Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cancer Immunol Immunother ; 70(11): 3155-3166, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33786638

RESUMO

There is an urgent need for new treatment options in metastatic drug-resistant prostate cancer. Combining immunotherapy with other targeted therapies may be an effective strategy for advanced prostate cancer. In the present study, we sought to investigate to enhance the efficacy of anti-CTLA-4 therapy against prostate cancer by the combination with STAT3 inhibition.Male C57BL6 mice were subcutaneously inoculated with the murine prostate cancer cell line RM-1. Tumor progression was monitored following treatment with vehicle, the small molecule STAT3 inhibitor GPB730, anti-CTLA-4 or GPB730 + anti-CTLA-4. Treatment with anti-CTLA-4 or anti-CTLA-4 + GPB730 significantly inhibited tumor growth and enhanced survival compared to vehicle. Combining anti-CTLA-4 treatment with GPB730 resulted in a significantly prolonged survival compared to anti-CTLA-4 alone. GPB730 significantly increased infiltration of CD45 + cells in tumors of anti-CTLA-4-treated mice compared to anti-CTLA-4 alone. The levels of tumor-infiltrating Tregs were significantly decreased and the CD8:Treg ratio significantly increased by GPB730 treatment in combination with anti-CTLA-4 compared to anti-CTLA-4 alone. Immunohistochemical analysis showed a significant increase in CD45-positive cells in anti-CTLA-4 and anti-CTLA-4 + GPB730-treated tumors compared to vehicle or GPB730 monotherapy. Plasma levels of IL10 were significantly increased by anti-CTLA-4 compared to vehicle but no increase was observed when combining anti-CTLA-4 with GPB730.In conclusion, STAT3 inhibition by GPB730 enhances the antitumoral activity of anti-CTLA-4 and decreases the intratumoral Treg frequency in a prostate cancer mouse model. These results support the combination of STAT3 inhibition with anti-CTLA-4 therapy to increase clinical responses in patients with prostate cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias de Próstata Resistentes à Castração/patologia , Fator de Transcrição STAT3/antagonistas & inibidores , Animais , Lactonas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Blood ; 128(11): 1475-89, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27465917

RESUMO

Treatment of hematological malignancies by adoptive transfer of activated natural killer (NK) cells is limited by poor postinfusion persistence. We compared the ability of interleukin-2 (IL-2) and IL-15 to sustain human NK-cell functions following cytokine withdrawal to model postinfusion performance. In contrast to IL-2, IL-15 mediated stronger signaling through the IL-2/15 receptor complex and provided cell function advantages. Genome-wide analysis of cytosolic and polysome-associated messenger RNA (mRNA) revealed not only cytokine-dependent differential mRNA levels and translation during cytokine activation but also that most gene expression differences were primed by IL-15 and only manifested after cytokine withdrawal. IL-15 augmented mammalian target of rapamycin (mTOR) signaling, which correlated with increased expression of genes related to cell metabolism and respiration. Consistently, mTOR inhibition abrogated IL-15-induced cell function advantages. Moreover, mTOR-independent STAT-5 signaling contributed to improved NK-cell function during cytokine activation but not following cytokine withdrawal. The superior performance of IL-15-stimulated NK cells was also observed using a clinically applicable protocol for NK-cell expansion in vitro and in vivo. Finally, expression of IL-15 correlated with cytolytic immune functions in patients with B-cell lymphoma and favorable clinical outcome. These findings highlight the importance of mTOR-regulated metabolic processes for immune cell functions and argue for implementation of IL-15 in adoptive NK-cell cancer therapy.


Assuntos
Citotoxicidade Imunológica/imunologia , Imunoterapia Adotiva , Interleucina-15/farmacologia , Células Matadoras Naturais/imunologia , Neoplasias Experimentais/terapia , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Citocinas/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Mitocondriais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Transdução de Sinais
3.
J Immunol ; 196(2): 759-66, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26673145

RESUMO

Treatment of cancer patients by adoptive T cell therapy has yielded promising results. In solid tumors, however, T cells encounter a hostile environment, in particular with increased inflammatory activity as a hallmark of the tumor milieu that goes along with abundant reactive oxygen species (ROS) that substantially impair antitumor activity. We present a strategy to render antitumor T cells more resilient toward ROS by coexpressing catalase along with a tumor specific chimeric Ag receptor (CAR) to increase their antioxidative capacity by metabolizing H2O2. In fact, T cells engineered with a bicistronic vector that concurrently expresses catalase, along with the CAR coexpressing catalase (CAR-CAT), performed superior over CAR T cells as they showed increased levels of intracellular catalase and had a reduced oxidative state with less ROS accumulation in both the basal state and upon activation while maintaining their antitumor activity despite high H2O2 levels. Moreover, CAR-CAT T cells exerted a substantial bystander protection of nontransfected immune effector cells as measured by CD3ζ chain expression in bystander T cells even in the presence of high H2O2 concentrations. Bystander NK cells, otherwise ROS sensitive, efficiently eliminate their K562 target cells under H2O2-induced oxidative stress when admixed with CAR-CAT T cells. This approach represents a novel means for protecting tumor-infiltrating cells from tumor-associated oxidative stress-mediated repression.


Assuntos
Catalase/imunologia , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Estresse Oxidativo/fisiologia , Linfócitos T/imunologia , Western Blotting , Efeito Espectador/imunologia , Linhagem Celular , Separação Celular , Humanos , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/imunologia , Transfecção
4.
Front Immunol ; 11: 621225, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584718

RESUMO

Natural killer (NK) cells are endowed with germline-encoded receptors that enable them to detect and kill malignant cells without prior priming. Over the years, overwhelming evidence has identified an essential role for NK cells in tumor immune surveillance. More recently, clinical trials have also highlighted their potential in therapeutic settings. Yet, data show that NK cells can be dysregulated within the tumor microenvironment (TME), rendering them ineffective in eradicating the cancer cells. This has been attributed to immune suppressive factors, including the tumor cells per se, stromal cells, regulatory T cells, and soluble factors such as reactive oxygen species and cytokines. However, the TME also hosts myeloid cells such as dendritic cells, macrophages, neutrophils, and myeloid-derived suppressor cells that influence NK cell function. Although the NK-myeloid cell crosstalk can promote anti-tumor responses, myeloid cells in the TME often dysregulate NK cells via direct cell-to-cell interactions down-regulating key NK cell receptors, depletion of nutrients and growth factors required for NK cell growth, and secretion of metabolites, chemokines and cytokines that ultimately alter NK cell trafficking, survival, and cytotoxicity. Here, we review the complex functions of myeloid-derived cytokines in both supporting and suppressing NK cells in the TME and how NK cell-derived cytokines can influence myeloid subsets. We discuss challenges related to these interactions in unleashing the full potential of endogenous and adoptively infused NK cells. Finally, we present strategies aiming at improving NK cell-based cancer immunotherapies via pathways that are involved in the NK-myeloid cell crosstalk in the TME.


Assuntos
Comunicação Celular/imunologia , Citocinas/imunologia , Imunoterapia , Células Matadoras Naturais , Células Mieloides , Neoplasias , Microambiente Tumoral/imunologia , Animais , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células Matadoras Naturais/transplante , Células Mieloides/imunologia , Células Mieloides/patologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
5.
Methods Mol Biol ; 1913: 197-206, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30666608

RESUMO

Syngeneic mouse models enable us to study the interaction of the immune system with tumor cells during tumor development and metastasis. It further allows us to evaluate the efficacy of immunotherapies on the different stages of tumor growth and the metastatic process. Here we describe two syngeneic mouse models, a murine mammary carcinoma and a murine melanoma model, that are used to study metastasis. Metastases occur spontaneously in the murine mammary carcinoma. The presence and number of foci are evaluated by culturing the lung tissue in a colony formation assay. For the murine melanoma model, tumor cells are injected intravenously, and metastatic burden is analyzed by counting of metastatic lesions.


Assuntos
Carcinoma/patologia , Linhagem Celular Tumoral/transplante , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/patologia , Animais , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Feminino , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C
6.
Methods Mol Biol ; 2032: 105-114, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31522415

RESUMO

Therapies based on activating the immune system, that is, immunotherapy, are now widely implemented in clinical praxis in patients with advanced cancer. Although cancer immunotherapy can result in long-lasting clinical responses, the majority of patients do not respond or develop resistance. Furthermore, cancer immunotherapy is being increasingly combined with other forms of immunotherapy or conventional cancer therapies. It is therefore much needed to identify biomarkers that can precisely classify what patients will benefit from the treatment without any major adverse events and to further develop the efficacy of cancer immunotherapy. While much attention has been focused on monitoring T cell responses in cancer immunotherapy, recent reports have shown that NK cells also play a major role in the response to cancer immunotherapy. The gold standard for immunoprofiling of NK cells is flow cytometry, but other technologies have emerged and include mass cytometry, multiplex immunohistochemistry, and single-cell RNA-sequencing. In this chapter we provide a detailed protocol to profile NK cells using flow cytometry, and a brief introduction to other techniques.


Assuntos
Citometria de Fluxo/métodos , Imunofenotipagem/métodos , Células Matadoras Naturais/imunologia , Humanos , Imuno-Histoquímica/métodos , Neoplasias/diagnóstico , Neoplasias/imunologia , Análise de Sequência de RNA , Análise de Célula Única/métodos
7.
Cancer Immunol Res ; 6(11): 1417-1425, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30143536

RESUMO

Metastatic breast cancer is a fatal disease that responds poorly to treatment. Cancer vaccines targeting antigens expressed by metastatic breast cancer cells and cancer stem cells could function as anticancer therapies. Cripto-1 is an oncofetal protein overexpressed in invasive breast cancer and cancer-initiating cells. In this study, we explored the potential of a Cripto-1-encoding DNA vaccine to target breast cancer in preclinical mouse models. BALB/c mice and BALB-neuT mice were treated with a DNA vaccine encoding mouse Cripto-1 (mCr-1). BALB/c mice were challenged with murine breast cancer 4T1 cells or TUBO spheres; BALB-neuT mice spontaneously developed breast cancer. Tumor growth was followed in all mouse models and lung metastases were evaluated. In vitro assays were performed to identify the immune response elicited by vaccination. Vaccination against mCr-1 reduced primary tumor growth in the 4T1 metastatic breast cancer model and reduced lung metastatic burden. In BALB-neuT mice, because the primary tumors are Cripto-1 negative, vaccination against mCr-1 did not affect primary tumors but did reduce lung metastatic burden. Spheroid-cultured TUBO cells, derived from a BALB/neuT primary tumor, develop a cancer stem cell-like phenotype and express mCr-1. We observed reduced tumor growth in vaccinated mice after challenge with TUBO spheres. Our data indicate that vaccination against Cripto-1 results in a protective immune response against mCr-1 expressing and metastasizing cells. Targeting Cripto-1 by vaccination holds promise as an immunotherapy for treatment of metastatic breast cancer. Cancer Immunol Res; 6(11); 1417-25. ©2018 AACR.


Assuntos
Vacinas Anticâncer/farmacologia , Fator de Crescimento Epidérmico/genética , Neoplasias Mamárias Experimentais/terapia , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Vacinas de DNA/farmacologia , Animais , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/imunologia , Feminino , Imunidade Humoral , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Glicoproteínas de Membrana/imunologia , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/imunologia , Células-Tronco Neoplásicas/imunologia
8.
J Biomed Mater Res B Appl Biomater ; 82(2): 494-505, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17279565

RESUMO

The nanocrystalline hydroxyapatite paste Ostim represents a fully degradable synthetic bone substitute for the filling of bone defects. Herein, we investigated in vivo the inflammatory and angiogenic host tissue response to this biomaterial after implantation. For this purpose, Ostim was implanted into the dorsal skinfold chambers of Syrian golden hamsters. The hydroxyapatite ceramic Cerabone and isogeneic transplanted cancellous bone served as controls. Angiogenesis, microhemodynamics, microvascular permeability, and leukocyte-endothelial cell interaction of the host tissue were analyzed over 2 weeks using intravital fluorescence microscopy. Ostim exhibited good biocompatibility comparable to that of Cerabone and cancellous bone, as indicated by a lack of venular leukocyte activation after implantation. Cancellous bone induced a more pronounced angiogenic response and an increased microvessel density when compared with the synthetic bone substitutes. In contrast to Cerabone, however, Ostim showed a guided neovascularization directed toward areas of degradation. Histology confirmed the ingrowth of proliferating vascularized tissue into the hydroxyapatite paste at sites of degradation, while the hydroxyapatite ceramic was not pierced by new microvessels. Thus, Ostim represents an injectable synthetic bone substitute, which may optimize the conditions for the formation of new bone at sites of bone defects by supporting a guided vascularization during biodegradation.


Assuntos
Cimentos Ósseos/farmacologia , Regeneração Óssea , Substitutos Ósseos/farmacologia , Osso e Ossos/efeitos dos fármacos , Durapatita/farmacologia , Neovascularização Fisiológica , Animais , Cimentos Ósseos/efeitos adversos , Substitutos Ósseos/administração & dosagem , Substitutos Ósseos/efeitos adversos , Osso e Ossos/irrigação sanguínea , Osso e Ossos/ultraestrutura , Cricetinae , Durapatita/administração & dosagem , Durapatita/química , Inflamação/induzido quimicamente , Teste de Materiais , Mesocricetus , Microcirculação , Nanopartículas/química
9.
Oncoimmunology ; 6(8): e1338238, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28920001

RESUMO

Regulatory T cells (Treg) suppress anti-tumor immune responses and their infiltration in the tumor microenvironment is associated with inferior prognosis in cancer patients. Thus, in order to enhance anti-tumor immune responses, selective depletion of Treg is highly desired. We found that treatment with zoledronic acid (ZA) resulted in a selective decrease in the frequency of Treg that was associated with a significant increase in proliferation of T cells and natural killer (NK) cells in peripheral blood of patients with metastatic cancer. In vitro, genome-wide transcriptomic analysis revealed alterations in calcium signaling pathways in Treg following treatment with ZA. Furthermore, co-localization of the nuclear factor of activated T cells (NFAT) and forkhead box P3 (FOXP3) was significantly reduced in Treg upon ZA-treatment. Consequently, reduced expression levels of CD25, STAT5 and TGFß were observed. Functionally, ZA-treated Treg had reduced capacity to suppress T and NK cell proliferation and anti-tumor responses compared with untreated Treg in vitro. Treatment with ZA to selectively inhibit essential signaling pathways in Treg resulting in reduced capacity to suppress effector T and NK cell responses represents a novel approach to inhibit Treg activity in patients with cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA