Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 161: 105094, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32795509

RESUMO

Oncolytic virus (OV) immunotherapy has demonstrated to be a promising approach in cancer treatment due to tumor-specific oncolysis. However, their clinical use so far has been largely limited due to the lack of suitable delivery strategies with high efficacy. Direct 'intratumoral' injection is the way to cross the hurdles of systemic toxicity, while providing local effects. Progress in this field has enabled the development of alternative way using 'systemic' oncolytic virotherapy for producing better results. One major potential roadblock to systemic OV delivery is the low virus persistence in the face of hostile immune system. The delivery challenge is even greater when attempting to target the oncolytic viruses into the entire tumor mass, where not all tumor cells are equally exposed to exactly the same microenvironment. The microenvironment of many tumors is known to be massively infiltrated with various types of leucocytes in both primary and metastatic sites. Interestingly, this intratumoral immune cell heterogeneity exhibits a degree of organized distribution inside the tumor bed as evidenced, for example, by the hypoxic tumor microenviroment where predominantly recruits tumor-associated macrophages. Although in vivo OV delivery seems complicated and challenging, recent results are encouraging for decreasing the limitations of systemically administered oncolytic viruses and an improved efficiency of oncolytic viral therapy in targeting cancerous tissues in vitro. Here, we review the latest developments of carrier cell-based oncolytic virus delivery using tumor-infiltrating immune cells with a focus on the main features of each cellular vehicle.


Assuntos
Fibroblastos Associados a Câncer/virologia , Células Matadoras Induzidas por Citocinas/virologia , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/virologia , Monócitos/virologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Linfócitos T/virologia , Animais , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/transplante , Células Matadoras Induzidas por Citocinas/imunologia , Células Matadoras Induzidas por Citocinas/transplante , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/transplante , Monócitos/imunologia , Monócitos/transplante , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/virologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/virologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Fenótipo , Linfócitos T/imunologia , Linfócitos T/transplante , Hipóxia Tumoral , Microambiente Tumoral
2.
Cell Mol Biol (Noisy-le-grand) ; 66(2): 36-40, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32415924

RESUMO

Hepatocellular carcinoma is known to be a common predominant cancer in adults, especially in eastern countries. Immune response and cancer-associated fibroblasts (CAFs) have significant influences on tumor development. However, the interaction between CAFs and immunotherapy is unclear in hepatocellular carcinoma. We measured the number of activated fibroblasts in hepatocellular carcinoma samples and samples taken from normal liver tissues. A total of 20 patients' fresh hepatocellular carcinoma and normal tissues which were surrounding the tumor were obtained from the surgery and used for evaluating alpha-SMA expression. We investigated the effects of CAFs in anti-tumor immunity in hepatocellular carcinoma animal model. The effects of CAFs in inducing anti-PD-1 treatment resistance were also measured in a preclinical animal model. Activated fibroblasts were highly accumulated in hepatocellular carcinoma tissues but not in surrounding normal tissues. CAFs showed a significant tumor-promoting effect in an immunocompetent model. The infiltration and function of some immune cells like myeloid-derived suppressive cells and T-cells were increased by CAFs. CAFs also reduced the number and activation of tumor-infiltrating cytotoxic T-cell in tumor tissue. In the treatment model, tumors with a higher amount of CAFs had been insensitive to therapy with anti-PD-1. CAFs are potent inducers of immunosuppression in hepatocellular carcinoma. Depleting CAFs rescued the antitumor immunity in the hepatocellular model and could be a novel treatment to combine with the existing immunotherapy.


Assuntos
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Actinas/metabolismo , Animais , Antineoplásicos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Fibroblastos Associados a Câncer/citologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/transplante , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Dasatinibe/uso terapêutico , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Imunoterapia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Células Supressoras Mieloides/citologia , Células Supressoras Mieloides/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
3.
Cancer Sci ; 110(5): 1790-1803, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30843645

RESUMO

Heat shock factor 1 (HSF1) is highly expressed in various malignancies and is a potential modulator of tumor progression. Emerging evidence suggests that HSF1 activation in stromal cells is closely related to poor patient prognosis. However, the role of HSF1 in oral squamous cell carcinoma (OSCC) remains elusive. We aimed to investigate the function of HSF1 in cancer-associated fibroblasts (CAFs) of the tumor microenvironment (TME) and in tumor development. In the present study, we found that HSF1 was highly expressed in both CAFs and tumor cells, and was significantly correlated with poor prognosis and overall survival. Moreover, HSF1 overexpression in CAFs resulted in a fibroblast-like phenotype of Cal27 cells, induced epithelial-mesenchymal transition (EMT), and promoted proliferation, migration and invasion in Cal27 cells. HSF1 knockdown attenuated features of CAFs and reduced EMT, proliferation, migration and invasion in Cal27 cells. Furthermore, HSF1 in CAFs promoted tumor growth in nude mice. Taken together, these data suggest that HSF1 expression in CAFs drive OSCC progression, and could serve as an independent prognostic marker of patients with OSCC. Thus, HSF1 is a potent mediator of OSCC malignancy.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Carcinoma de Células Escamosas/patologia , Fatores de Transcrição de Choque Térmico/metabolismo , Neoplasias Bucais/patologia , Regulação para Cima , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Fibroblastos Associados a Câncer/transplante , Carcinoma de Células Escamosas/metabolismo , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Bucais/metabolismo , Estadiamento de Neoplasias , Transplante de Neoplasias , Prognóstico , Análise de Sobrevida , Microambiente Tumoral
4.
Clin Cancer Res ; 23(16): 4769-4779, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28420724

RESUMO

Purpose: Cancer metastasis can occur at the early stage of tumor development when a primary tumor is at the microscopic size. In particular, the interaction of malignant cells with other cell types including cancer-associated fibroblasts (CAF) in promoting metastasis at the early stage of tumor development remains largely unknown. Here, we investigated the role of CAFs in facilitating the initial events of cancer metastasis when primary tumors were at microscopic sizes.Experimental Design: Multicolor-coded cancer cells and CAFs were coimplanted into the transparent zebrafish body and metastasis at a single-cell level was monitored in living animals. Healthy fibroblasts, tumor factor-educated fibroblasts, and CAFs isolated from various tumors were tested for their ability to facilitate metastasis.Results: We showed that CAFs promoted cancer cell metastasis at the very early stage during primary tumor development. When a primary tumor was at the microscopic size consisting of a few hundred cells, CAFs were able to hijack cancer cells for dissemination from the primary site. Surprisingly, a majority of metastatic cancer cells remained in tight association with CAFs in the circulation. Furthermore, stimulation of non-metastasis-promoting normal fibroblasts with TGF-B, FGF-2, HGF, and PDGF-BB led to acquisition of their metastatic capacity.Conclusions: Cancer metastasis occurs at the very early stage of tumor formation consisting of only a few hundred cells. CAFs are the key cellular determinant for metastasis. Our findings provide novel mechanistic insights on CAFs in promoting cancer metastasis and targeting CAFs for cancer therapy should be aimed at the early stage during cancer development. Clin Cancer Res; 23(16); 4769-79. ©2017 AACR.


Assuntos
Fibroblastos Associados a Câncer/patologia , Embrião não Mamífero/patologia , Neoplasias/patologia , Peixe-Zebra/embriologia , Células A549 , Animais , Animais Geneticamente Modificados , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/transplante , Linhagem Celular Tumoral , Movimento Celular/genética , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Transplante Heterólogo , Peixe-Zebra/genética
5.
Cancer Lett ; 385: 39-45, 2017 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-27826041

RESUMO

Tumor-associated-fibroblasts (TAFs) are the most important host cells in the stroma and take part in extracellular matrix construction and cancer colony development. During cancer colonization, seed cells from primary tumor can reconstruct the microenvironment by recruiting circulating cancer cells and TAFs to the metastasis site. Previous studies have established that SMC1A, a subunit of cohesin, is an important trigger signal for liver metastasis in colorectal cancer. We investigated the particular effects as well as the underlying mechanism of SMC1A on TAFs recruitment during liver metastasis of colorectal cancer. Here, We found that: first, the high expression of SMC1A in colorectal cancer cells promotes the invasiveness and the viability of these cells by recruiting circulating TAFs, facilitating early tumor construction and tumorigenesis; second, different expression levels of SMC1A influenced the reformation of fibroblasts, which assisted tumorigenesis, and third, expression of SMC1A stimulated the secretion of the inflammatory mediators of TNF-α and IL-1ß, and up-regulated the transcriptional expression of MMP2 and VEGF-ß, both of which were involved in the tumor-related gene pathway.


Assuntos
Adenocarcinoma/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quimiotaxia , Proteínas Cromossômicas não Histona/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Hepáticas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/secundário , Animais , Fibroblastos Associados a Câncer/patologia , Fibroblastos Associados a Câncer/transplante , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Humanos , Interleucina-1beta/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Transdução de Sinais , Células Tumorais Cultivadas , Microambiente Tumoral , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Fator B de Crescimento do Endotélio Vascular/genética , Fator B de Crescimento do Endotélio Vascular/metabolismo
6.
Cancer Lett ; 383(1): 18-27, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27693460

RESUMO

Vascular mimicry (VM) describes the phenomenon that tumor cells but not endothelial cells form vascular-like channels, which provide blood perfusion for tumor tissues. VM is associated with tumor growth, metastasis and worse survival of different cancers. The mechanisms of VM formation remain largely unknown. We showed that the conditioned medium of cancer-associated fibroblast (CM-CAF) promoted tumor cells to form capillary-like structure in vitro. Consistently, co-implantation of CAFs with tumor cells significantly enhanced VM formation in mouse xenografts, and higher amount of CAFs was found in VM+ human HCC tissues compared to VM- ones. However, the CM-CAF-promoted VM formation was attenuated when TGF-ß or SDF1 signaling was abrogated. Similar to CM-CAF, recombinant TGF-ß1 and SDF1 induced VM formation. We further disclosed that the CAF-secreted TGF-ß and SDF1 enhanced the expression of VE-cadherin, MMP2 and laminin5γ2 via TGF-ßR1 and CXCR4 in tumor cells, thereby promoted VM formation. Moreover, tumor cells with high activity of self-sustaining TGF-ß signaling displayed strong capability of VM formation. Subsequent investigations showed that miR-101, which was down-regulated in both tumor cells and CAFs, suppressed the CAF-promoted VM formation in vitro and in vivo. Gain- and loss-of-function analyses revealed that miR-101 attenuated TGF-ß signaling transduction by targeting TGF-ßR1 and Smad2 in tumor cells, and simultaneously abrogated SDF1 signaling by suppressing SDF1 expression in CAFs and inhibiting VE-cadherin expression in tumor cells. Our findings suggest that the miR-101-TGF-ß/SDF1-VE-cadherin/MMP2/LAMC2 networks regulate VM formation and represent the potential targets for cancer therapy.


Assuntos
Mimetismo Biológico , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Hepatocelular/metabolismo , Quimiocina CXCL12/metabolismo , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo , Neovascularização Patológica , Comunicação Parácrina , Fator de Crescimento Transformador beta1/metabolismo , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Fibroblastos Associados a Câncer/patologia , Fibroblastos Associados a Câncer/transplante , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/metabolismo , Feminino , Humanos , Laminina/metabolismo , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
7.
Sci Rep ; 6: 21782, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26899873

RESUMO

Cancer-associated fibroblasts (CAFs), the principal components of the tumor stroma, play a central role in cancer development and progression. As an important regulator of the crosstalk between breast cancer cells and CAFs, the cytokine leptin has been associated to breast carcinogenesis. The nuclear Farnesoid X Receptor-(FXR) seems to exert an oncosuppressive role in different tumors, including breast cancer. Herein, we demonstrated, for the first time, that the synthetic FXR agonist GW4064, inhibiting leptin signaling, affects the tumor-promoting activities of CAFs in breast malignancy. GW4064 inhibited growth, motility and invasiveness induced by leptin as well as by CAF-conditioned media in different breast cancer cell lines. These effects rely on the ability of activated FXR to increase the expression of the suppressor of the cytokine signaling 3 (SOCS3) leading to inhibition of leptin-activated signaling and downregulation of leptin-target genes. In vivo xenograft studies, using MCF-7 cells alone or co-injected with CAFs, showed that GW4064 administration markedly reduced tumor growth. Interestingly, GW4064-treated tumors exhibited decreased levels of leptin-regulated proteins along with a strong staining intensity for SOCS3. Thus, FXR ligands might represent an emerging potential anti-cancer therapy able to block the tumor supportive role of activated fibroblasts within the breast microenvironment.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Fibroblastos Associados a Câncer/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Isoxazóis/farmacologia , Receptores Citoplasmáticos e Nucleares/agonistas , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Fibroblastos Associados a Câncer/transplante , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Comunicação Celular , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Leptina/genética , Leptina/metabolismo , Células MCF-7 , Camundongos Nus , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...