Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Cancer Prev Res (Phila) ; 14(12): 1075-1088, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34535449

RESUMO

Aging is a complex physiological process that leads to the progressive decline of metabolic and immune function, among other biological mechanisms. As global life expectancy increases, it is important to understand determinants of healthy aging-including environmental and genetic factors-and thus slow the onset or progression of age-related disease. Environmental enrichment (EE) is a housing environment wherein laboratory animals engage with complex physical and social stimulation. EE is a prime model to understand environmental influences on aging dynamics, as it confers an antiobesity and anticancer phenotype that has been implicated in healthy aging and health span extension. Although EE is frequently used to study malignancies in young mice, fewer studies characterize EE-cancer outcomes in older mice. Here, we used young (3-month-old) and aged (14-month-old) female C57BL/6 mice to determine whether EE would be able to mitigate age-related deficiencies in metabolic function and thus alter Lewis lung carcinoma (LLC) growth. Overall, EE improved metabolic function, resulting in reduced fat mass, increased lean mass, and improved glycemic processing; many of these effects were stronger in the aged cohort than in the young cohort, indicating an age-driven effect on metabolic responses. In the aged-EE cohort, subcutaneously implanted LLC tumor growth was inhibited and tumors exhibited alterations in various markers of apoptosis, proliferation, angiogenesis, inflammation, and malignancy. These results validate EE as an anticancer model in aged mice and underscore the importance of understanding environmental influences on cancer malignancy in aged populations. PREVENTION RELEVANCE: Environmental enrichment (EE) serves as a model of complex physical and social stimulation. This study validates EE as an anticancer intervention paradigm in aged mice and underscores the importance of understanding environmental influences on cancer malignancy in aged populations.


Assuntos
Carcinoma Pulmonar de Lewis , Envelhecimento , Animais , Carcinoma Pulmonar de Lewis/prevenção & controle , Meio Ambiente , Feminino , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
2.
Neurosci Lett ; 754: 135851, 2021 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-33781910

RESUMO

Psychological stress is a common etiology among patients with lung cancer and serves as a potential indication of poor prognosis and advanced cancer clinical stage. Evidence indicates that depression is positively correlated with the evolvement of lung carcinoma. Nevertheless, the mechanisms underlying the effects of mental disorder on lung cancer have not been considerably and systemically explored. We hypothesized that mental disorder may affect the adjustment of the tumor microenvironment and immune cells. We used the chronic unpredictable mild stress (CUMS) procedure to induce depressed mice models and established tumor-bearing models of C57BL/6 J mice. Results revealed that the worsening of lung cancer was notably hastened in the CUMS + tumor group. Notably, the expression of PD-L1 in tumor issues increased in the tumor microenvironment, accompanied with a decline in the levels of CD8. On the basis of the date of tumor migration, our results indicated that MMPs and VEGF significantly increased after CUMS + tumor treatment. Thus, we demonstrated that modulation of the tumor microenvironment is pivotal for depression-promoted lung cancer migration.


Assuntos
Carcinoma Pulmonar de Lewis/secundário , Depressão/complicações , Neoplasias Pulmonares/patologia , Estresse Psicológico/complicações , Microambiente Tumoral/imunologia , Animais , Antidepressivos/administração & dosagem , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Carcinoma Pulmonar de Lewis/psicologia , Linhagem Celular Tumoral , Depressão/tratamento farmacológico , Depressão/imunologia , Depressão/psicologia , Progressão da Doença , Humanos , Pulmão/imunologia , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/imunologia , Estresse Psicológico/psicologia , Linfócitos T Citotóxicos , Microambiente Tumoral/efeitos dos fármacos
3.
J Recept Signal Transduct Res ; 41(5): 504-510, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32998602

RESUMO

BACKGROUND: Cell division control (CDC) 42 has been involved in the regulation of diverse cancers. Macrophage recruitment plays an important role in the pathogenesis and development of tumor. However, it remains unclear whether CDC42 contributes to macrophage recruitment and lung tumorigenesis in vivo. METHODS: Small interference RNA (siRNA) was used to knock down CDC42 in the Lewis lung carcinoma (LLC)1. The invasion capability of CDC42 knockdown LLC1 cells was evaluated. LLC1 cells with CDC42 targeted small hairpin RNA (shRNA) were inoculated into C57BL/6 mice to establish the tumor-bearing animal model Tumor size and metastasis related proteins were measured. In addition, the invasion of macrophages in the tumor site as well as macrophage chemokine were also determined in the model. RESULTS: The capacity of invasion and metastasis of LLC1 cells significantly decreased when CDC42 was knocked down. When inoculated with CDC42 knockdown LLC1 cells in vivo, the tumor size and metastasis related proteins levels both decreased. The invasion capacity of macrophages and the associated macrophage chemokine were also significantly down-regulated. CONCLUSION: Our data suggest that the inhibition of CDC42 expression in lung cancer cells can significantly prevent the pathogenesis and development of tumor in an allograft tumor model in vivo, which might provide a novel therapeutic target and potential strategy for lung cancer treatment in the future.


Assuntos
Carcinogênese/patologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Modelos Animais de Doenças , Macrófagos/imunologia , RNA Interferente Pequeno/genética , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Animais , Apoptose , Carcinogênese/imunologia , Carcinogênese/metabolismo , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína cdc42 de Ligação ao GTP/genética
4.
JCI Insight ; 5(19)2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33004686

RESUMO

Engineering T cells to express chimeric antigen receptors (CARs) specific for antigens on hematological cancers has yielded remarkable clinical responses, but with solid tumors, benefit has been more limited. This may reflect lack of suitable target antigens, immune evasion mechanisms in malignant cells, and/or lack of T cell infiltration into tumors. An alternative approach, to circumvent these problems, is targeting the tumor vasculature rather than the malignant cells directly. CLEC14A is a glycoprotein selectively overexpressed on the vasculature of many solid human cancers and is, therefore, of considerable interest as a target antigen. Here, we generated CARs from 2 CLEC14A-specific antibodies and expressed them in T cells. In vitro studies demonstrated that, when exposed to their target antigen, these engineered T cells proliferate, release IFN-γ, and mediate cytotoxicity. Infusing CAR engineered T cells into healthy mice showed no signs of toxicity, yet these T cells targeted tumor tissue and significantly inhibited tumor growth in 3 mouse models of cancer (Rip-Tag2, mPDAC, and Lewis lung carcinoma). Reduced tumor burden also correlated with significant loss of CLEC14A expression and reduced vascular density within malignant tissues. These data suggest the tumor vasculature can be safely and effectively targeted with CLEC14A-specific CAR T cells, offering a potent and widely applicable therapy for cancer.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Carcinoma Ductal Pancreático/prevenção & controle , Moléculas de Adesão Celular/metabolismo , Imunoterapia Adotiva/métodos , Lectinas Tipo C/metabolismo , Neovascularização Patológica/prevenção & controle , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Moléculas de Adesão Celular/genética , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Lectinas Tipo C/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/prevenção & controle
5.
Anticancer Res ; 39(4): 1729-1738, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30952712

RESUMO

AIM: Monocyte chemotactic protein-1 (MCP1) is a potent adipokine. This study tested the hypothesis that adipose-produced MCP1 contributes to metastasis. MATERIALS AND METHODS: In a spontaneous metastasis model of Lewis lung carcinoma (LLC), male adipose MCP1-deficient (Mcp1-/-) and wild-type (WT) mice were fed the AIN93G diet or a high-fat diet (HFD) for 11 weeks. Lung metastasis from a subcutaneous tumor was the primary endpoint. RESULTS: The adipose expression of MCP1 was lower in Mcp1-/- mice than in WT controls. The HFD increased the number of lung metastases in WT mice. The number of metastasis was significantly lower in the HFD-fed Mcp1-/- mice than in the HFD-fed WT mice. Compared to the WT mice, adipose MCP1 deficiency lowered plasma concentrations of insulin, proinflammatory adipokines (leptin, plasminogen activator inhibitor-1, and resistin), and angiogenic markers (vascular endothelial growth factor, hepatocyte growth factor, and angiopoietin-2). CONCLUSION: Adipose MCP1 deficiency attenuates HFD-enhanced pulmonary metastasis of LLC.


Assuntos
Tecido Adiposo/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundário , Quimiocina CCL2/deficiência , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Adipocinas/metabolismo , Adiposidade , Proteínas Angiogênicas/metabolismo , Animais , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/prevenção & controle , Quimiocina CCL2/genética , Dieta Hiperlipídica , Mediadores da Inflamação/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , Transdução de Sinais
6.
Anticancer Res ; 39(4): 1739-1748, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30952713

RESUMO

BACKGROUND/AIM: Obesity is a risk factor for cancer. Disruption of the daily feeding and fasting rhythm can contribute to obesity. This study tested the hypothesis that time-restricted feeding (TRF) attenuates obesity-enhanced metastasis. MATERIALS AND METHODS: In a spontaneous metastasis model of Lewis lung carcinoma (LLC), male C57BL/6 mice were fed the standard AIN93G diet or a high-fat diet (HFD) with or without dark-phase restricted feeding (12 h per day) for 10 weeks. Pulmonary metastases from a subcutaneous tumor were quantified. RESULTS: The number and size of lung metastases were greater in the HFD group than in the AIN93G group, but did not differ between the TRF and AIN93G groups. TRF prevented HFD-induced increases in plasma concentrations of glucose, insulin, proinflammatory cytokines (leptin, monocyte chemotactic protein-1, plasminogen activator inhibitor-1), and angiogenic factors (angiopoietin-2, hepatic growth factor, vascular endothelial growth factor). CONCLUSION: TRF attenuates the HFD-enhanced spontaneous metastasis of LLC in mice.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Carcinoma Pulmonar de Lewis/secundário , Dieta Hiperlipídica/efeitos adversos , Jejum , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Obesidade/prevenção & controle , Adipocinas/sangue , Adiposidade , Proteínas Angiogênicas/sangue , Animais , Glicemia/metabolismo , Carcinoma Pulmonar de Lewis/sangue , Carcinoma Pulmonar de Lewis/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Citocinas/sangue , Mediadores da Inflamação/sangue , Insulina/sangue , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/genética , Masculino , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/etiologia , Obesidade/genética , Fatores de Tempo , Aumento de Peso
7.
Cancer Res ; 79(4): 783-794, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30770367

RESUMO

Costimulation through 4-1BB (CD137) receptor generates robust CD8+ T-effector and memory responses. The only known ligand, 4-1BBL, is a trimeric transmembrane protein that has no costimulatory activity as a soluble molecule. Thus, agonistic antibodies to the receptor have been used for cancer immunotherapy in preclinical models and are currently being evaluated in the clinic. Here, we report that treatment with an oligomeric form of the ligand, SA-4-1BBL, as a single agent is able to protect mice against subsequent tumor challenge irrespective of the tumor type. Protection was long-lasting (>8 weeks) and a bona fide property of SA-4-1BBL, as treatment with an agonistic antibody to the 4-1BB receptor was ineffective in generating immune protection against tumor challenge. Mechanistically, SA-4-1BBL significantly expanded IFNγ-expressing, preexisting memory-like CD44+CD4+ T cells and NK cells in naïve mice as compared with the agonistic antibody. In vivo blockade of IFNγ or depletion of CD4+ T or NK cells, but not CD8+ T or B cells, abrogated the immunopreventive effects of SA-4-1BBL against cancer. SA-4-1BBL as a single agent also exhibited robust efficacy in controlling postsurgical recurrences. This work highlights unexpected features of SA-4-1BBL as a novel immunomodulator with implications for cancer immunoprevention and therapy. SIGNIFICANCE: This study demonstrates the unique and unexpected immunomodulatory features of SA-4-1BBL that bridge innate and adaptive immune responses with both preventive and therapeutic efficacy against cancer.


Assuntos
Ligante 4-1BB/imunologia , Adjuvantes Imunológicos/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Carcinoma Pulmonar de Lewis/prevenção & controle , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Ligante 4-1BB/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Vacinas Anticâncer/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/metabolismo , Memória Imunológica , Interferon gama/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas
8.
Cancer Res ; 79(8): 1925-1937, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30796053

RESUMO

The unbalanced production of pro- and antiangiogenic factors in tumors can lead to aberrant vasculature morphology, angiogenesis, and disease progression. In this study, we report that disease progression in various murine models of solid tumors is associated with increased cleavage of full-length chromogranin A (CgA), a circulating vasoregulatory neurosecretory protein. Cleavage of CgA led to the exposure of the highly conserved PGPQLR site, which corresponds to residues 368-373 of human CgA1-373, a fragment that has proangiogenic activity. Antibodies against this site, unable to bind full-length CgA, inhibited angiogenesis and reduced tumor perfusion and growth. The PGPQLR sequence of the fragment, but not of the precursor, bound the VEGF-binding site of neuropilin-1; the C-terminal arginine (R373) of the sequence was crucial for binding. The proangiogenic activity of the CgA1-373 was blocked by anti-neuropilin-1 antibodies as well as by nicotinic acetylcholine receptor antagonists, suggesting that these receptors, in addition to neuropilin-1, play a role in the proangiogenic activity of CgA1-373. The R373 residue was enzymatically removed in plasma, causing loss of neuropilin-1 binding and gain of antiangiogenic activity. These results suggest that cleavage of the R373R374 site of circulating human CgA in tumors and the subsequent removal of R373 in the blood represent an important "on/off" switch for the spatiotemporal regulation of tumor angiogenesis and may serve as a novel therapeutic target. SIGNIFICANCE: This work reveals that the interaction between fragmented chromogranin A and neuropilin-1 is required for tumor growth and represents a novel potential therapeutic target.


Assuntos
Neoplasias da Mama/prevenção & controle , Carcinoma Pulmonar de Lewis/prevenção & controle , Cromogranina A/metabolismo , Melanoma/prevenção & controle , Neovascularização Patológica/prevenção & controle , Neuropilina-1/metabolismo , Animais , Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Proliferação de Células , Feminino , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Análise Espaço-Temporal , Células Tumorais Cultivadas
9.
Life Sci ; 201: 102-110, 2018 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-29572180

RESUMO

AIMS: Active immunization with human vascular endothelial growth factor (hVEGF) vaccines provides a therapeutic option instead of bevacizumab therapy. However, the immunity to self-molecule is difficult to elicit due to immune tolerance. A bioactive peptide of two tandem repeats of mHSP70407-426 (M2) has exhibited potent adjuvant ability in our previous study, and the aim of this study was to explore whether M2 could assist hVEGF to display enhanced therapeutic anti-tumor effects. MAIN METHODS: The anti-tumor effects of hVEGF-M2 vaccine were evaluated in both H22 hepatocellular carcinoma and Lewis lung tumor models. CD31 analysis of excised tumors was used to evaluate anti-angiogenesis effects. The titers of anti-VEGF antibody was detected by ELISA and verified by western blot analyses, and the effects of immune sera on HUVEC differentiation were investigated by tube formation assay. KEY FINDINGS: M2 could assist hVEGF to exhibit more favorable therapeutic anti-tumor growth and metastasis effects than hVEGF. Meanwhile, high titer of anti-VEGF antibody was detected in hVEGF-M2 immunized mice sera by ELISA and verified by western blot analysis. Sera from hVEGF-M2 immunized mice could more significantly inhibit HUVEC tube formation than hVEGF immune serum. The hVEGF-M2-immune sera could more effectively inhibit H22 tumor growth and extend the survival rates of H22 tumor bearing mice than hVEGF-immune sera. CD31 analysis of the excised tumors verified a significant reduction in vessel density after hVEGF-M2 vaccination. SIGNIFICANCE: M2 could assist hVEGF to display enhanced anti-tumor effects, which are important for the further application of M2 to enhance antigen-specific immune responses.


Assuntos
Vacinas Anticâncer/farmacologia , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/farmacologia , Repetições Minissatélites/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Inibidores da Angiogênese/farmacologia , Animais , Vacinas Anticâncer/efeitos adversos , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Metástase Neoplásica/prevenção & controle , Proteínas Recombinantes , Vacinação
10.
Oncotarget ; 7(40): 65669-65675, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27582541

RESUMO

The objective of this study was to determine whether a reduction in energy intake ameliorated the high-fat diet-enhanced spontaneous metastasis of Lewis lung carcinoma in mice. Male C57BL/6 mice were fed the AIN93G diet, a high-fat diet or a high-fat diet with a 5% restriction of the intake. Energy restriction reduced body adiposity and body weight, but maintained growth similar to mice fed the AIN93G diet. The high-fat diet significantly increased the number and size (cross-sectional area and volume) of metastases formed in lungs. Restricted feeding reduced the number of metastases by 23%, metastatic cross-sectional area by 32% and volume by 45% compared to the high-fat diet. The high-fat diet elevated plasma concentrations of proinflammatory cytokines (monocyte chemotactic protein-1, plasminogen activator inhibitor-1, leptin), angiogenic factors (vascular endothelial growth factor, tissue inhibitor of metalloproteinase-1) and insulin. Restricted feeding significantly reduced the high-fat diet-induced elevations in plasma concentrations of proinflammatory cytokines, angiogenic factors and insulin. These results demonstrated that a reduction in diet intake by 5% reduced high-fat diet-enhanced metastasis, which may be associated with the mitigation of adiposity and down-regulation of cancer-promoting proinflammatory cytokines and angiogenic factors.


Assuntos
Restrição Calórica , Carcinoma Pulmonar de Lewis/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Dieta , Ingestão de Energia , Neoplasias Pulmonares/prevenção & controle , Animais , Carcinoma Pulmonar de Lewis/etiologia , Carcinoma Pulmonar de Lewis/patologia , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus
11.
BMC Cancer ; 16: 462, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27412241

RESUMO

BACKGROUND: The ever increasing knowledge in the areas of cell biology, the immune system and the mechanisms of cancer are allowing a new phase of immunotherapy to develop. The aim of cancer vaccination is to activate the host immune system and some success has been observed particularly in the use of the BCG vaccine for bladder cancer as an immunostimulant. Reovirus, an orphan virus, has proven itself as an oncolytic virus in vitro and in vivo. Over 80 % of tumour cell lines have been found to be susceptible to Reovirus infection and it is currently in phase III clinical trials. It has been shown to induce immune responses to tumours with very low toxicities. METHODS: In this study, Reovirus was examined in two main approaches in vivo, in mice, using the melanoma B16F10 and Lewis Lung Carcinoma (LLC) models. Initially, mice were treated intratumourally (IT) with Reovirus and the immune responses determined by cytokine analysis. Mice were also vaccinated using a cell-based Reovirus vaccine and subsequently exposed to a tumourigenic dose of cells (B16F10 or LLC). Using the same cell-based Reovirus vaccine, established tumours were treated and subsequent immune responses and virus retrieval investigated. RESULTS: Upregulation of several cytokines was observed following treatment and replication-competent virus was also retrieved from treated tumours. Varying levels of cytokine upregulation were observed and no replication-competent virus was retrieved in vaccine-treated mice. Prolongation of survival and delayed tumour growth were observed in all models and an immune response to Reovirus, either using Reovirus alone or a cell-based vaccine was also observed in all mice. CONCLUSION: This study provides evidence of immune response to tumours using a cell-based Reovirus vaccine in both tumour models investigated, B16F10 and LLC, cytokine induction was observed with prolongation of survival in almost all cases which may suggest a new method for using Reovirus in the clinic.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Pulmonar de Lewis/terapia , Imunoterapia/métodos , Orthoreovirus Mamífero 3 , Melanoma Experimental/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Cutâneas/terapia , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Linhagem Celular Tumoral , Citocinas/metabolismo , Imunoterapia/efeitos adversos , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/prevenção & controle , Taxa de Sobrevida , Vacinação
12.
J Cancer Res Clin Oncol ; 142(1): 111-23, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26139353

RESUMO

PURPOSE: Human umbilical endothelial cells (HUVECs) have been proved as an effective whole-cell vaccine inhibiting tumor angiogenesis. However, HUVECs divide a very limited number of passages before entering replicative senescence, which limits its application for clinical situation. Here, we fused HUVECs with human pulmonary adenocarcinoma cell line A549s and investigated the anti-tumor immunity of the hybrids against mice Lewis lung cancer. METHODS: HUVECs were fused with A549s using polyethylene glycol and were sorted by flow cytometry. The fusion cells (HUVEC-A549s) were confirmed by testing the expression of telomerase and VE-cadherin, the senescence-associated ß-galactosidase activity, and tube formation ability. HUVEC-A549s were then irradiated and injected into the C57BL/6 mice of protective, therapeutic, and metastatic models. The mechanism of the anti-tumor immunity was explored by analyzing mice sera, spleen T lymphocytes, tumor microenvironment, and histological changes. RESULTS: HUVEC-A549s coexpressed tumor and endothelial markers and maintained the vascular function of tube forming at passage 30 without showing signs of senescence. HUVEC-A549s could induce protective and therapeutic anti-tumor activity for LL(2) model and presented stronger activity against metastasis than HUVECs. Both humoral and cellular immunity were participated in the anti-angiogenic activity, as HUVECs-neutralizing IgG and HUVECs-toxic lymphocytes were increased. Angiogenic mediators (VEGF and TGF-ß) and tumor microenvironment cells MDSCs and Tregs were also diminished. CONCLUSIONS: Our findings might provide a novel strategy for HUVECs-related immunotherapy, and this vaccine requires lower culture condition than primary HUVECs while enhancing the anti-tumor immunity.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Endotélio Vascular/imunologia , Imunidade Celular/imunologia , Neovascularização Patológica/imunologia , Linfócitos T Citotóxicos/imunologia , Veias Umbilicais/imunologia , Animais , Western Blotting , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/imunologia , Proliferação de Células , Células Cultivadas , Senescência Celular , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana/imunologia , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/metabolismo , Veias Umbilicais/citologia , Vacinação
13.
Oncotarget ; 6(38): 40667-79, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26512779

RESUMO

The gelatinase inhibitor doxycycline is the prototypical antitumor antibiotic. We investigated the effects of doxycycline on the migration, invasion, and metastasis of human lung cancer cell lines and in a mouse model. We also measured the effect of doxycycline on the transcription of epithelial-mesenchymal transition (EMT) markers, and used immunohistochemistry to determine whether EMT reversal was associated with doxycycline inhibition. Doxycycline dose-dependently inhibited proliferation, migration, and invasion of NCI-H446 human small cell lung cancer cells. It also suppressed tumor growth from NCI-H446 and A549 lung cancer cell xenografts without altering body weight, inhibited Lewis lung carcinoma cell migration, and prolonged survival. The activities of the transcription factors Twist1/2, SNAI1/2, AP1, NF-κB, and Stat3 were suppressed by doxycycline, which reversed EMT and inhibited signal transduction, thereby suppressing tumor growth and metastasis. Our data demonstrate functional targeting of transcription factors by doxycycline to reverse EMT and suppress tumor proliferation and metastasis. Thus, doxycycline selectively targets malignant tumors and reduces its metastatic potential with less cytotoxicity in lung cancer patients.


Assuntos
Antibacterianos/farmacologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Doxiciclina/farmacologia , Neoplasias Pulmonares/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Transição Epitelial-Mesenquimal , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Células Tumorais Cultivadas , Cicatrização/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Oncotarget ; 6(13): 11009-22, 2015 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-25826092

RESUMO

Leucine-rich α2-glycoprotein (LRG) is an approximately 50-kDa glycoprotein that has been found to be elevated in the sera of patients with several types of cancer. LRG directly binds to transforming growth factor beta 1 (TGFß1) and modulates TGFß1 signaling in endothelial cells; however, the precise function of LRG in cancer remains unclear. This study aimed to investigate the role of LRG in cancer. Lewis lung carcinoma (LLC) cells hardly expressed LRG. The growth of LLC tumors allografted in the LRG knockout (KO) mice was significantly increased compared with wild-type (WT) mice. Conversely, overexpression of LRG significantly inhibited the growth of LLC tumors in WT mice. In the presence of LRG, TGFß1 significantly inhibited the proliferation of LLC cells and human hepatocellular carcinoma Hep3B cells in vitro by inducing apoptosis via the potent activation of smad2 and its downstream signaling pathway. Furthermore, administration of a TGFßR1 inhibitor (SB431542) significantly enhanced the growth of LLC tumors in WT mice compared with LRG KO mice via inhibition of apoptosis. We propose that LRG potentiates the effect of TGFß1 in cancer cells whose growth is suppressed in the presence of TGFß1.


Assuntos
Apoptose , Carcinoma Hepatocelular/prevenção & controle , Carcinoma Pulmonar de Lewis/prevenção & controle , Glicoproteínas/fisiologia , Neoplasias Hepáticas/prevenção & controle , Fator de Crescimento Transformador beta1/metabolismo , Animais , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Proliferação de Células , Feminino , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Oncotarget ; 6(7): 5382-411, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25742784

RESUMO

The efficacy of anti-angiogenic therapies on cancer patients is limited by the emergence of drug resistance, urging the search for second-generation drugs. In this study, we screened an academic chemical library (DCM, University of Grenoble-Alpes) and identified a leader molecule, COB223, that inhibits endothelial cell migration and proliferation. It inhibits also Lewis lung carcinoma (LLC/2) cell proliferation whereas it does not affect fibroblast proliferation. The anti-angiogenic activity of COB223 was confirmed using several in vitro and in vivo assays. In a mouse LLC/2 tumor model, ip administration of doses as low as 4 mg/kg COB223 efficiently reduced the tumor growth rate. We observed that COB223 inhibits endothelial cell ERK1/2 phosphorylation induced by VEGF, FGF-2 or serum and that it acts downstream of PKC and upstream of Ras. This molecule represents a novel anti-angiogenic and anti-tumorigenic agent with an original mechanism of action that deserves further development as an anti-cancer drug.


Assuntos
Antineoplásicos/farmacologia , Carbamatos/farmacologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Sulfonamidas/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Proteínas ras/metabolismo , Animais , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Western Blotting , Carbamatos/síntese química , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/patologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Pele/citologia , Pele/efeitos dos fármacos , Pele/metabolismo , Sulfonamidas/síntese química , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Genet Mol Res ; 13(3): 6827-36, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25177962

RESUMO

Recent studies have found that glucocorticoids are closely associated with oncogenesis and the development of many types of tumors. The aim of this study was to observe the effect of dexamethasone on the growth and angiogenesis of transplanted Lewis lung carcinoma in mice. Lewis lung carcinoma cells were inoculated subcutaneously into the right axilla of C57BL/6 mice, and the mice were randomly divided into 3 groups: the control group, cisplatin group, and dexamethasone group. From day 7 after inoculation, all the mice were given different treatments for 10 days, and changes in xenograft tumor volumes were monitored. All mice were sacrificed on day 17, and the tumors were obtained and weighed and the tumor inhibitory rate was calculated. The expression levels of hypoxia inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF), as well as the microvessel density (MVD) in the tumor mass, were measured by immunohistochemistry. Tumor growth was suppressed in the cisplatin group and dexamethasone group. The weights of tumors were markedly decreased in the cisplatin group and dexamethasone group compared with the control group (P < 0.05). The expression levels of HIF-1α and VEGF and the MVD were significantly lower in the cisplatin group and dexamethasone group than in the control group (P < 0.05). However, these levels were not significantly different between the cisplatin group and dexamethasone group (P > 0.05). Dexamethasone can effectively inhibit the growth and angiogenesis of Lewis lung carcinoma by inhibiting the expression of HIF-1α and VEGF.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Dexametasona/farmacologia , Neovascularização Patológica/prevenção & controle , Carga Tumoral/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Feminino , Glucocorticoides/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Distribuição Aleatória , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Oncotarget ; 5(9): 2761-77, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24811731

RESUMO

Tumor blood vessels are leaky and immature, which causes inadequate blood supply to tumor tissues resulting in hypoxic microenvironment and promotes metastasis. Here we have explored tumor vessel modulating activity of Sac-1004, a recently developed molecule in our lab, which directly potentiates VE-cadherin-mediated endothelial cell junction. Sac-1004 could enhance vascular junction integrity in tumor vessels and thereby inhibit vascular leakage and enhance vascular perfusion. Improved perfusion enabled Sac-1004 to have synergistic anti-tumor effect on cisplatin-mediated apoptosis of tumor cells. Interestingly, characteristics of normalized blood vessels namely reduced hypoxia, improved pericyte coverage and decreased basement membrane thickness were readily observed in tumors treated with Sac-1004. Remarkably, Sac-1004 was also able to inhibit lung and lymph node metastasis in MMTV and B16BL6 tumor models. This was in correlation with a reduction in epithelial-to-mesenchymal transition of tumor cells with considerable diminution in expression of related transcription factors. Moreover, cancer stem cell population dropped substantially in Sac-1004 treated tumor tissues. Taken together, our results showed that direct restoration of vascular junction could be a significant strategy to induce normalization of tumor blood vessels and reduce metastasis.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Carcinoma Pulmonar de Lewis/prevenção & controle , Endotélio Vascular/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/prevenção & controle , Neovascularização Patológica/prevenção & controle , Saponinas/farmacologia , Animais , Antígenos CD/genética , Apoptose/efeitos dos fármacos , Western Blotting , Caderinas/genética , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Retinopatia Diabética/prevenção & controle , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células HeLa , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Metástase Linfática , Células MCF-7 , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Int J Cancer ; 135(3): 742-50, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24382818

RESUMO

Tumor necrosis factor (TNF) has remarkable antitumor effects, but its systemic therapeutic use is prevented by its lethal inflammatory effects. TNFR1 (P55) is essential for both the antitumor and toxic effects because both of them are absent in P55-deficient mice. In previous work we demonstrated that P55+/- mice are completely resistant to TNF toxicity, while the antitumor effects induced by TNF combined with interferon gamma (IFNγ) remain fully functional in these mice. Hence, a high dose of TNF/IFNγ has an excellent therapeutic potential when P55 levels are reduced, because TNF induces tumor regression without systemic toxicity. Here, we provide proof of principle for therapeutic application of this approach by using antisense oligonucleotides (ASOs). Treatment of mice with ASOs targeting P55 resulted in a strong reduction in P55 protein levels in liver, small intestine and blood mononuclear cells. This P55 downregulation was associated with significant protection of mice against acute TNF toxicity as measured by hypothermia, systemic inflammation and lethality. This treatment also protected mice against toxicity of TNF/IFNγ treatment in several cancer models: B16Bl6, Lewis lung carcinoma and a lung colony model. Our results confirm the therapeutic value of this strategy, which could lead to the development of a safer and more effective TNF/IFNγ antitumor therapy.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Modelos Animais de Doenças , Interferon gama/toxicidade , Melanoma Experimental/prevenção & controle , Oligonucleotídeos Antissenso/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/toxicidade , Animais , Carcinoma Pulmonar de Lewis/induzido quimicamente , Carcinoma Pulmonar de Lewis/genética , Feminino , Intestino Delgado/metabolismo , Fígado/metabolismo , Dose Máxima Tolerável , Melanoma Experimental/induzido quimicamente , Melanoma Experimental/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Transdução de Sinais
19.
Int J Cancer ; 134(4): 849-58, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23913502

RESUMO

The tumor microenvironment plays a pivotal role during cancer development and progression. The balance between suppressive and cytotoxic responses of the tumor immune microenvironment has been shown to have a direct effect on the final outcome in various human and experimental tumors. Recently, we demonstrated that the oxygen sensor HIF-prolyl hydroxylase-2 (PHD2) plays a detrimental role in tumor cells, stimulating systemic growth and metastasis in mice. In our current study, we show that the conditional ablation of PHD2 in the hematopoietic system also leads to reduced tumor volume, intriguingly generated by an imbalance between enhanced cell death and improved proliferation of tumor cells. This effect seems to rely on the overall downregulation of protumoral as well as antitumoral cytokines. Using different genetic approaches, we were able to confine this complex phenotype to the crosstalk of PHD2-deficient myeloid cells and T-lymphocytes. Taken together, our findings reveal a multifaceted role for PHD2 in several hematopoietic lineages during tumor development and might have important implications for the development of tumor therapies in the future.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Prolina Dioxigenases do Fator Induzível por Hipóxia/fisiologia , Melanoma Experimental/prevenção & controle , Células Mieloides/patologia , Linfócitos T/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Medula Óssea/metabolismo , Medula Óssea/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular , Proliferação de Células , Citocinas/genética , Citocinas/metabolismo , Progressão da Doença , Citometria de Fluxo , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Integrases/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia , Linfócitos T/metabolismo
20.
Asian Pac J Cancer Prev ; 14(4): 2307-10, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23725132

RESUMO

Curcumin previously was proven to inhibit angiogenesis and display potent antitumor activity in vivo and in vitro. In the present study, we investigated whether a combination curcumin with hyperthermia would have a synergistic antitumor effect in the LL/2 model. The results indicated that combination therapy significantly inhibited cell proliferation of MS-1 and LL/2 in vitro. LL/2 experiment model also demonstrated that the combination therapy inhibited tumor growth and prolonged the life span in vivo. Furthermore, combination therapy reduced angiogenesis and increased tumor apoptosis. Our findings suggest that the combination therapy exerted synergistic antitumor effects, providing a new perspective fpr clinical tumor therapy.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose , Carcinoma Pulmonar de Lewis/prevenção & controle , Curcumina/uso terapêutico , Hipertermia Induzida , Neovascularização Patológica/prevenção & controle , Animais , Western Blotting , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Proliferação de Células , Terapia Combinada , Feminino , Imunofluorescência , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...