Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Ther ; 23(8): 1380-1390, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25939491

RESUMO

Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.


Assuntos
Endorribonucleases/genética , Regulação Neoplásica da Expressão Gênica , Linfócitos do Interstício Tumoral/imunologia , Melanoma/terapia , Receptor de Morte Celular Programada 1/genética , Dedos de Zinco , Alelos , Animais , Separação Celular , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Memória Imunológica , Imunoterapia Adotiva , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Fenótipo , Receptor de Morte Celular Programada 1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
PLoS One ; 9(5): e96137, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24846143

RESUMO

Following spinal cord injury (SCI) there are drastic changes that occur in the spinal microvasculature, including ischemia, hemorrhage, endothelial cell death and blood-spinal cord barrier disruption. Vascular endothelial growth factor-A (VEGF-A) is a pleiotropic factor recognized for its pro-angiogenic properties; however, VEGF has recently been shown to provide neuroprotection. We hypothesized that delivery of AdV-ZFP-VEGF--an adenovirally delivered bio-engineered zinc-finger transcription factor that promotes endogenous VEGF-A expression--would result in angiogenesis, neuroprotection and functional recovery following SCI. This novel VEGF gene therapy induces the endogenous production of multiple VEGF-A isoforms; a critical factor for proper vascular development and repair. Briefly, female Wistar rats--under cyclosporin immunosuppression--received a 35 g clip-compression injury and were administered AdV-ZFP-VEGF or AdV-eGFP at 24 hours post-SCI. qRT-PCR and Western Blot analysis of VEGF-A mRNA and protein, showed significant increases in VEGF-A expression in AdV-ZFP-VEGF treated animals (p<0.001 and p<0.05, respectively). Analysis of NF200, TUNEL, and RECA-1 indicated that AdV-ZFP-VEGF increased axonal preservation (p<0.05), reduced cell death (p<0.01), and increased blood vessels (p<0.01), respectively. Moreover, AdV-ZFP-VEGF resulted in a 10% increase in blood vessel proliferation (p<0.001). Catwalk™ analysis showed AdV-ZFP-VEGF treatment dramatically improves hindlimb weight support (p<0.05) and increases hindlimb swing speed (p<0.02) when compared to control animals. Finally, AdV-ZFP-VEGF administration provided a significant reduction in allodynia (p<0.01). Overall, the results of this study indicate that AdV-ZFP-VEGF administration can be delivered in a clinically relevant time-window following SCI (24 hours) and provide significant molecular and functional benefits.


Assuntos
Adenoviridae , Terapia Genética/métodos , Hiperalgesia/terapia , Traumatismos da Medula Espinal/terapia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Dedos de Zinco , Animais , Feminino , Células HEK293 , Humanos , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Neovascularização Fisiológica/genética , Ratos , Ratos Wistar , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Fator A de Crescimento do Endotélio Vascular/genética
3.
N Engl J Med ; 370(10): 901-10, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24597865

RESUMO

BACKGROUND: CCR5 is the major coreceptor for human immunodeficiency virus (HIV). We investigated whether site-specific modification of the gene ("gene editing")--in this case, the infusion of autologous CD4 T cells in which the CCR5 gene was rendered permanently dysfunctional by a zinc-finger nuclease (ZFN)--is safe. METHODS: We enrolled 12 patients in an open-label, nonrandomized, uncontrolled study of a single dose of ZFN-modified autologous CD4 T cells. The patients had chronic aviremic HIV infection while they were receiving highly active antiretroviral therapy. Six of them underwent an interruption in antiretroviral treatment 4 weeks after the infusion of 10 billion autologous CD4 T cells, 11 to 28% of which were genetically modified with the ZFN. The primary outcome was safety as assessed by treatment-related adverse events. Secondary outcomes included measures of immune reconstitution and HIV resistance. RESULTS: One serious adverse event was associated with infusion of the ZFN-modified autologous CD4 T cells and was attributed to a transfusion reaction. The median CD4 T-cell count was 1517 per cubic millimeter at week 1, a significant increase from the preinfusion count of 448 per cubic millimeter (P<0.001). The median concentration of CCR5-modified CD4 T cells at 1 week was 250 cells per cubic millimeter. This constituted 8.8% of circulating peripheral-blood mononuclear cells and 13.9% of circulating CD4 T cells. Modified cells had an estimated mean half-life of 48 weeks. During treatment interruption and the resultant viremia, the decline in circulating CCR5-modified cells (-1.81 cells per day) was significantly less than the decline in unmodified cells (-7.25 cells per day) (P=0.02). HIV RNA became undetectable in one of four patients who could be evaluated. The blood level of HIV DNA decreased in most patients. CONCLUSIONS: CCR5-modified autologous CD4 T-cell infusions are safe within the limits of this study. (Funded by the National Institute of Allergy and Infectious Diseases and others; ClinicalTrials.gov number, NCT00842634.).


Assuntos
Linfócitos T CD4-Positivos/transplante , Terapia Genética , Infecções por HIV/terapia , Transfusão de Linfócitos , Receptores CCR5/genética , Adulto , Terapia Antirretroviral de Alta Atividade , Transfusão de Sangue Autóloga , Linfócitos T CD4-Positivos/química , Terapia Combinada , DNA Viral/sangue , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , HIV/genética , HIV/isolamento & purificação , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Humanos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , RNA Viral/sangue , Reto/imunologia , Carga Viral
4.
Clin Cancer Res ; 18(3): 858-68, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22147941

RESUMO

PURPOSE: Listeria monocytogenes (Lm)-based vaccines stimulate both innate and adaptive immunity. ANZ-100 is a live-attenuated Lm strain (Lm ΔactA/ΔinlB). Uptake by phagocytes in the liver results in local inflammatory responses and activation and recruitment of natural killer (NK) and T cells, in association with increased survival of mice bearing hepatic metastases. The Lm ΔactA/ΔinlB strain, engineered to express human mesothelin (CRS-207), a tumor-associated antigen expressed by a variety of tumors, induces mesothelin-specific T-cell responses against mesothelin-expressing murine tumors. These two phase I studies test ANZ-100 and CRS-207 in subjects with liver metastases and mesothelin-expressing cancers, respectively. EXPERIMENTAL DESIGN: A single intravenous injection of ANZ-100 was evaluated in a dose escalation study in subjects with liver metastases. Nine subjects received 1 × 10(6), 3 × 10(7), or 3 × 10(8) colony-forming units (cfu). CRS-207 was evaluated in a dose-escalation study in subjects with mesothelioma, lung, pancreatic, or ovarian cancers. Seventeen subjects received up to 4 doses of 1 × 10(8), 3 × 10(8), 1 × 10(9), or 1 × 10(10) cfu. RESULTS: A single infusion of ANZ-100 was well tolerated to the maximum planned dose. Adverse events included transient laboratory abnormalities and symptoms associated with cytokine release. Multiple infusions of CRS-207 were well tolerated up to 1 × 10(9) cfu, the determined maximum tolerated dose. Immune activation was observed for both ANZ-100 and CRS-207 as measured by serum cytokine/chemokine levels and NK cell activation. In the CRS-207 study, listeriolysin O and mesothelin-specific T-cell responses were detected and 37% of subjects lived ≥15 months. CONCLUSIONS: ANZ-100 and CRS-207 administration was safe and resulted in immune activation.


Assuntos
Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Proteínas Ligadas por GPI/imunologia , Listeria monocytogenes/imunologia , Neoplasias Hepáticas/terapia , Adulto , Idoso , Vacinas Bacterianas/efeitos adversos , Vacinas Anticâncer/efeitos adversos , Carcinoma/secundário , Carcinoma/terapia , Citocinas/sangue , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Masculino , Dose Máxima Tolerável , Mesotelina , Mesotelioma/patologia , Mesotelioma/terapia , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia
5.
J Immunother ; 32(8): 856-69, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19752748

RESUMO

Immunotherapeutic vaccine is potentially an effective strategy to combat cancer. Essential components of an effective vaccine must include antigens that are processed by the major histocompatibility complex class I pathway, presented by the tumor major histocompatibility complex molecules, and an effective antigen delivery platform that is capable of breaking self-tolerance. In this study, we characterized a set of ovarian cancer-specific T-cell epitopes delivered by live-attenuated recombinant Listeria monocytogenes (Lm DeltaactADeltainlB) as a vaccine vector. We present data that peptide-specific T cells recognize the human monocytic cell line THP-1 infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Furthermore, we demonstrate that recombinant L. monocytogenes (Lm)-infected antigen-presenting cells can prime and expand epitope-specific CD8 T cells in vitro and such CD8 T cells recognize not only peptide-loaded targets but also ovarian and breast tumor cells presenting endogenous epitopes. Finally, peptide-specific T cells generated using peripheral blood mononuclear cell from ovarian cancer patients recognize target cells infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Our results demonstrate that live-attenuated recombinant Lm can be used effectively as a vehicle to deliver cancer peptide antigens singly or as a multiepitope construct. Thus, the use of recombinant live-attenuated Lm strains encoding endogenously processed and presented tumor epitopes/antigens represents an attractive strategy for active cancer immunotherapy in a clinical setting.


Assuntos
Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Listeria monocytogenes/imunologia , Neoplasias Ovarianas/terapia , Apresentação de Antígeno , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , Mapeamento de Epitopos , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Feminino , Antígeno HLA-A2/metabolismo , Humanos , Listeriose/imunologia , Ativação Linfocitária , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia
6.
J Immunol ; 179(11): 7376-84, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18025181

RESUMO

NK cells can identify and eliminate emerging tumors due to altered expression of activating and inhibitory ligands on aberrant cells, a process that is greatly enhanced following NK cell activation. As a principal site of both tumor metastases and immature NK cells, the liver represents a unique anatomic location in which activation of the innate immune system could provide substantial therapeutic benefit. We describe here the NK cell-dependent destruction of a primary hepatic tumor following infection with an attenuated intracellular bacterium derived from Listeria monocytogenes. NK cell-mediated immunity correlated with the ordered migration and maturation of NK cells within the liver. Cytolytic activity was partially dependent on NKG2D-mediated tumor cell recognition, but surprisingly was still effective in the absence of type I IFN. Significantly, NK cell-mediated destruction of a primary hepatic tumor in infected mice led to long-lived CD4- and CD8 T cell-dependent tumor-specific adaptive immunity. These findings establish that activation and differentiation of immature NK cells using complex microbial stimuli can elicit potent anti-tumor activity within the liver, promote cross-presentation of tumor-derived Ags leading to long-lived systemic anti-tumor immunity, and suggests a paradigm for clinical intervention of liver metastatic carcinoma.


Assuntos
Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Listeria monocytogenes/imunologia , Neoplasias Hepáticas/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Imunidade Celular , Imunidade Inata , Interferon gama/biossíntese , Ligantes , Listeriose/imunologia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores Imunológicos/imunologia , Receptores Imunológicos/uso terapêutico , Receptores de Células Matadoras Naturais
7.
Cancer Res ; 66(2): 1096-104, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424046

RESUMO

Improved immunization and ex vivo T-cell culture strategies can generate larger numbers and more potent tumor-specific effector cells than previously possible. Nonetheless, the capacity of these cells to eliminate established tumors is limited by their ability to efficiently enter tumor-bearing organs and mediate their effector function. In the current study, we show that the administration of an engineered organ-homing microbe selectively targets tumor-specific immune responses to metastases within that organ. Specifically, an attenuated Listeria monocytogenes strain, which preferentially infects the liver following systemic administration, dramatically enhances the activity of a cancer vaccine against liver metastases but not metastases in the lung. This enhanced activity results from both local recruitment of innate immune effectors as well as concentration and increased activation of vaccine-induced antitumor T cells within the liver. These findings show a general approach to focus systemic cancer immunotherapies to specific organs bearing tumor metastases by taking advantage of differential tropisms and the proinflammatory nature of microbes.


Assuntos
Vacinas Anticâncer/imunologia , Engenharia Genética , Listeria monocytogenes/genética , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/terapia , Animais , Feminino , Hepatite/virologia , Humanos , Imunoterapia/métodos , Inflamação , Listeria monocytogenes/patogenicidade , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T
9.
Proc Natl Acad Sci U S A ; 101(38): 13832-7, 2004 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-15365184

RESUMO

The facultative intracellular bacterium Listeria monocytogenes is being developed as a cancer vaccine platform because of its ability to induce potent innate and adaptive immunity. For successful clinical application, it is essential to develop a Listeria platform strain that is safe yet retains the potency of vaccines based on wild-type bacteria. Here, we report the development of a recombinant live-attenuated vaccine platform strain that retains the potency of the fully virulent pathogen, combined with a >1,000-fold reduction in toxicity, as compared with wild-type Listeria. By selectively deleting two virulence factors, ActA (DeltaactA) and Internalin B (DeltainlB), the immunopotency of Listeria was maintained and its toxicity was diminished in vivo, largely by blocking the direct internalin B-mediated infection of nonphagocytic cells, such as hepatocytes, and the indirect ActA-mediated infection by cell-to-cell spread from adjacent phagocytic cells. In contrast, infection of phagocytic cells was not affected, leaving intact the ability of Listeria to stimulate innate immunity and to induce antigenspecific cellular responses. Listeria DeltaactA/DeltainlB-based vaccines were rapidly cleared from mice after immunization and induced potent and durable effector and memory T-cell responses with no measurable liver toxicity. Therapeutic vaccination of BALB/c mice bearing murine CT26 colon tumor lung metastases or palpable s.c. tumors (>100 mm(3)) with recombinant Listeria DeltaactA/DeltainlB expressing an endogenous tumor antigen resulted in breaking of self-tolerance and long-term survival. We propose that recombinant Listeria DeltaactA/DeltainlB expressing human tumor-associated antigens represents an attractive therapeutic strategy for further development and testing in human clinical trials.


Assuntos
Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Neoplasias do Colo/imunologia , Hepatócitos/imunologia , Listeria monocytogenes/imunologia , Neoplasias Pulmonares/imunologia , Monócitos/imunologia , Linfócitos T/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Deleção de Genes , Hepatócitos/efeitos dos fármacos , Humanos , Memória Imunológica , Listeria monocytogenes/genética , Neoplasias Pulmonares/secundário , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/efeitos dos fármacos , RNA Bacteriano/genética , RNA de Transferência de Arginina/genética , Linfócitos T/efeitos dos fármacos
10.
J Immunol ; 173(1): 420-7, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15210801

RESUMO

The bacterium L. monocytogenes is a proposed vaccine carrier based upon the observation that this pathogen replicates within the intracytoplasmic environment facilitating delivery of Ag to the endogenous Ag processing and presentation pathway with subsequent stimulation of peptide specific MHC class I-restricted CD8(+) effector cells. In this report, we evaluate virulence-attenuated strains of Listeria monocytogenes as vaccine vectors and examine whether existing antivector (antilisterial) immunity limits or alters its efficacy as a therapeutic cancer vaccine. Following immunization with virulence-attenuated mutants, we found that the effectiveness of L. monocytogenes as a recombinant cancer vaccine remains intact. In addition, we found that antibiotic treatment initiated 24 or 36 h following therapeutic immunization with recombinant L. monocytogenes allows full development of the antitumor response. We also demonstrate that the vaccine vector potential of L. monocytogenes is not limited in animals with existing antilisterial immunity. For these latter studies, mice previously immunized with wild-type L. monocytogenes were infused with melanoma cells and then 5 days later challenged with recombinant tumor Ag expressing L. monocytogenes. Collectively, these results add additional support for the use of L. monocytogenes as a vaccine vector and underscore its potential to be used repeatedly for stimulation of recall responses concomitant with primary cell-mediated responses to newly delivered heterologous tumor-associated epitopes.


Assuntos
Vacinas Anticâncer/uso terapêutico , Vetores Genéticos , Oxirredutases Intramoleculares/genética , Listeria monocytogenes/genética , Melanoma Experimental/terapia , Vacinas Sintéticas/uso terapêutico , Animais , Antibacterianos/farmacologia , Linhagem Celular , Feminino , Oxirredutases Intramoleculares/imunologia , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Virulência
11.
Cancer Cell ; 4(4): 241-3, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14585348

RESUMO

Selected mutant strains of vesicular stomatitis virus (VSV) are described that are unable to combat endogenous IFN-beta signaling within infected normal cells and as a result are dramatically more selective for productive growth in tumor cells having a defective antiviral response. The VSV mutants may have the potential to be used clinically as a systemic oncolytic agent for the treatment of distal and metastatic cancers.


Assuntos
Interferon beta/metabolismo , Infecções por Vírus de RNA/terapia , Vírus da Estomatite Vesicular Indiana/fisiologia , Vírus/metabolismo , Animais , Humanos , Interferon beta/antagonistas & inibidores , Mutação , Neoplasias/terapia , Neoplasias/virologia , Infecções por Vírus de RNA/virologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA