Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Oncol ; 13: 1145817, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37064104

RESUMO

Background: Three-dimensional primary slice cultures (SC) of head and neck squamous cell carcinomas (HNC) are realistic preclinical models. Until now, preserving structure and viability ex vivo for several days has been difficult. The aim of this study was to optimize cultivation conditions for HNC SC and analyze the added effects of platelet rich fibrin (PRF) on these conditions. Methods: SC were prepared from the tumor biopsies of 9 HNC patients. Cultures were incubated for 1 and 7 days in three different media- Keratinocyte serum-free medium (SFM), RPMI-1640i, and 1:1 mix of both, with and without addition of PRF. After culturing, SC were fixated, embedded, and stained with Hematoxylin-Eosin (HE) and cleaved caspase-3. In addition, triple immune fluorescence staining for cytokeratin, vimentin and CD45 was performed. Outcome parameters were cell count and cell density, viability and apoptosis, SC total area and proportions of keratinocytes, mesenchymal and immune cells. The effects of culture time, medium, and addition of PRF were calculated in an SPSS generalized linear model and using the Wald Chi-Squared test. Results: Ninety-four slice cultures were analyzed. Viability remained stable for 7 days in culture. After addition of PRF, cell viability increased (p=0.05). SC total area decreased (0.44 ± 0.04 mm2 on day 1 (95% CI: 0.35 to 0.56) to 0.29 ± 0.03 mm2 on day 7 (95% CI: 0.22 to 0.36), but cell density and cell proportions remained stable. Differences in cultivation media had no significant impact on outcome parameters. Conclusion: HNC SC can be preserved for up to 7 days using the tested cultivation media. Cell viability was best preserved with addition of PRF. HNC SC are a versatile experimental tool to study physiology and drug actions. Autologous PRF can help simulate realistic conditions in vitro.

2.
Sci Rep ; 12(1): 15334, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36097280

RESUMO

Head and neck cancer etiology and architecture is quite diverse and complex, impeding the prediction whether a patient could respond to a particular cancer immunotherapy or combination treatment. A concomitantly arising caveat is obviously the translation from pre-clinical, cell based in vitro systems as well as syngeneic murine tumor models towards the heterogeneous architecture of the human tumor ecosystems. To bridge this gap, we have established and employed a patient-derived HNSCC (head and neck squamous cell carcinoma) slice culturing system to assess immunomodulatory effects as well as permissivity and oncolytic virus (OV) action. The heterogeneous contexture of the human tumor ecosystem including tumor cells, cancer-associated fibroblasts and immune cells was preserved in our HNSCC slice culturing approach. Importantly, the immune cell compartment remained to be functional and cytotoxic T-cells could be activated by immunostimulatory antibodies. In addition, we uncovered that a high proportion of the patient-derived HNSCC slice cultures were susceptible to the OV VSV-GP. More specifically, VSV-GP infects a broad spectrum of tumor-associated lineages including epithelial and stromal cells and can induce apoptosis. In sum, this human tumor ex vivo platform might complement pre-clinical studies to eventually propel cancer immune-related drug discovery and ease the translation to the clinics.


Assuntos
Neoplasias de Cabeça e Pescoço , Vírus Oncolíticos , Animais , Ecossistema , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Imunoterapia , Camundongos , Vírus Oncolíticos/fisiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia
3.
Laryngorhinootologie ; 101(10): 787-796, 2022 10.
Artigo em Alemão | MEDLINE | ID: mdl-35977557

RESUMO

OBJECTIVE: Oncolytic viruses (OV) infect and kill cancer cells and elicit an antitumoral immune response. With their potential to break through tumor immunoresistance, OV might be a future combination treatment option in patients with advanced head and neck cancer (HNC). Modes of action, biological modifications, handling and side effects of OV for treatment of HNC are reviewed. Results of preclinical and clinical trials are reported. METHODS: Publications and clinical trials dealing with OV and HNC were searched in PubMed and international platforms for clinical study records. Studies on preclinical and clinical trials regarding oncolytic Herpes Simplex Virus (HSV), Adenovirus, Vacciniavirus and Reovirus were selected. RESULTS: Enhanced infection and killing of tumor cells through capsid and genome modifications of OV were reported in recent preclinical studies. Most of the clinical studies were phase-I/II trials. In phase III studies, tumor regression and prolonged survival were observed after treatment with oncolytic HSV, Adenoviruses and Reoviruses. In most trials, OV were combined with chemoradiotherapy or immunotherapy. CONCLUSION: In the published studies, OV treatment of HNC patients was safe, often well tolerated and showed promising results with regard to response and survival, especially in combination with chemoradiotherapy or checkpoint inhibitors.


Assuntos
Neoplasias de Cabeça e Pescoço , Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Imunoterapia/métodos , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética
4.
Nat Cell Biol ; 23(5): 497-510, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33972730

RESUMO

The Drosophila trachea, as the functional equivalent of mammalian blood vessels, senses hypoxia and oxygenates the body. Here, we show that the adult intestinal tracheae are dynamic and respond to enteric infection, oxidative agents and tumours with increased terminal branching. Increased tracheation is necessary for efficient damage-induced intestinal stem cell (ISC)-mediated regeneration and is sufficient to drive ISC proliferation in undamaged intestines. Gut damage or tumours induce HIF-1α (Sima in Drosophila), which stimulates tracheole branching via the FGF (Branchless (Bnl))-FGFR (Breathless (Btl)) signalling cascade. Bnl-Btl signalling is required in the intestinal epithelium and the trachea for efficient damage-induced tracheal remodelling and ISC proliferation. Chemical or Pseudomonas-generated reactive oxygen species directly affect the trachea and are necessary for branching and intestinal regeneration. Similarly, tracheole branching and the resulting increase in oxygenation are essential for intestinal tumour growth. We have identified a mechanism of tracheal-intestinal tissue communication, whereby damage and tumours induce neo-tracheogenesis in Drosophila, a process reminiscent of cancer-induced neoangiogenesis in mammals.


Assuntos
Transformação Celular Neoplásica/metabolismo , Hipóxia/metabolismo , Oxigênio/metabolismo , Regeneração/fisiologia , Animais , Animais Geneticamente Modificados/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/genética
5.
Br J Cancer ; 121(8): 647-658, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31530903

RESUMO

BACKGROUND: Oncolytic virotherapy is thought to result in direct virus-induced lytic tumour killing and simultaneous activation of innate and tumour-specific adaptive immune responses. Using a chimeric vesicular stomatitis virus variant VSV-GP, we addressed the direct oncolytic effects and the role of anti-tumour immune induction in the syngeneic mouse lung cancer model LLC1. METHODS: To study a tumour system with limited antiviral effects, we generated interferon receptor-deficient cells (LLC1-IFNAR1-/-). Therapeutic efficacy of VSV-GP was assessed in vivo in syngeneic C57BL/6 and athymic nude mice bearing subcutaneous tumours. VSV-GP treatment effects were analysed using bioluminescent imaging (BLI), immunohistochemistry, ELISpot, flow cytometry, multiplex ELISA and Nanostring® assays. RESULTS: Interferon insensitivity correlated with VSV-GP replication and therapeutic outcome. BLI revealed tumour-to-tumour spread of viral progeny in bilateral tumours. Histological and gene expression analysis confirmed widespread and rapid infection and cell killing within the tumour with activation of innate and adaptive immune-response markers. However, treatment outcome was increased in the absence of CD8+ T cells and surviving mice showed little protection from tumour re-challenge, indicating limited therapeutic contribution by the activated immune system. CONCLUSION: These studies present a case for a predominantly lytic treatment effect of VSV-GP in a syngeneic mouse lung cancer model.


Assuntos
Carcinoma Pulmonar de Lewis/terapia , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica/métodos , Vesiculovirus , Imunidade Adaptativa/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD8-Positivos/imunologia , Carcinoma Pulmonar de Lewis/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Quimera , Citocinas/imunologia , Técnicas de Inativação de Genes , Imunidade Inata/imunologia , Técnicas In Vitro , Interferon Tipo I/imunologia , Interferon-alfa/imunologia , Interferon gama/imunologia , Neoplasias Pulmonares/genética , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Nus , Receptor de Interferon alfa e beta/genética , Vesiculovirus/genética , Vesiculovirus/imunologia , Proteínas do Envelope Viral/genética , Proteínas Virais/genética
6.
Int J Cancer ; 145(7): 1958-1969, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30972741

RESUMO

The efficacy of cancer vaccines has been limited by the immunosuppressive tumor microenvironment, which can be alleviated by immune checkpoint inhibitor (ICI) therapy. Here, we tested if oncolytic viruses (OVs), similar to ICI, can also synergize with cancer vaccines by modulating the tumor microenvironment. VSV-GP, a chimeric vesicular stomatitis virus (VSV) pseudotyped with the glycoprotein (GP) of the lymphocytic choriomeningitis virus, is a promising new OV candidate. Here, we show that in mouse B16-OVA melanoma, combination treatment of VSV-GP with an ovalbumin (OVA) peptide-loaded dendritic cell (DC) vaccine (DCVacc) significantly enhanced survival over the single agent therapies, although both DCVacc and DCVacc/VSV-GP treatments induced comparable levels of OVA-specific CD8 T cell responses. Virus replication was minimal so that direct viral oncolysis in B16-OVA did not contribute to this synergism. The strong therapeutic effect of the DCVacc/VSV-GP combination treatment was associated with high numbers of tumor-infiltrating, highly activated T cells and the relative reduction of regulatory T cells in treated and contra-lateral nontreated tumors. Accordingly, depletion of CD8 T cells but not natural killer cells abrogated the therapeutic effect of DCVacc/VSV-GP supporting the crucial role of CD8 T cells. In addition, a drastic increase in several proinflammatory cytokines was observed in VSV-GP-treated tumors. Taken together, OVs, similar to ICI, have the potential to markedly increase the efficacy of cancer vaccines by alleviating local immune suppression in the tumor microenvironment.


Assuntos
Vacinas Anticâncer/administração & dosagem , Glicoproteínas/metabolismo , Melanoma Experimental/terapia , Terapia Viral Oncolítica/métodos , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Glicoproteínas/genética , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Melanoma Experimental/imunologia , Camundongos , Vírus Oncolíticos/fisiologia , Ovalbumina/imunologia , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Int J Cancer ; 143(7): 1786-1796, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29696636

RESUMO

Oncolytic viruses, including the oncolytic rhabdovirus VSV-GP tested here, selectively infect and kill cancer cells and are a promising new therapeutic modality. Our aim was to study the efficacy of VSV-GP, a vesicular stomatitis virus carrying the glycoprotein of lymphocytic choriomeningitis virus, against prostate cancer, for which current treatment options still fail to cure metastatic disease. VSV-GP was found to infect 6 of 7 prostate cancer cell lines with great efficacy. However, susceptibility was reduced in one cell line with low virus receptor expression and in 3 cell lines after interferon alpha treatment. Four cell lines had developed resistance to interferon type I at different levels of the interferon signaling pathway, resulting in a deficient antiviral response. In prostate cancer mouse models, long-term remission was achieved upon intratumoral and, remarkably, also upon intravenous treatment of subcutaneous tumors and bone metastases. These promising efficacy data demonstrate that treatment of prostate cancer with VSV-GP is feasible and safe in preclinical models and encourage further preclinical and clinical development of VSV-GP for systemic treatment of metastatic prostate cancer.


Assuntos
Efeito Citopatogênico Viral , Modelos Animais de Doenças , Terapia Viral Oncolítica , Neoplasias da Próstata/terapia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Apoptose , Proliferação de Células , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/patologia , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nat Commun ; 6: 5874, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25608467

RESUMO

Epithelial cancer constitutes a major clinical challenge and molecular mechanisms underlying the process of tumour initiation are not well understood. Here we demonstrate that hair follicle bulge stem cells (SCs) give rise to well-differentiated sebaceous tumours and show that SCs are not only crucial in tumour initiation, but are also involved in tumour plasticity and heterogeneity. Our findings reveal that SC-specific expression of mutant Lef1, which mimics mutations found in human sebaceous tumours, drives sebaceous tumour formation. Mechanistically, we demonstrate that mutant Lef1 abolishes p53 activity in SCs. Intriguingly, mutant Lef1 induces DNA damage and interferes with SC-specific gatekeeper functions normally protecting against accumulations of DNA lesions and cell loss. Thus, normal control of SC proliferation is disrupted by mutant Lef1, thereby allowing uncontrolled propagation of tumour-initiating SCs. Collectively, these findings identify underlying molecular and cellular mechanisms of tumour-initiating events in tissue SCs providing a potential target for future therapeutic strategies.


Assuntos
Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Neoplasias das Glândulas Sebáceas/patologia , Neoplasias Cutâneas/patologia , Células-Tronco/citologia , Proteína Supressora de Tumor p53/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animais , Animais Recém-Nascidos , Apoptose , Carcinogênese , Carcinógenos , Linhagem da Célula , Proliferação de Células , Separação Celular , Transformação Celular Neoplásica/patologia , Cruzamentos Genéticos , Dano ao DNA , Progressão da Doença , Epiderme/metabolismo , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Neoplasias das Glândulas Sebáceas/metabolismo , Neoplasias Cutâneas/metabolismo , Raios Ultravioleta
9.
Methods Mol Biol ; 989: 45-60, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23483386

RESUMO

Lineage tracing of tissue stem cells represents a powerful tool to address fundamental questions of deve-lopment, differentiation and cellular renewal in a natural tissue environment. The Cre/lox site-specific recombination system is increasingly used to genetically label specific cell populations to perform cell lineage tracing or fate mapping experiments in sophisticated mouse models. Here we describe a method of labeling and subsequent tracking stem cells of the hair follicle bulge region in mouse skin. Hair follicle stem cells are specifically labeled by expressing the Cre recombinase under control of keratin15 (K15) regulatory sequences and by crossing the Cre-containing animals with Cre-sensitive Rosa26R (R26R) reporter mice. To achieve a temporal control of recombinase activity in stem cells, Cre is fused to a modified estrogen receptor (CreER(G)T2). In the K15CreER(G)T2/R26R mouse model, hair follicle stem cells (HFSCs) are specifically labeled after Cre activation upon treatment of mice with tamoxifen. By analyzing the skin tissue at different time points following genetic labeling, important information on stem cell behavior and contribution of labeled stem cells to epidermal structures during tissue homeostasis and hair follicle regeneration are obtained. Combining the lineage tracing approach with the whole mount technique allows examining large areas of the epidermis containing many hair follicles and sebaceous glands and reveals the complex three-dimensional relationship of labeled stem cell clones within the tissue.


Assuntos
Células Epidérmicas , Folículo Piloso/citologia , Células-Tronco/citologia , Animais , Linhagem da Célula , Células Cultivadas , Epiderme/efeitos dos fármacos , Folículo Piloso/efeitos dos fármacos , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pele/citologia , Pele/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Tamoxifeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA