Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 6(6): 658-670, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29622580

RESUMO

Type I interferon (IFN-I) is a class of antiviral immunomodulatory cytokines involved in many stages of tumor initiation and progression. IFN-I acts directly on tumor cells to inhibit cell growth and indirectly by activating immune cells to mount antitumor responses. To understand the role of endogenous IFN-I in spontaneous, oncogene-driven carcinogenesis, we characterized tumors arising in HER2/neu transgenic (neuT) mice carrying a nonfunctional mutation in the IFNI receptor (IFNAR1). Such mice are unresponsive to this family of cytokines. Compared with parental neu+/- mice (neuT mice), IFNAR1-/- neu+/- mice (IFNAR-neuT mice) showed earlier onset and increased tumor multiplicity with marked vascularization. IFNAR-neuT tumors exhibited deregulation of genes having adverse prognostic value in breast cancer patients, including the breast cancer stem cell (BCSC) marker aldehyde dehydrogenase-1A1 (ALDH1A1). An increased number of BCSCs were observed in IFNAR-neuT tumors, as assessed by ALDH1A1 enzymatic activity, clonogenic assay, and tumorigenic capacity. In vitro exposure of neuT+ mammospheres and cell lines to antibodies to IFN-I resulted in increased frequency of ALDH+ cells, suggesting that IFN-I controls stemness in tumor cells. Altogether, these results reveal a role of IFN-I in neuT-driven spontaneous carcinogenesis through intrinsic control of BCSCs. Cancer Immunol Res; 6(6); 658-70. ©2018 AACR.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Interferon Tipo I/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Receptor ErbB-2/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Camundongos Knockout , Camundongos Transgênicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Receptor ErbB-2/genética , Ensaio Tumoral de Célula-Tronco
2.
Blood ; 119(6): 1407-17, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-22184405

RESUMO

Cross-presentation allows antigen-presenting cells to present exogenous antigens to CD8(+) T cells, playing an essential role in controlling infections and tumor development. IFN-α induces the rapid differentiation of human mono-cytes into dendritic cells, known as IFN-DCs, highly efficient in mediating cross-presentation, as well as the cross-priming of CD8(+) T cells. Here, we have investigated the mechanisms underlying the cross-presentation ability of IFN-DCs by studying the intracellular sorting of soluble ovalbumin and nonstructural-3 protein of hepatitis C virus. Our results demonstrate that, independently from the route and mechanism of antigen entry, IFN-DCs are extraordinarily competent in preserving internalized proteins from early degradation and in routing antigens toward the MHC class-I processing pathway, allowing long-lasting, cross-priming capacity. In IFN-DCs, both early and recycling endosomes function as key compartments for the storage of both antigens and MHC-class I molecules and for proteasome- and transporter-associated with Ag processing-dependent auxiliary cross-presentation pathways. Because IFN-DCs closely resemble human DCs naturally occurring in vivo in response to infections and other danger signals, these findings may have important implications for the design of vaccination strategies in neoplastic or chronic infectious diseases.


Assuntos
Antígenos/metabolismo , Apresentação Cruzada/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Interferon-alfa/farmacologia , Antígenos/imunologia , Western Blotting , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endossomos/química , Endossomos/metabolismo , Citometria de Fluxo , Hepacivirus/imunologia , Hepacivirus/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Microscopia Confocal , Ovalbumina/imunologia , Ovalbumina/metabolismo , Ovalbumina/farmacocinética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/farmacocinética
3.
Vaccine ; 29(23): 3935-44, 2011 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-21481326

RESUMO

Human gammaherpesviruses such as Epstein-Barr virus (EBV) cause lifelong infections and associated diseases, by virtue of their ability to establish latent infection. Many studies performed in the past years in murine herpesvirus 68 (MHV-68) model of infection suggested that the limited immunity generated against isolated viral components by subunit vaccines cannot counteract the multiple immune evasion strategies operated by gammaherpesviruses. Indeed, a significant inhibition of long-term latency establishment could be observed in mice vaccinated with strains of genetically modified MHV-68 defective in reactivation or establishment of latency. In this study, we focused on the effects of interferon-α (IFN-α) on both the lytic and latent phase of MHV-68 infection, as exerted by the constitutive release of IFN-α1 by a clone of MHV-68 genetically modified to produce this cytokine (MHV-68mIFNα1). Although the MHV-68mIFNα1 recombinant virus exhibited in vitro replication features indistinguishable from those of the wild type MHV-68, its pathological properties were severely attenuated in vivo in immunocompetent mice and not in mice rendered genetically unresponsive to type I IFN, suggesting that a stronger immune response was primed in the presence of the cytokine. Notably, MHV-68mIFNα1 attenuation did not result in a reduced level of long-term spleen latency establishment. These results prompted us to evaluate the efficacy of MHV-68mIFNα1 in a prophylactic vaccination regimen aimed at inhibiting the symptoms of acute virus infection and the establishment of long-term latency after MHV-68 challenge. Our results show that mice vaccinated with MHV-68mIFNα1, administered as a live-attenuated or partially inactivated (by Psoralen and UV treatment) vaccine, were protected against the challenge with wt MHV-68 from all phases of infection. The ability of MHV-68mIFNα1 to produce IFN-α at the site of the infection, thus efficiently stimulating the immune system in case of virus reactivation from latency, makes this recombinant virus a safer live-attenuated vaccine as compared to the previously reported latency-deficient clones.


Assuntos
Infecções por Herpesviridae/prevenção & controle , Interferon-alfa/metabolismo , Organismos Geneticamente Modificados/fisiologia , Rhadinovirus/patogenicidade , Infecções Tumorais por Vírus/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Animais , Linhagem Celular , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Humanos , Interferon-alfa/genética , Camundongos , Camundongos Endogâmicos C57BL , Organismos Geneticamente Modificados/genética , Organismos Geneticamente Modificados/metabolismo , Recombinação Genética , Rhadinovirus/genética , Rhadinovirus/metabolismo , Baço , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Replicação Viral
4.
PLoS One ; 6(2): e17364, 2011 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-21387004

RESUMO

IFN-α exerts multiple effects leading to immune protection against pathogens and cancer as well to autoimmune reactions by acting on monocytes and dendritic cells. We analyzed the versatility of human monocytes conditioned by IFN-α towards dendritic cell differentiation (IFN-DC) in shaping the autologous T-helper response. Priming of naïve CD4 T cells with autologous IFN-DC in the presence of either SEA or anti-CD3, resulted, in addition to a prominent expansion of CXCR3+ IFN-γ-producing CD4 Th1 cells, in the emergence of two distinct subsets of IL-17-producing CD4 T cells: i) a predominant Th17 population selectively producing IL-17 and expressing CCR6; ii) a minor Th1/Th17 population, producing both IL-17 and IFN-γ. After phagocytosis of apoptotic cells, IFN-DC induced Th17 cell expansion and IL-17 release. Notably, the use of neutralizing antibodies revealed that IL-23 was an essential cytokine in mediating Th17 cell development by IFN-DC. The demonstration of the IFN-DC-induced expansion of both Th1 and Th17 cell populations reveals the intrinsic plasticity of these DC in orienting the immune response and provides a mechanistic link between IFN-α and the onset of autoimmune phenomena, which have been correlated with both IL-17 production and exposure to IFN-α.


Assuntos
Interferon-alfa/farmacologia , Interleucina-12/fisiologia , Interleucina-23/fisiologia , Monócitos/efeitos dos fármacos , Células Th1/fisiologia , Células Th17/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/fisiologia , Humanos , Interleucina-12/metabolismo , Interleucina-23/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/fisiologia , Modelos Biológicos , Monócitos/metabolismo , Monócitos/fisiologia , Células Th1/efeitos dos fármacos , Células Th17/metabolismo , Células Th17/fisiologia , Células Th17/transplante
5.
Immunobiology ; 216(5): 537-47, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21093097

RESUMO

We have investigated the molecular mechanisms underlying the peculiar cross-presentation efficiency of human dendritic cells (DCs) differentiated from monocytes in the presence of Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) and Interferon (IFN)-α (IFN-DCs). To this end, we evaluated the capability of IFN-DCs to present and cross-present epitopes derived from Epstein-Barr Virus (EBV) or human melanoma-associated antigens after exposure to cell lysates or apoptotic cells. In an autologous setting, IFN-DCs loaded with Lymphoblastoid Cell Lines (LCL) lysates or apoptotic LCL were highly efficient in expanding, respectively, EBV-specific class II- or class I-restricted memory T cell responses. Of note, IFN-DCs loaded with apoptotic LCL were more potent than fully mature DCs in triggering the cytotoxicity of CD8(+) T lymphocytes recognizing a subdominant HLA-A*0201-restricted epitope derived from EBV latent membrane protein 2 (LMP2). In addition, IFN-DCs loaded with apoptotic human melanoma cells were highly efficient in cross-presenting the MART-1(27-35) epitope to a specific CD8(+) cytotoxic T cell clone, and this functional activity was proteasome-dependent. These IFN-DC properties were associated with an enhanced expression of all the immunoproteasome subunits as well as of TAP-1, TAP-2 and tapasin, and, interestingly, to a higher proteasome proteolytic activity as compared to immature or mature DCs. Altogether, these results highlight new mechanisms by which IFN-α influences antigen processing and cross-presentation ability of monocyte-derived DCs, with potentially important implications for the development of DC-based therapeutic vaccines.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/metabolismo , Imunoterapia Adotiva , Interferon-alfa/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Apresentação Cruzada , Citotoxicidade Imunológica , Células Dendríticas/citologia , Células Dendríticas/imunologia , Epitopos/metabolismo , Epitopos de Linfócito T/metabolismo , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Memória Imunológica , Interferon-alfa/imunologia , Ativação Linfocitária , Proteínas de Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Ligação Proteica , Proteínas da Matriz Viral/metabolismo
6.
Cancer Res ; 70(2): 490-500, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20068181

RESUMO

Many cancer cells express Toll-like receptors (TLR) that offer possible therapeutic targets. Polyadenylic-polyuridylic acid [poly(A:U)] is an agonist of the Toll-like receptor TLR3 that displays anticancer properties. In this study, we illustrate how the immunostimulatory and immunosuppressive effects of this agent can be uncoupled to therapeutic advantage. We took advantage of two TLR3-expressing tumor models that produced large amounts of CCL5 (a CCR5 ligand) and CXCL10 (a CXCR3 ligand) in response to type I IFN and poly(A:U), both in vitro and in vivo. Conventional chemotherapy or in vivo injection of poly(A:U), alone or in combination, failed to reduce tumor growth unless an immunochemotherapeutic regimen of vaccination against tumor antigens was included. CCL5 blockade improved the efficacy of immunochemotherapy, whereas CXCR3 blockade abolished its beneficial effects. These findings show how poly(A:U) can elicit production of a range of chemokines by tumor cells that reinforce immunostimulatory or immunosuppressive effects. Optimizing the anticancer effects of TLR3 agonists may require manipulating these chemokines or their receptors.


Assuntos
Vacinas Anticâncer/farmacologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Poli A-U/farmacologia , Receptor 3 Toll-Like/antagonistas & inibidores , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Quimiocina CCL5/biossíntese , Quimiocina CCL5/metabolismo , Sinergismo Farmacológico , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Receptores CCR5/biossíntese , Receptores CCR5/metabolismo , Receptores CXCR3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 3 Toll-Like/imunologia , Receptor 3 Toll-Like/metabolismo
7.
Handb Exp Pharmacol ; (188): 295-317, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19031032

RESUMO

Dendritic cells (DCs) play a crucial role in linking innate and adaptive immunity, by virtue of their unique ability to take up and process antigens in the peripheral blood and tissues and, upon migration to draining lymph nodes, to present antigen to resting lymphocytes. Notably, these DC functions are modulated by cytokines and chemokines controlling the activation and maturation of these cells, thus shaping the response towards either immunity or tolerance.An ensemble of recent studies have emphasized an important role of type I IFNs in the DC differentiation/activation, suggesting the existence of a natural alliance between these cytokines and DCs in linking innate and adaptive immunity. Herein, we will review how type I IFNs can promote the ex vivo differentiation of human DCs and orient DC functions towards the priming and expansion of protective antitumor immune responses. We will also discuss how the knowledge on type I IFN-DC interactions could be exploited for the design of more selective and effective strategies of cancer immunotherapy.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Vacinas Anticâncer , Células Dendríticas/transplante , Imunoterapia Adotiva , Interferon-alfa/uso terapêutico , Neoplasias/terapia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Células Dendríticas/imunologia , Humanos , Imunidade Inata , Interferon-alfa/metabolismo , Neoplasias/imunologia , Fenótipo
8.
Tumori ; 94(2): 182-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18564605

RESUMO

Biotherapy of cancer holds great promise for its potential to lead to the identification of novel, selective, and effective treatments against cancer. However, the clinical development of biopharmaceuticals and biotherapy products is hampered by several and diverse barriers. Herein, we will address some of the critical issues identified both at the national and European level as the major obstacles for the translation of knowledge into clinical applications in the field of biotherapy and immunotherapy of cancer. We will also illustrate specific initiatives undertaken both in Europe and in Italy in order to support the translational and clinical research and that are expected to have a favorable impact on the process of clinical development of novel and more effective therapeutic interventions against cancer. The contents of this article are directly referred to the event "International Clinical Trials' Day on Biotherapy of Cancer" organized in the context of the OECI Genoa 2008, with the sponsorship of Alliance Against Cancer (ACC) and the Istituto Superiore di Sanità (ISS, the Italian National Institute of Health), and under the auspices of the European Clinical Research Infrastructures Network (ECRIN). This event sees the active participation of representatives of the ISS and of the Italian Network for Tumor Biotherapy, both involved in a project recently funded by ACC and aimed at the promotion of clinical research in the field of cancer biotherapy and immunotherapy, through the creation of a national network of clinical cancer research centers and GMP facilities dedicated to the production of biological drugs and advanced medicinal products.


Assuntos
Pesquisa Biomédica/tendências , Redes Comunitárias , Imunoterapia , Neoplasias/terapia , Academias e Institutos , Animais , Terapia Biológica , Comportamento Cooperativo , Modelos Animais de Doenças , Europa (Continente) , Humanos , Itália , Neoplasias/imunologia , Desenvolvimento de Programas
9.
J Immunol ; 180(10): 6477-83, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18453565

RESUMO

The synergistic antitumor effects of the combination therapy imatinib mesylate (IM) and IL-2 depended upon NK1.1- expressing cells and were associated with the accumulation of CD11c(int)B220(+)NK1.1(+) IFN-producing killer dendritic cells (IKDC) into tumor beds. In this study, we show that the antitumor efficacy of the combination therapy was compromised in IL-15 and IFN-type 1R loss-of-function mice. IL-15Ralpha was required for the proliferation of IKDC during IM plus IL-2 therapy. Trans-presentation of IL-15/IL-15Ralpha activated IKDC to express CCR2 and to respond to type 1 IFN by producing CCL2. Moreover, the antitumor effects of the combination therapy correlated with a CCL2-dependent recruitment of IKDC, but not B220(-) NK cells, into tumor beds. Altogether, the IL-15-driven peripheral expansion and the CCL-2-dependent intratumoral chemoattraction of IKDC are two critical parameters dictating the antitumor efficacy of IM plus IL-2 in mice.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Células Dendríticas/imunologia , Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Experimentais/tratamento farmacológico , Animais , Benzamidas , Quimiocina CCL2/metabolismo , Citometria de Fluxo , Humanos , Mesilato de Imatinib , Interleucina-2/administração & dosagem , Camundongos , Camundongos Mutantes , Piperazinas/administração & dosagem , Pirimidinas/administração & dosagem , Receptores CCR2/metabolismo , Receptores de Interferon/imunologia , Receptores de Interferon/metabolismo , Receptor de Interferon gama
10.
Cytokine Growth Factor Rev ; 19(1): 93-107, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18054517

RESUMO

Dendritic cells (DCs) play a crucial role in linking innate and adaptive immunity and, thus, in the generation of a protective immune response against both infectious diseases and tumors. The ability of DCs to prime and expand an immune response is regulated by signals acting through soluble mediators, mainly cytokines and chemokines. Understanding how cytokines influence DC functions and orchestrate the interactions of DCs with other immune cells is strictly instrumental to the progress in cancer immunotherapy. Herein, we will illustrate how certain cytokines and immune stimulating molecules can induce and sustain the antitumor immune response by acting on DCs. We will also discuss these cytokine-DC interactions in the light of clinical results in cancer patients.


Assuntos
Antígenos de Neoplasias/imunologia , Citocinas/fisiologia , Células Dendríticas/imunologia , Neoplasias/imunologia , Adjuvantes Imunológicos/fisiologia , Aminoquinolinas/uso terapêutico , Animais , Apresentação de Antígeno , Vacinas Anticâncer/uso terapêutico , Células Cultivadas , Ensaios Clínicos como Assunto , Ilhas de CpG/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Imiquimode , Imunoterapia , Interferon-alfa/uso terapêutico , Neoplasias/prevenção & controle , Oligodesoxirribonucleotídeos/uso terapêutico , Receptor Toll-Like 9/agonistas
11.
Biochimie ; 89(6-7): 884-93, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17532550

RESUMO

Interferons-alpha (IFN-alpha) are pleiotropic cytokines belonging to type I IFNs, extensively used in the treatment of patients with some types of cancer and viral disease. IFN-alpha can affect tumor cell functions by multiple mechanisms. In addition, these cytokines can promote the differentiation and activity of host immune cells. Early studies in mouse tumor models showed the importance of host immune mechanisms in the generation of a long-lasting antitumor response after treatment of the animals with IFN-alpha/beta. Subsequently, an ensemble of studies based on the use of genetically modified tumor cells expressing specific IFN molecules provided important information on the host-mediated antitumor mechanisms induced by the local production of IFN-alpha. Of note, several studies have then underscored new immunomodulatory effects of IFN-alpha, including activities on T cells and dendritic cells, which may lead to IFN-induced antitumor immunity. In addition, recent reports on new immune correlates in cancer patients responding to IFN-alpha represent additional evidence on the importance of the interactions of IFN-alpha with the immune system for the generation of a durable antitumor response. On the whole, this knowledge suggests the advantage of using these cytokines as adjuvants of cancer vaccines and for the in vitro generation of highly active dendritic cells to be utilized for therapeutic vaccination of cancer patients.


Assuntos
Antineoplásicos/farmacologia , Imunoterapia/métodos , Interferon-alfa/fisiologia , Neoplasias/metabolismo , Animais , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Técnicas de Transferência de Genes , Humanos , Interferon-alfa/metabolismo , Modelos Biológicos , Monócitos/metabolismo , Neoplasias/terapia
12.
Cancer Immunol Immunother ; 56(4): 581-5, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17131119

RESUMO

The main aims of the international meeting "Immunotherapy of Cancer: Challenges and Needs" were to review the state of the art of cancer immunotherapy and to identify critical issues which deserve special attention for promoting progress of research in this field, with a particular focus on the perspectives of clinical research. Novel concepts and strategies for identifying, monitoring and predicting effective responses to cancer immunotherapy protocols were presented, focused on the use of adjuvants (CpG oligonucleotides) or cytokines (IFN-alpha) to enhance the efficacy of cancer vaccines. Moreover, the possible advantages of using different types of dendritic cells (for active immunization strategies) or T cells (for adoptive immunotherapy protocols) were debated. A consensus was achieved on the need for enhancing the efficacy of cancer vaccines or adoptive cell immunotherapy by combining these strategies with other anti-cancer treatments, including chemotherapy. Finally, initiatives for promoting clinical research by establishing a strategic cooperation in the field of cancer immunotherapy based on the active participation of all the relevant actors, including public institutions responsible of Public Health, National Cancer Institutes, industry, representatives of regulatory bodies, and patients' organizations were proposed.


Assuntos
Imunoterapia/métodos , Neoplasias/terapia , Animais , Ensaios Clínicos como Assunto , Humanos
13.
Cancer Res ; 66(9): 4943-51, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651452

RESUMO

The use of IFN-alpha in clinical oncology has generally been based on the rationale of exploiting its antiproliferative and antiangiogenic activities. However, IFN-alpha also exhibits enhancing effects on T-cell and dendritic cell functions, which may suggest a novel use as a vaccine adjuvant. We have carried out a pilot phase I-II trial to determine the effects of IFN-alpha, administered as an adjuvant of Melan-A/MART-1:26-35(27L) and gp100:209-217(210M) peptides, on immune responses in stage IV melanoma patients. In five of the seven evaluable patients, a consistent enhancement of CD8(+) T cells recognizing modified and native MART-1 and gp100 peptides and MART-1(+)gp100(+) melanoma cells was observed. Moreover, vaccination induced an increase in CD8(+) T-cell binding to HLA tetramers containing the relevant peptides and an increased frequency of CD45RA(+)CCR7(-) (terminally differentiated effectors) and CD45RA(-)CCR7(-) (effector memory) cells. In all patients, treatment augmented significantly the percentage of CD14(+) monocytes and particularly of the CD14(+)CD16(+) cell fraction. An increased expression of CD40 and CD86 costimulatory molecules in monocytes was also observed. Notably, postvaccination monocytes from two of the three patients showing stable disease or long disease-free survival showed an enhanced antigen-presenting cell function and capability to secrete IP10/CXCL10 when tested in mixed leukocyte reaction assays, associated to a boost of antigen and melanoma-specific CD8(+) T cells. Although further clinical studies are needed to show the adjuvant activity of IFN-alpha, the present data represent an important starting point for considering a new clinical use of IFN-alpha and new immunologic end points, potentially predictive of clinical response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Interferon-alfa/uso terapêutico , Melanoma/terapia , Glicoproteínas de Membrana/imunologia , Proteínas de Neoplasias/imunologia , Adjuvantes Imunológicos/uso terapêutico , Apresentação de Antígeno , Antígenos de Neoplasias , Vacinas Anticâncer/imunologia , Células Dendríticas/citologia , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Imunofenotipagem , Ativação Linfocitária , Antígeno MART-1 , Melanoma/imunologia , Melanoma/patologia , Monócitos/citologia , Monócitos/imunologia , Estadiamento de Neoplasias , Projetos Piloto , Antígeno gp100 de Melanoma
14.
Cancer Res ; 64(18): 6827-30, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15375003

RESUMO

The major aims of the International Meeting on Cancer Vaccines were to review the state-of-the-art research on cancer vaccines, to compare different experimental approaches of therapeutic vaccination and to discuss critical issues and perspectives. The results from recent clinical trials in patients treated with different types of cancer vaccines were presented. Reasons for the limited response and possible modalities for enhancing efficacy of therapeutic vaccines were subjects of major discussion. A consensus was achieved on the need of combining cancer vaccines with other anticancer treatments. Of note, evidence stemming from studies in animal models pointed out new rationales for a selective combination of cancer vaccines with chemotherapy. In addition, some main presentations focused on new adjuvants (CpG oligonucleotides) and on the role of cytokines (i.e., type I IFN, interleukin 12, and interleukin 15) in promoting an antitumor immune response to vaccines. A considerable attention was given to regulatory T cells and to strategies for suppressing their function, thus enhancing vaccine efficacy. An entire session was devoted to the use of dendritic cells for the development of cancer vaccines. The results of clinical studies and the advantages of using new modalities for preparing dendritic cell-based vaccines were discussed.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias/terapia , Vacinas Anticâncer/imunologia , Ensaios Clínicos como Assunto , Humanos , Imunoterapia/métodos , Neoplasias/imunologia
15.
Vaccine ; 22(11-12): 1433-40, 2004 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15063566

RESUMO

Human gammaherpesviruses such as Epstein-Barr virus (EBV) cause lifelong infections and associated diseases, including malignancies, and the development of an effective vaccine against this class of viral infections is of considerable interest. The murine herpesvirus 68 (MHV-68) model provides a useful experimental setting to investigate the immune response to gammaherpesvirus infections and to evaluate the efficacy of vaccination strategies. In this study, we tested a heat-inactivated MHV-68 vaccine in immunocompetent mice as well as in B cell-deficient or type I IFN receptor knockout mice. Vaccination with heat-inactivated MHV-68 protected immunocompetent mice from the acute MHV-68 infection in the lung and strongly reduced the expansion of latently infected cells in the spleen and the development of splenomegaly. A similar inhibition of the acute viral replication in the lung was also observed in vaccinated B cell-deficient mice. Of note, the inactivated MHV-68 vaccine completely protected type I IFN receptor knockout mice from the infection with a lethal dose of MHV-68.


Assuntos
Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Herpesviridae/imunologia , Vacinas contra Herpesvirus/imunologia , Interferon Tipo I/fisiologia , Receptores de Interferon/fisiologia , Latência Viral/imunologia , Replicação Viral/fisiologia , Animais , Linfócitos B/imunologia , Linfócitos B/fisiologia , Linhagem Celular , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes de Neutralização , Receptores de Interferon/genética , Vacinação , Vacinas de Produtos Inativados/imunologia
16.
J Immunol ; 172(4): 2137-46, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14764679

RESUMO

Ideal vaccines should be stable, safe, molecularly defined, and out-of-shelf reagents efficient at triggering effector and memory Ag-specific T cell-based immune responses. Dendritic cell-derived exosomes could be considered as novel peptide-based vaccines because exosomes harbor a discrete set of proteins, bear functional MHC class I and II molecules that can be loaded with synthetic peptides of choice, and are stable reagents that were safely used in pioneering phase I studies. However, we showed in part I that exosomes are efficient to promote primary MHC class I-restricted effector CD8(+) T cell responses only when transferred onto mature DC in vivo. In this work, we bring evidence that among the clinically available reagents, Toll-like receptor 3 and 9 ligands are elective adjuvants capable of triggering efficient MHC-restricted CD8(+) T cell responses when combined to exosomes. Exosome immunogenicity across species allowed to verify the efficacy of good manufactory procedures-manufactured human exosomes admixed with CpG oligonucleotides in prophylactic and therapeutic settings of melanoma in HLA-A2 transgenic mice. CpG adjuvants appear to be ideal adjuvants for exosome-based cancer vaccines.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas Anticâncer/imunologia , Endossomos/imunologia , Rejeição de Enxerto/imunologia , Melanoma Experimental/imunologia , Oligodesoxirribonucleotídeos/imunologia , Linfócitos T Reguladores/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Adjuvantes Imunológicos/metabolismo , Animais , Vacinas Anticâncer/administração & dosagem , Sistema Livre de Células/imunologia , Sistema Livre de Células/transplante , Ilhas de CpG/imunologia , Proteínas de Ligação a DNA/metabolismo , Endossomos/transplante , Antígeno HLA-A2/biossíntese , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Humanos , Interfase/imunologia , Ligantes , Melanoma Experimental/prevenção & controle , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/metabolismo , RNA de Cadeia Dupla/imunologia , Receptores de Superfície Celular/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Reguladores/citologia , Receptor 3 Toll-Like , Receptor Toll-Like 9 , Receptores Toll-Like , Vacinas de Subunidades Antigênicas/administração & dosagem , Antígeno gp100 de Melanoma
17.
J Immunol ; 170(10): 5195-202, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12734367

RESUMO

Cellular immune responses are crucial for the control of EBV-associated lymphoproliferative diseases. To induce an anti-EBV cell-mediated immunity, we have used dendritic cells (DCs) generated by a 3-day culture of human CD14(+) monocytes in the presence of GM-CSF and type I IFN (IFN-DCs) and pulsed with peptides corresponding to CTL EBV epitopes. The functional activity of IFN-DCs was compared with that of APCs differentiated by culturing monocytes for 3 days with GM-CSF and IL-4 and indicated as IL-4-DCs. Stimulation of PBLs from EBV-seropositive donors with EBV peptide-pulsed autologous IFN-DCs resulted in a stronger expansion of specific T lymphocytes producing IFN-gamma with respect to stimulation with peptide-loaded IL-4-DCs, as assessed by ELISPOT assays. When purified CD8(+) T cells were cocultured with EBV peptide-pulsed IFN-DCs or IL-4-DCs, significantly higher levels of specific cytotoxic activity were observed in CD8(+) T cell cultures stimulated with IFN-DCs. Injection of peptide-pulsed IFN-DCs into SCID mice transplanted with autologous PBLs led to the recovery of a significantly greater number of EBV-specific human CD8(+) T cells from the spleen and the peritoneal cavity with respect to that recovered from mice injected with peptide-pulsed IL-4-DCs. Moreover, a significant delay in lymphoma development was observed when peptide-pulsed IFN-DCs were injected into SCID mice reconstituted with PBMCs endowed with a high capability of lymphoma induction, whereas injection of unpulsed IFN-DCs was ineffective. Our results indicate that IFN-DCs efficiently promote in vitro and in vivo the expansion of CD8(+) T lymphocytes acting as cytotoxic effectors against EBV-transformed cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Herpesvirus Humano 4/imunologia , Interferon-alfa/farmacologia , Monócitos/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/transplante , Técnicas de Cultura de Células/métodos , Divisão Celular/imunologia , Linhagem Celular Transformada , Citotoxicidade Imunológica , Células Dendríticas/citologia , Células Dendríticas/transplante , Feminino , Humanos , Memória Imunológica/imunologia , Injeções Intraperitoneais , Ativação Linfocitária/imunologia , Linfoma de Células B/imunologia , Linfoma de Células B/prevenção & controle , Camundongos , Camundongos SCID , Monócitos/citologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia
19.
Am J Pathol ; 160(4): 1507-20, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11943735

RESUMO

Our previous article reported that retroviral transduction of human type I consensus interferon-coding sequence into two human melanoma cells increased their susceptibility to cisplatin-induced apoptosis. Importantly, primary melanoma cells were significantly more sensitive to cisplatin-induced apoptosis with respect to metastatic melanoma cells. The aim of this study was to elucidate the subcellular mechanisms involved in this interferon-induced apoptotic proneness. Our results indicate that 1) cisplatin-induced apoptosis can be referred to as the type II apoptosis, ie, to the mitochondrially driven cascade; 2) treatment of interferon-producing melanoma cells with other type II apoptotic stimuli, such as radiation or staurosporine, also resulted in massive apoptosis, whereas type I stimuli, ie, anti-Fas, were ineffective; 3) interferon sensitization involved the caspase cascade in primary melanoma cells and the alternative pathway represented by cathepsin-mediated apoptosis in metastatic melanoma cells; 4) interferon production sensitizes cells to apoptosis by inducing, as the earliest event, mitochondrial membrane hyperpolarization. These results suggest that constitutive production of type I interferon by melanoma cells can act as an intracellular booster capable of increasing cell proneness to apoptosis by specifically modifying mitochondrial homeostasis and independently from the apoptotic cascade involved.


Assuntos
Apoptose/fisiologia , Interferon Tipo I/fisiologia , Melanoma/fisiopatologia , Mitocôndrias/fisiologia , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistência a Medicamentos/fisiologia , Técnicas de Transferência de Genes , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Interferon Tipo I/genética , Melanoma/genética , Melanoma/patologia , Mitocôndrias/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas , Ubiquitina/metabolismo
20.
Cytokine Growth Factor Rev ; 13(2): 119-34, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11900988

RESUMO

Interferon-alpha (IFN-alpha) is a pleiotropic cytokine belonging to type I IFN, currently used in cancer patients. Early studies in mouse tumor models have shown the importance of host immune mechanisms in the generation of a long-lasting antitumor response to type I IFN. Recent studies have underscored new immunomodulatory effects of IFN-alpha, including activities on T and dendritic cells, which may explain IFN-induced tumor immunity. Reports on new immune correlates in cancer patients responding to IFN-alpha represent additional evidence on the importance of the interactions of IFN-alpha with the immune system for the generation of durable antitumor response. This knowledge, together with results from studies on genetically modified tumor cells expressing IFN-alpha, suggest novel strategies for using these cytokines in cancer immunotherapy and in particular the use of IFN-alpha as an immune adjuvant for the development of cancer vaccines.


Assuntos
Vacinas Anticâncer , Imunoterapia/métodos , Interferon-alfa/fisiologia , Animais , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Humanos , Interferon-alfa/farmacologia , Melanoma/cirurgia , Melanoma/terapia , Camundongos , Modelos Biológicos , Neoplasias/mortalidade , Neoplasias/terapia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA