Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Macromol Biosci ; 22(2): e2100356, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34822219

RESUMO

Cancer vaccination is a powerful strategy to combat cancer. A very attractive approach to prime the immune system against cancer cells involves the use of tumor lysate as antigen source. The immunogenicity of tumor lysate can be further enhanced by treatment with hypochlorous acid. This study explores poly(lactic-co-glycolic acid) (PLGA) nanoparticles to enhance the delivery of oxidized tumor lysate to dendritic cells. Using human donor-derived dendritic cells, it is found that the use of PLGA nanoparticles enhances antigen uptake and dendritic cell maturation, as compared to the use of the free tumor lysate. The ability of the activated dendritic cells to stimulate autologous peripheral blood mononuclear cells (PBMCs) is assessed in vitro by coculturing PBMCs with A375 melanoma cells. Live cell imaging analysis of this experiment highlights the potential of nanoparticle-mediated dendritic-cell-based vaccination approaches. Finally, the efficacy of the PLGA nanoparticle formulation is evaluated in vivo in a therapeutic vaccination study using B16F10 tumor-bearing C57BL/6J mice. Animals that are challenged with the polymer nanoparticle-based oxidized tumor lysate formulation survive for up to 50 days, in contrast to a maximum of 41 days for the group that receives the corresponding free oxidized tumor lysate-based vaccine.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Animais , Células Dendríticas , Leucócitos Mononucleares , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
2.
Front Immunol ; 12: 695150, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149738

RESUMO

The emergence of ablative therapies has revolutionized the treatment of inoperable solid tumors. Cryoablation stands out for its uniqueness of operation based on hypothermia, and for its ability to unleash the native tumor antigens, resulting in the generation of anti-tumor immune responses. It is not clearly understood how alterations in the rate of freeze impact the immune response outcomes. In this study, we tested fast freeze and slow freeze rates for their locoregional effectiveness and their ability to elicit immune responses in a B16F10 mouse model of melanoma. Tumor bearing mice treated with fast freeze protocol survived better than the ones treated with slow freeze protocol. Fast freeze resulted in a higher magnitude of CD4+ and CD8+ T-cell responses, and a significantly extended survival post re-challenge. Thus, fast freeze rate should be applied in any future studies employing cryoablation as an in vivo vaccination tool in conjunction with targeted immunotherapies.


Assuntos
Criocirurgia , Melanoma Experimental/cirurgia , Neoplasias Cutâneas/cirurgia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Congelamento , Cinética , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Células Mieloides/imunologia , Células Mieloides/metabolismo , Necrose , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Carga Tumoral
4.
Curr Opin Biotechnol ; 65: 60-64, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32088576

RESUMO

Cryoablation is a hypothermic modality exercised as a focal therapy for annihilating cancer lesions. Its application leaves the ablated tumor in situ, allowing multifarious tumor antigens to be available to the host's immune system. This ensues the activation of innate and adaptive immunity against the tumor antigens. Therefore, cryoablation can be employed as an in vivo vaccination tool to fortify the impact of immunotherapies. Application of checkpoint inhibitors, toll-like receptor agonists, adoptive cell therapies and epigenetic modulators has been shown to galvanize the immune system against tumors. Preliminary data demonstrate an excellent synergy between cryoablation and immunotherapies. Future endeavors should focus on tailoring cryo-immunotherapies based on the tumor's immune signature and testing alternative approaches to circumvent treatment-associated toxicities and maximize efficacy.


Assuntos
Criocirurgia , Neoplasias , Antígenos de Neoplasias , Humanos , Sistema Imunitário , Imunoterapia , Neoplasias/terapia
5.
Front Immunol ; 10: 2283, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31608067

RESUMO

Treatment of solid tumors by ablation techniques has gained momentum in the recent years due to their technical simplicity and reduced morbidity as juxtaposed to surgery. Cryoablation is one of such techniques, known for its uniqueness to destroy the tumors by freezing to lethal temperatures. Freezing the tumor locally and allowing it to remain in situ unleashes an array of tumor antigens to be exposed to the immune system, paving the way for the generation of anti-tumor immune responses. However, the immune responses triggered in most cases are insufficient to eradicate the tumors with systemic spread. Therefore, combination of cryoablation and immunotherapy is a new treatment strategy currently being evaluated for its efficacy, notably in patients with metastatic disease. This article examines the mechanistic fabric of cryoablation for the generation of an effective immune response against the tumors, and various possibilities of its combination with different immunotherapies that are capable of inducing exceptional therapeutic responses. The combinatorial treatment avenues discussed in this article if explored in sufficient profundity, could reach the pinnacle of future cancer medicine.


Assuntos
Antígenos de Neoplasias/imunologia , Criocirurgia/métodos , Sistema Imunitário/imunologia , Imunoterapia/métodos , Neoplasias/terapia , Ensaios Clínicos como Assunto , Terapia Combinada , Humanos , Neoplasias/imunologia , Resultado do Tratamento
6.
Proc Natl Acad Sci U S A ; 116(22): 10943-10951, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31097580

RESUMO

Here, we highlight the potential translational benefits of delivering FLASH radiotherapy using ultra-high dose rates (>100 Gy⋅s-1). Compared with conventional dose-rate (CONV; 0.07-0.1 Gy⋅s-1) modalities, we showed that FLASH did not cause radiation-induced deficits in learning and memory in mice. Moreover, 6 months after exposure, CONV caused permanent alterations in neurocognitive end points, whereas FLASH did not induce behaviors characteristic of anxiety and depression and did not impair extinction memory. Mechanistic investigations showed that increasing the oxygen tension in the brain through carbogen breathing reversed the neuroprotective effects of FLASH, while radiochemical studies confirmed that FLASH produced lower levels of the toxic reactive oxygen species hydrogen peroxide. In addition, FLASH did not induce neuroinflammation, a process described as oxidative stress-dependent, and was also associated with a marked preservation of neuronal morphology and dendritic spine density. The remarkable normal tissue sparing afforded by FLASH may someday provide heretofore unrealized opportunities for dose escalation to the tumor bed, capabilities that promise to hasten the translation of this groundbreaking irradiation modality into clinical practice.


Assuntos
Disfunção Cognitiva , Neuroproteção/efeitos da radiação , Doses de Radiação , Radioterapia/métodos , Espécies Reativas de Oxigênio/metabolismo , Animais , Encéfalo/patologia , Encéfalo/efeitos da radiação , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/prevenção & controle , Feminino , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Radioterapia/efeitos adversos , Espécies Reativas de Oxigênio/análise
7.
Br J Radiol ; 92(1093): 20180008, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29694234

RESUMO

Normal tissue damages induced by radiation therapy remain dose-limiting factors in radiation oncology and this is still true despite recent advances in treatment planning and delivery of image-guided radiation therapy. Additionally, as the number of long-term cancer survivors increases, unacceptable complications emerge and dramatically reduce the patients' quality of life. This means that patients and clinicians expect discovery of new options for the therapeutic management of radiation-induced complications. Over the past four decades, research has enhanced our understanding of the pathophysiological, cellular and molecular processes governing normal tissue toxicity. Those processes are complex and involve the cross-talk between the various cells of a tissue, including fibroblasts, endothelial, immune and epithelial cells as well as soluble paracrine factors including growth factors and proteases. We will review the translatable pharmacological approaches that have been developed to prevent, mitigate, or reverse radiation injuries based upon the targeting of cellular and signalling pathways. We will summarize the different steps of the research strategy, from the definition of initial biological hypotheses to preclinical studies and clinical translation. We will also see how novel research and therapeutic hypotheses emerge along the way as well as briefly highlight innovative approaches based upon novel radiotherapy delivery procedures.


Assuntos
Neoplasias/radioterapia , Oxirredução/efeitos dos fármacos , Segurança do Paciente , Lesões por Radiação/prevenção & controle , Radioterapia Guiada por Imagem/métodos , Animais , Morte Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Neoplasias/patologia , Terapia com Prótons/efeitos adversos , Terapia com Prótons/métodos , Radioterapia/efeitos adversos , Radioterapia/métodos , Valores de Referência
8.
Cell Stem Cell ; 21(2): 225-240.e5, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28736216

RESUMO

Bacterial infection leads to consumption of short-lived innate immune effector cells, which then need to be replenished from hematopoietic stem and progenitor cells (HSPCs). HSPCs express pattern recognition receptors, such as Toll-like receptors (TLRs), and ligation of these receptors induces HSPC mobilization, cytokine production, and myeloid differentiation. The underlying mechanisms involved in pathogen signal transduction in HSCs and the resulting biological consequences remain poorly defined. Here, we show that in vivo lipopolysaccharide (LPS) application induces proliferation of dormant HSCs directly via TLR4 and that sustained LPS exposure impairs HSC self-renewal and competitive repopulation activity. This process is mediated via TLR4-TRIF-ROS-p38, but not MyD88 signaling, and can be inhibited pharmacologically without preventing emergency granulopoiesis. Live Salmonella Typhimurium infection similarly induces proliferative stress in HSCs, in part via TLR4-TRIF signals. Thus, while direct TLR4 activation in HSCs might be beneficial for controlling systemic infection, prolonged TLR4 signaling has detrimental effects and may contribute to inflammation-associated HSPC dysfunction.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Imunidade Inata , Salmonella typhimurium/fisiologia , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Salmonella typhimurium/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Gastric Cancer ; 20(1): 116-125, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26782287

RESUMO

BACKGROUND: An increase of regulatory T cells, defined as CD25high- and/or FOXP3+-expressing CD4+ T cells, within tumors has been reported in several studies. Tregs promote tumor growth by modulating the antitumor immune response, mainly through inhibition of T-cell-mediated tumor cell killing: this has been suggested to be dependent on IL-10 and/or TGF-ß. In stomach cancer, the mechanisms behind the accumulation of Tregs in tumor tissue has not been fully elucidated, and neither has Treg gene expression in situ. MATERIALS AND METHODS: Stomach tissue from gastric cancer patients undergoing gastric resection was analyzed using flow cytometry and cell sorting, followed by RT-PCR. RESULTS: We observed that stomach CD4+ FOXP3+ T cells proliferated to a higher degree than CD4+ FOXP3- T cells, which may contribute to Treg accumulation in the mucosa. By analyzing DNA methylation, we demonstrated that both proliferating and nonproliferating FOXP3+ T cells exhibited complete demethylation of the FOXP3 gene, indicating a stable FOXP3 expression in both cell populations. Furthermore, analysis of T-cell populations isolated directly from the tumor and tumor-free mucosa demonstrated that CD4+ CD25high T cells have a higher IL-10/IFN-γ gene expression ratio but express lower levels of TGF-ß than CD4+ CD25low/- T cells. CONCLUSION: We demonstrate strong proliferation among regulatory CD4+ FOXP3+ CD25high T cells in the gastric cancer mucosa. These local Treg express a suppressive cytokine profile characterized by high IL-10 and low TGF-ß and IFN-γ production.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Mucosa Gástrica/imunologia , Interleucina-10/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/genética , Idoso , Idoso de 80 Anos ou mais , Apoptose , Biomarcadores Tumorais/genética , Metilação de DNA , Feminino , Citometria de Fluxo , Seguimentos , Fatores de Transcrição Forkhead/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/sangue , Neoplasias Gástricas/genética , Células Tumorais Cultivadas
10.
PLoS One ; 7(2): e30695, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22319577

RESUMO

BACKGROUND: Colorectal cancer usually gives rise to a specific anti-tumor immune response, but for unknown reasons the resulting immunity is not able to clear the tumor. Recruitment of activated effector lymphocytes to the tumor is important for efficient anti-tumor responses, while the presence of regulatory T cells (Treg) down-modulate tumor-specific immunity. We therefore aimed to determine homing mechanisms and activation stage of Treg and effector T cell infiltrating colon tumors compared to cells from the unaffected mucosa in patients suffering from colon adenocarcinoma. METHODOLOGY/PRINCIPAL FINDINGS: Lymphocytes were isolated from unaffected and tumor mucosa from patients with colon adenocarcinoma, and flow cytometry, immunohistochemistry, and quantitative PCR was used to investigate the homing mechanisms and activation stage of infiltrating Treg and conventional lymphocytes. We detected significantly higher frequencies of CD25(high)FOXP3⁺CD127(low) putative Treg in tumors than unaffected mucosa, which had a complete demethylation in the FOXP3 promotor. Tumor-associated Treg had a high expression of CTLA-4, and some appeared to be antigen experienced effector/memory cells based on their expression of αEß7 (CD103). There were also significantly fewer activated T cells and more CTLA-4⁺ conventional T cells susceptible to immune regulation in the tumor-associated mucosa. In contrast, CD8⁺granzyme B⁺ putative cytotoxic cells were efficiently recruited to the tumors. The frequencies of cells expressing α4ß7 and the Th1 associated chemokine receptor CXCR3 were significantly decreased among CD4⁺ T cells in the tumor, while frequencies of CD4⁺CCR4⁺ lymphocytes were significantly increased. CONCLUSIONS/SIGNIFICANCE: This study shows that CCR4⁺CTLA4(hi) Treg accumulate in colon tumors, while the frequencies of activated conventional Th1 type T cells are decreased. The altered lymphocyte composition in colon tumors will probably diminish the ability of the immune system to effectively attack tumor cells, and reducing the Treg activity is an important challenge for future immunotherapy protocols.


Assuntos
Adenocarcinoma Mucinoso/imunologia , Neoplasias do Colo/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/imunologia , Adenocarcinoma/imunologia , Antígeno CTLA-4 , Humanos , Imunidade , Subunidade alfa de Receptor de Interleucina-2 , Mucosa , Receptores CCR4 , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA