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.
J Immunol ; 201(7): 1837-1841, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30135180

RESUMO

Various malignancies are reproducibly cured in mouse models, but most cancer immunotherapies show objective responses in a fraction of treated patients. One reason for this disconnect may be the use of young, lean mice lacking immune-altering comorbidities present in cancer patients. Although many cancer patients are overweight or obese, the effect of obesity on antitumor immunity is understudied in preclinical tumor models. We examined the effect of obesity on two immunotherapeutic models: systemic anti-CTLA-4 mAb and intratumoral delivery of a TRAIL-encoding adenovirus plus CpG. Both therapies were effective in lean mice, but neither provided a survival benefit to diet-induced obese BALB/c mice. Interestingly, tumor-bearing leptin-deficient (ob/ob) obese BALB/c mice did respond to treatment. Moreover, reducing systemic leptin with soluble leptin receptor:Fc restored the antitumor response in diet-induced obese mice. These data demonstrate the potential of targeting leptin to improve tumor immunotherapy when immune-modulating comorbidities are present.


Assuntos
Adenocarcinoma/metabolismo , Envelhecimento/fisiologia , Anticorpos Monoclonais/uso terapêutico , Imunoterapia/métodos , Neoplasias Renais/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Adenocarcinoma/terapia , Adenoviridae/genética , Animais , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Dieta , Modelos Animais de Doenças , Feminino , Humanos , Imunidade , Neoplasias Renais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Obesidade/terapia , Oligodesoxirribonucleotídeos/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
2.
Biomaterials ; 164: 38-53, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29482062

RESUMO

Cytotoxic T lymphocytes (CTLs) play a major role in cancer immunotherapy because of their ability to directly kill tumor cells and secrete tumor suppressive cytokines. Anticancer vaccines aim to provoke tumor-specific CTL responses, which require activation of antigen presenting cells (APCs) including dendritic cells (DCs) and macrophages. Therefore, a potent immunostimulatory adjuvant capable of activating APCs is an essential component of anticancer vaccines. In this study, we introduce novel TLR 7/8 bi-specific agonists that significantly enhance cytokine secretion compared to TLR7 mono-selective compounds. Encapsulation of these TLR 7/8 agonists in poly(lactide-co-glycolide) (PLGA) nanoparticles increased the co-stimulatory molecule expression and antigen presentation via MHC I by DCs compared to the soluble agonist. When administered subcutaneously, these nanoparticles migrated to draining lymph node and triggered DC activation and expansion. This lead to expansion of antigen-specific CD8 T cells and enhanced CTL response, which resulted in significant prophylactic and therapeutic efficacy in melanoma, bladder and renal cell carcinoma tumor models. Importantly, our studies demonstrate significant reductions in systemic metastasis with the nanoparticle vaccine. Our results suggest novel TLR 7/8 agonist-encapsulated nanoparticles are potent immunostimulatory adjuvants for cancer immunotherapy.


Assuntos
Portadores de Fármacos , Imidazóis/farmacologia , Nanocápsulas , Poliglactina 910 , Quinolinas/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Adjuvantes Imunológicos/farmacologia , Animais , Antineoplásicos/farmacologia , Portadores de Fármacos/química , Feminino , Humanos , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Linfócitos T Citotóxicos/efeitos dos fármacos
3.
J Vis Exp ; (122)2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28448047

RESUMO

Renal cell carcinoma (RCC) affects > 60,000 people in the United States annually, and ~ 30% of RCC patients have multiple metastases at the time of diagnosis. Metastatic RCC (mRCC) is incurable, with a median survival time of only 18 months. Immune-based interventions (e.g., interferon (IFN) and interleukin (IL)-2) induce durable responses in a fraction of mRCC patients, and multikinase inhibitors (e.g., sunitinib or sorafenib) or anti-VEGF receptor monoclonal antibodies (mAb) are largely palliative, as complete remissions are rare. Such shortcomings in current therapies for mRCC patients provide the rationale for the development of novel treatment protocols. A key component in the preclinical testing of new therapies for mRCC is a suitable animal model. Beneficial features that recapitulate the human condition include a primary renal tumor, renal tumor metastases, and an intact immune system to investigate any therapy-driven immune effector responses and the formation of tumor-induced immunosuppressive factors. This report describes an orthotopic mRCC mouse model that has all of these features. We describe an intrarenal implantation technique using the mouse renal adenocarcinoma cell line Renca, followed by the assessment of tumor growth in the kidney (primary site) and lungs (metastatic site).


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Neoplasias Renais/patologia , Neoplasias Experimentais/patologia , Animais , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/secundário , Modelos Animais de Doenças , Neoplasias Renais/tratamento farmacológico , Camundongos , Neoplasias Experimentais/tratamento farmacológico
4.
FEBS J ; 282(24): 4747-4765, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26426449

RESUMO

Renal cell carcinoma (RCC) is resistant to traditional cancer therapies, and metastatic RCC (mRCC) is incurable. The shortcomings in current therapeutic options for patients with mRCC provide the rationale for the development of novel treatment protocols. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has proven to be a potent inducer of tumor cell death in vitro and in vivo, and a number of TRAIL death receptor agonists (recombinant TRAIL or TRAIL death receptor-specific mAb) have been developed and tested clinically. Unfortunately the clinical efficacy of TRAIL has been underwhelming and is likely due to a number of possible mechanisms that render tumors resistant to TRAIL, prompting the search for drugs that increase tumor cell susceptibility to TRAIL. The objective of this study was to determine the effectiveness of combining the diterpene triepoxide triptolide, or its water-soluble prodrug, Minnelide, with TRAIL receptor agonists against RCC in vitro or in vivo, respectively. TRAIL-induced apoptotic death of human RCC cells was increased in the presence of triptolide. The triptolide-induced sensitization was accompanied by increased TRAIL-R2 (DR5) and decreased heat shock protein 70 expression. In vivo treatment of mice bearing orthotopic RCC (Renca) tumors showed the combination of Minnelide and agonistic anti-DR5 mAb significantly decreased tumor burden and increased animal survival compared to either therapy alone. Our data suggest triptolide/Minnelide sensitizes RCC cells to TRAIL-induced apoptosis through altered TRAIL death receptor and heat shock protein expression.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Diterpenos/farmacologia , Neoplasias Renais/tratamento farmacológico , Fenantrenos/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Diterpenos/uso terapêutico , Sinergismo Farmacológico , Compostos de Epóxi/farmacologia , Compostos de Epóxi/uso terapêutico , Feminino , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Organofosfatos/farmacologia , Organofosfatos/uso terapêutico , Fenantrenos/uso terapêutico , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Organismos Livres de Patógenos Específicos , Análise de Sobrevida , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Carga Tumoral/efeitos dos fármacos
5.
Hum Vaccin Immunother ; 11(7): 1612-20, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996049

RESUMO

Clinical observations of spontaneous disease regression in some renal cell carcinoma (RCC) patients implicate a role for tumor immunity in controlling this disease. Puzzling, however, are findings that high levels of tumor infiltrating lymphocytes (TIL) are common to RCC. Despite expression of activation markers by TILs, functional impairment of innate and adaptive immune cells has been consistently demonstrated contributing to the failure of the immune system to control RCC. Immunotherapy can overcome the immunosuppressive effects of the tumor and provide an opportunity for long-term disease free survival. Unfortunately, complete response rates remain sub-optimal indicating the effectiveness of immunotherapy remains limited by tumor-specific factors and/or cell types that inhibit antitumor immune responses. Here we discuss immunotherapies and the function of multiple immune system components to achieve an effective response. Understanding these complex interactions is essential to rationally develop novel therapies capable of renewing the immune system's ability to respond to these tumors.


Assuntos
Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/terapia , Imunidade Inata , Neoplasias Renais/imunologia , Neoplasias Renais/terapia , Carcinoma de Células Renais/secundário , Humanos , Imunoterapia , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Metástase Neoplásica/imunologia , Linfócitos T/imunologia
6.
J Immunol ; 194(4): 1609-20, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595784

RESUMO

Patients surviving the acute stages of sepsis develop compromised T cell immunity and increased susceptibility to infection. Little is known about the decreased CD4 T cell function after sepsis. We tracked the loss and recovery of endogenous Ag-specific CD4 T cell populations after cecal ligation and puncture-induced sepsis and analyzed the CD4 T cell response to heterologous infection during or after recovery. We observed that the sepsis-induced early loss of CD4 T cells was followed by thymic-independent numerical recovery in the total CD4 T cell compartment. Despite this numerical recovery, we detected alterations in the composition of naive CD4 T cell precursor pools, with sustained quantitative reductions in some populations. Mice that had experienced sepsis and were then challenged with epitope-bearing, heterologous pathogens demonstrated significantly reduced priming of recovery-impaired Ag-specific CD4 T cell responses, with regard to both magnitude of expansion and functional capacity on a per-cell basis, which also correlated with intrinsic changes in Vß clonotype heterogeneity. Our results demonstrate that the recovery of CD4 T cells from sepsis-induced lymphopenia is accompanied by alterations to the composition and function of the Ag-specific CD4 T cell repertoire.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Precursoras de Linfócitos T/imunologia , Sepse/imunologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Infecções Bacterianas/imunologia , Linfócitos T CD4-Positivos/citologia , Modelos Animais de Doenças , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Linfócitos T/citologia , Subpopulações de Linfócitos T/citologia , Timo/citologia , Timo/imunologia
7.
Cancer Immunol Immunother ; 63(11): 1213-27, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25143233

RESUMO

Tumor progression occurs through the modulation of a number of physiological parameters, including the development of immunosuppressive mechanisms to prevent immune detection and response. Among these immune evasion mechanisms, the mobilization of myeloid-derived suppressor cells (MDSC) is a major contributor to the suppression of antitumor T-cell immunity. Patients with renal cell carcinoma (RCC) show increased MDSC, and methods are being explored clinically to reduce the prevalence of MDSC and/or inhibit their function. In the present study, we investigated the relationship between MDSC and the therapeutic potential of a TRAIL-encoding recombinant adenovirus (Ad5-TRAIL) in combination with CpG-containing oligodeoxynucleotides (Ad5-TRAIL/CpG) in an orthotopic mouse model of RCC. This immunotherapy effectively clears renal (Renca) tumors and enhances survival, despite the presence of a high frequency of MDSC in the spleens and primary tumor-bearing kidneys at the time of treatment. Subsequent analyses revealed that the CpG component of the immunotherapy was responsible for decreasing the frequency of MDSC in Renca-bearing mice; further, treatment with CpG modulated the phenotype and function of MDSC that remained after immunotherapy and correlated with an increased T-cell response. Interestingly, the CpG-dependent alterations in MDSC frequency and function did not occur in tumor-bearing mice complicated with diet-induced obesity. Collectively, these data suggest that in addition to its adjuvant properties, CpG also enhances antitumor responses by altering the number and function of MDSC.


Assuntos
Carcinoma de Células Renais/terapia , Imunoterapia/métodos , Neoplasias Renais/terapia , Oligodesoxirribonucleotídeos/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Adenoviridae/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Carcinoma de Células Renais/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Rim/metabolismo , Neoplasias Renais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/citologia , Obesidade , Oligonucleotídeos , Fenótipo , Baço/metabolismo , Linfócitos T/citologia
8.
Cancer Immunol Immunother ; 63(7): 685-97, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24711083

RESUMO

It is now appreciated that there are distinct subsets of dendritic cells (DC) with specialized functions. Plasmacytoid DC (pDC) and CD8α DC can contribute to the priming, activation and function of antitumor CD8 T cells; however, their specific roles and necessity in stimulating antitumor immunity are not clearly understood. We examined the importance of pDC and CD8α DC during immunotherapy of an orthotopic model of metastatic renal cell carcinoma. Immunotherapy that utilizes a recombinant adenovirus encoding tumor necrosis factor-related apoptosis-inducing ligand (Ad5-TRAIL) in combination with an immunostimulatory CpG-containing oligodeoxynucleotide (CpG) resulted in the clearance of primary and metastatic tumors in wild-type (WT) replete BALB/c mice and prolonged survival. In comparison, mice deficient in either pDC (accomplished using a depleting mAb specific for PDCA1) or CD8α DC (through utilization of CD8α DC-deficient Batf3(-/-) BALB/c mice) had uncontrolled tumor growth and high mortality after Ad5-TRAIL/CpG administration. The ineffectiveness of Ad5-TRAIL/CpG therapy in the anti-PDCA1-treated and Batf3(-/-) BALB/c mice was marked by an altered activation phenotype of the DC, as well as significantly reduced expression of type I IFN-stimulated genes and IL-15/IL-15R complex production. In addition, pDC-depleted and Batf3(-/-) BALB/c mice had significantly decreased effector CD8 T cell infiltration in the primary tumor site compared with WT mice after therapy. These data collectively suggest that pDC and CD8α DC carry out independent, but complementary, roles that are necessary to initiate an efficacious antitumor immune response after Ad5-TRAIL/CpG therapy.


Assuntos
Antígenos CD8/imunologia , Carcinoma de Células Renais/tratamento farmacológico , Células Dendríticas/imunologia , Imunoterapia/métodos , Neoplasias Renais/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Carcinoma de Células Renais/imunologia , Linhagem Celular Tumoral , Células Dendríticas/citologia , Feminino , Neoplasias Renais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
9.
Nat Protoc ; 9(1): 209-22, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24385150

RESUMO

Characterization of the cellular participants in tissue immune responses is crucial to understanding infection, cancer, autoimmunity, allergy, graft rejection and other immunological processes. Previous reports indicate that leukocytes in lung vasculature fail to be completely removed by perfusion. Several studies suggest that intravascular staining may discriminate between tissue-localized and blood-borne cells in the mouse lung. Here we outline a protocol for the validation and use of intravascular staining to define innate and adaptive immune cells in mice. We demonstrate application of this protocol to leukocyte analyses in many tissues and we describe its use in the contexts of lymphocytic choriomeningitis virus and Mycobacterium tuberculosis infections or solid tumors. Intravascular staining and organ isolation usually takes 5-30 min per mouse, with additional time required for any subsequent leukocyte isolation, staining and analysis. In summary, this simple protocol should help enable interpretable analyses of tissue immune responses.


Assuntos
Leucócitos/citologia , Pulmão/citologia , Imunidade Adaptativa , Animais , Linfócitos B/citologia , Biomarcadores , Permeabilidade Capilar , Imunidade Inata , Pulmão/imunologia , Camundongos , Mycobacterium tuberculosis/imunologia , Coloração e Rotulagem/métodos
10.
Leuk Lymphoma ; 54(11): 2500-5, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23452151

RESUMO

Both complement and antibody-dependent cellular cytotoxicity (ADCC) contribute to the clinical efficacy of anti-CD20 monoclonal antibody (mAb) therapy. Paradoxically, the C3b component of complement can block interaction between mAb and natural killer (NK) cells. The present study compared the effect of complement on the ability of two anti-CD20 mAbs, rituximab and GA101, to activate NK cells and mediate ADCC. Complement blocked adherence of NK cells to rituximab, but had little effect on NK binding to GA101. Target cells coated with rituximab or GA101 were able to activate NK cells in the absence of serum. Complement in serum blocked NK activation induced by rituximab, but not GA101. Complement blocked rituximab-induced NK-cell mediated ADCC, but not GA101-induced ADCC. These results demonstrate that the decreased ability of GA101 to fix complement relative to rituximab results in an enhanced ability of GA101 to bind to NK cells, activate NK cells and induce ADCC when serum is present.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Murinos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Proteínas do Sistema Complemento/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais Murinos/metabolismo , Linhagem Celular , Complemento C1q/imunologia , Complemento C1q/metabolismo , Complemento C5/imunologia , Complemento C5/metabolismo , Proteínas do Sistema Complemento/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Ligação Proteica/imunologia , Rituximab
11.
J Immunol ; 190(5): 1991-2000, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23355736

RESUMO

Patients who survive severe sepsis often display compromised immune function with impairment in innate and adaptive immune responses. These septic patients are highly susceptible to "secondary" infections with intracellular pathogens that are usually controlled by CD8(+) T cells. It is not known when and if this observed immunoparalysis of CD8(+) T cell immunity recovers, and the long-term consequences of sepsis on the ability of naive CD8(+) T cells to respond to subsequent infections are poorly understood. In this study, using the cecal-ligation and puncture mouse model of sepsis, we show that sepsis induces a rapid loss of naive CD8(+) T cells. However, IL-15-dependent numerical recovery is observed a month after initial septic insult. Numerical recovery is accompanied by IL-15-dependent phenotypic changes where a substantial proportion of naive (Ag-inexperienced) CD8(+) T cells display a "memory-like" phenotype (CD44(hi)/CD11a(hi)). Importantly, the impairment of naive CD8(+) T cells to respond to viral and bacterial infection was sustained for month(s) after sepsis induction. Incomplete recovery of naive CD8(+) T cell precursors was observed in septic mice, suggesting that the availability of naive precursors contributes to the sustained impairment in primary CD8(+) T cell responses. Thus, sepsis can result in substantial and long-lasting changes in the available CD8(+) T cell repertoire affecting the capacity of the host to respond to new infections.


Assuntos
Linfócitos T CD8-Positivos/patologia , Convalescença , Listeriose/patologia , Coriomeningite Linfocítica/patologia , Células Precursoras de Linfócitos T/patologia , Sepse/patologia , Vacínia/patologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Celular , Memória Imunológica , Interleucina-15/biossíntese , Interleucina-15/imunologia , Listeria monocytogenes/fisiologia , Listeriose/complicações , Listeriose/imunologia , Listeriose/microbiologia , Contagem de Linfócitos , Coriomeningite Linfocítica/complicações , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Camundongos Transgênicos , Células Precursoras de Linfócitos T/imunologia , Sepse/complicações , Sepse/imunologia , Fatores de Tempo , Vacínia/complicações , Vacínia/imunologia , Vacínia/virologia , Vaccinia virus/fisiologia
12.
Oncoimmunology ; 1(7): 1178-1180, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23170271

RESUMO

TNF-related apoptosis-inducing ligand (TRAIL) continues to be intently studied as a cancer therapeutic because of its selective tumoricial activity. We have been interesting in evaluating the ability of TRAIL to induce systemic antitumor immunity through the generation of apoptotic tumor cells. Recent observations suggest that localized administration of TRAIL in combination with CpG ODN generates a systemic antitumor immune response to eliminate the primary tumor and distant metastases.

13.
J Immunol ; 189(3): 1311-21, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22745381

RESUMO

Obesity is a mounting health concern in the United States and is associated with an increased risk for developing several cancers, including renal cell carcinoma (RCC). Despite this, little is known regarding the impact of obesity on antitumor immunity. Because dendritic cells (DC) are critical regulators of antitumor immunity, we examined the combined effects of obesity and tumor outgrowth on DC function. Using a diet-induced obesity (DIO) model, DC function was evaluated in mice bearing orthotopic RCC and in tumor-free controls. Tumor-free DIO mice had profoundly altered serum cytokine and chemokine profiles, with upregulation of 15 proteins, including IL-1α, IL-17, and LIF. Tumor-free DIO mice had elevated percentages of conventional splenic DC that were impaired in their ability to stimulate naive T cell expansion, although they were phenotypically similar to normal weight (NW) controls. In DIO mice, intrarenal RCC tumor challenge in the absence of therapy led to increased local infiltration by T cell-suppressive DC and accelerated early tumor outgrowth. Following administration of a DC-dependent immunotherapy, established RCC tumors regressed in normal weight mice. The same immunotherapy was ineffective in DIO mice and was characterized by an accumulation of regulatory DC in tumor-bearing kidneys, decreased local infiltration by IFN-γ-producing CD8 T cells, and progressive tumor outgrowth. Our results suggest that the presence of obesity as a comorbidity can impair the efficacy of DC-dependent antitumor immunotherapies.


Assuntos
Adenocarcinoma/imunologia , Células Dendríticas/imunologia , Gorduras na Dieta/administração & dosagem , Neoplasias Renais/imunologia , Obesidade/imunologia , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Células Dendríticas/patologia , Feminino , Interpretação de Imagem Assistida por Computador , Neoplasias Renais/diagnóstico , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Obesidade/etiologia , Obesidade/patologia , Distribuição Aleatória
14.
PLoS One ; 7(2): e31085, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22312440

RESUMO

Despite evidence that antitumor immunity can be protective against renal cell carcinoma (RCC), few patients respond objectively to immunotherapy and the disease is fatal once metastases develop. We asked to what extent combinatorial immunotherapy with Adenovirus-encoded murine TNF-related apoptosis-inducing ligand (Ad5mTRAIL) plus CpG oligonucleotide, given at the primary tumor site, would prove efficacious against metastatic murine RCC. To quantitate primary renal and metastatic tumor growth in mice, we developed a luciferase-expressing Renca cell line, and monitored tumor burdens via bioluminescent imaging. Orthotopic tumor challenge gave rise to aggressive primary tumors and lung metastases that were detectable by day 7. Intra-renal administration of Ad5mTRAIL+CpG on day 7 led to an influx of effector phenotype CD4 and CD8 T cells into the kidney by day 12 and regression of established primary renal tumors. Intra-renal immunotherapy also led to systemic immune responses characterized by splenomegaly, elevated serum IgG levels, increased CD4 and CD8 T cell infiltration into the lungs, and elimination of metastatic lung tumors. Tumor regression was primarily dependent upon CD8 T cells and resulted in prolonged survival of treated mice. Thus, local administration of Ad5mTRAIL+CpG at the primary tumor site can initiate CD8-dependent systemic immunity that is sufficient to cause regression of metastatic lung tumors. A similar approach may prove beneficial for patients with metastatic RCC.


Assuntos
Adenoviridae/genética , Carcinoma de Células Renais/terapia , Ilhas de CpG/genética , Imunoterapia/métodos , Neoplasias Renais/terapia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Animais , Antígenos CD8/metabolismo , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Estudos de Viabilidade , Feminino , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
J Virol ; 85(14): 6822-31, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21593179

RESUMO

The absence of interleukin-10 (IL-10), a potent anti-inflammatory cytokine results in increased immune-mediated demyelination in mice infected with a neurotropic coronavirus (recombinant J2.2-V-1 [rJ2.2]). Here, we examined the therapeutic effects of increased levels of IL-10 at early times after infection by engineering a recombinant J2.2 virus to produce IL-10. We demonstrate that viral expression of IL-10, which occurs during the peak of virus replication and at the site of disease, enhanced survival and diminished morbidity in rJ2.2-infected wild-type B6 and IL-10(-/-) mice. The protective effects of increased IL-10 levels were associated with reductions in microglial activation, inflammatory cell infiltration into the brain, and proinflammatory cytokine and chemokine production. Additionally, IL-10 increased both the frequency and number of Foxp3(+) regulatory CD4 T cells in the infected central nervous system. Most strikingly, the ameliorating effects of IL-10 produced during the first 5 days after infection were long acting, resulting in decreased demyelination during the resolution phase of the infection. Collectively, these results suggest that the pathogenic processes that result in demyelination are initiated early during infection and that they can be diminished by exogenous IL-10 delivered soon after disease onset. IL-10 functions by dampening the innate or very early T cell immune response. Further, they suggest that early treatment with IL-10 may be useful adjunct therapy in some types of viral encephalitis.


Assuntos
Coronavirus/genética , Doenças Desmielinizantes/prevenção & controle , Encefalomielite/prevenção & controle , Interleucina-10/fisiologia , Doença Aguda , Animais , Sequência de Bases , Doença Crônica , Primers do DNA , Doenças Desmielinizantes/imunologia , Encefalomielite/imunologia , Citometria de Fluxo , Interleucina-10/genética , Camundongos , Proteínas Recombinantes/metabolismo , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Cancers (Basel) ; 3(1): 603-20, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-24212631

RESUMO

Numerous biologic approaches are being investigated as anti-cancer therapies in an attempt to induce tumor regression while circumventing the toxic side effects associated with standard chemo- or radiotherapies. Among these, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has shown particular promise in pre-clinical and early clinical trials, due to its preferential ability to induce apoptotic cell death in cancer cells and its minimal toxicity. One limitation of TRAIL use is the fact that many tumor types display an inherent resistance to TRAIL-induced apoptosis. To circumvent this problem, researchers have explored a number of strategies to optimize TRAIL delivery and to improve its efficacy via co-administration with other anti-cancer agents. In this review, we will focus on TRAIL-based gene therapy approaches for the treatment of malignancies. We will discuss the main viral vectors that are being used for TRAIL gene therapy and the strategies that are currently being attempted to improve the efficacy of TRAIL as an anti-cancer therapeutic.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...