Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 113
Filtrer
1.
JTO Clin Res Rep ; 2(11): 100242, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34806054

RÉSUMÉ

INTRODUCTION: To assess the technical feasibility and safety of repeated percutaneous computed tomography (CT)-guided transthoracic biopsies and intratumoral injections of gene-modified dendritic cells in metastatic NSCLC. METHODS: A total of 15 patients with 15 NSCLC lesions measuring greater than 1.0 cm underwent two cycles of intratumoral biopsies and CCL21 dendritic cell injections separated by 7 days. All needle placements and injections were done under CT guidance. Clinical and imaging follow-up was done approximately 4 weeks after the first procedure. Safety and feasibility were determined as: (1) safety and feasibility similar to that of single-needle biopsy, and (2) an absence of serious adverse events defined as grade greater than or equal to three according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. RESULTS: A total of 30 percutaneous, transthoracic intratumoral biopsies and injections into the lung cancer were performed, two cycles (at d 0 and 7) received by each patient (311 biopsies and 96 intratumoral injections). All percutaneous cases achieved technical success with respect to needle placement for both biopsy and injection of CCL21 dendritic cells. Only minor complications were observed (grade <3), including pneumothorax (n = 10, 33%) and small postbiopsy hemorrhage (n = 2, 7%). Pneumothorax was moderate (n = 1) or trace (n = 9), with resolution of the moderate pneumothorax after manual aspiration without chest tube placement. No patient required chest tube placement. No other complications or serious adverse effects related to the biopsy or dendritic cell injection were noted. All patients were in stable condition after up to 4 hours in the recovery unit and were discharged home on the same day. No procedure-related complications were observed on imaging or clinical follow-up at 4 weeks. CONCLUSIONS: Repeated percutaneous, transthoracic CT-guided biopsies and intratumoral gene-modified cell-based immunotherapy injections into lung cancers are technically feasible, safe, and reproducible. There were no procedure-related serious (defined as grade ≥3) adverse events.

2.
Acta Pharm Sin B ; 11(5): 1274-1285, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-34094833

RÉSUMÉ

Liver is the most common metastatic site for colorectal cancer (CRC), there is no satisfied approach to treat CRC liver metastasis (CRCLM). Here, we investigated the role of a polycomb protein BMI-1 in CRCLM. Immunohistochemical analysis showed that BMI-1 expression in liver metastases was upregulated and associated with T4 stage, invasion depth and right-sided primary tumor. Knockdown BMI-1 in high metastatic HCT116 and LOVO cells repressed the migratory/invasive phenotype and reversed epithelial-mesenchymal transition (EMT), while BMI-1 overexpression in low metastatic Ls174T and DLD1 cells enhanced invasiveness and EMT. The effects of BMI-1 in CRC cells were related to upregulating snail via AKT/GSK-3ß pathway. Furthermore, knockdown BMI-1 in HCT116 and LOVO cells reduced CRCLM using experimental liver metastasis mice model. Meanwhile, BMI-1 overexpression in Ls174T and DLD1 significantly increased CRCLM. Moreover, sodium butyrate, a histone deacetylase and BMI-1 inhibitor, reduced HCT116 and LOVO liver metastasis in immunodeficient mice. Our results suggest that BMI-1 is a major regulator of CRCLM and provide a potent molecular target for CRCLM treatment.

3.
Vaccines (Basel) ; 8(4)2020 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-33167311

RÉSUMÉ

BACKGROUND: Immune checkpoint blockade that downregulates T cell evasion for effective immunity has provided a renewed interest in therapeutic cancer vaccines. METHODS: Utilizing murine lung cancer models, we determined: tumor burden, TIL cytolysis, immunohistochemistry, flow cytometry, RNA Sequencing, CD4 T cells, CD8 T cells, CXCL9 chemokine, and CXCL10 chemokine neutralization to evaluate the efficacy of Programmed cell death protein 1 (PD-1) blockade combined with chemokine (C-C motif) ligand 21-dendritic cell tumor antigen (CCL21-DC tumor Ag) vaccine. RESULTS: Anti-PD1 combined with CCL21-DC tumor Ag vaccine eradicated 75% of 12-day established tumors (150 mm3) that was enhanced to 90% by administering CCL21-DC tumor Ag vaccine prior to combined therapy. The effect of combined therapy was blocked by CD4, CD8, CXCL9, and CXCL10 neutralizing antibodies. CONCLUSION: PD-1 blockade therapy plus CCL21-DC tumor Ag vaccine could be beneficial to lung cancer patients.

4.
Vaccines (Basel) ; 8(2)2020 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-32570793

RÉSUMÉ

Background: Targeting inhibitory immune checkpoint molecules has highlighted the need to find approaches enabling the activation of immune responses against cancer. Therapeutic vaccination, which induces specific immune responses against tumor antigens (Ags), is an attractive option. Methods: Utilizing a K-RasG12Dp53null murine lung cancer model we determined tumor burden, tumor-infiltrating T cell (TIL) cytolysis, immunohistochemistry, flow cytometry, and CD4 and CD8 depletion to evaluate the efficacy of PD-1 blockade combined with CCL21-DC tumor lysate vaccine. Results: Anti-PD-1 plus CCL21-DC tumor lysate vaccine administered to mice bearing established tumors (150 mm3) increased expression of perforin and granzyme B in the tumor microenvironment (TME), increased tumor-infiltrating T cell (TIL) activity, and caused 80% tumor eradication. Mice with treatment-induced tumor eradication developed immunological memory, enabling tumor rejection upon challenge and cancer-recurrence-free survival. The depletion of CD4 or CD8 abrogated the antitumor activity of combined therapy. PD-1 blockade or CCL21-DC tumor lysate vaccine monotherapy reduced tumor burden without tumor eradication. Conclusion: Immune checkpoint blockade promotes the activity of the therapeutic cancer vaccine. PD-1 blockade plus CCL21-DC tumor lysate vaccine therapy could benefit lung cancer patients.

5.
Adv Exp Med Biol ; 1231: 67-78, 2020.
Article de Anglais | MEDLINE | ID: mdl-32060847

RÉSUMÉ

CCL21 promotes immune activity in the tumor microenvironment (TME) by colocalizing dendritic cells (DC) and T cells programing ectopic lymph node architectural structures that correlate with cancer prognosis. Innovative strategies to deliver CCL21 in cancer patients will reactivate the downregulated immune activity in the TME. Immune escape mechanisms are upregulated in the TME that promote tumor immune evasion. CCL21 combined with inhibition of dominant pathways of immune evasion will aid in the development of effective immunotherapy for cancer.


Sujet(s)
Chimiokine CCL21/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Microenvironnement tumoral/immunologie , Animaux , Cellules dendritiques/immunologie , Humains , Immunothérapie , Lymphocytes T/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie
6.
Oxid Med Cell Longev ; 2019: 2352079, 2019.
Article de Anglais | MEDLINE | ID: mdl-31346357

RÉSUMÉ

Radiation-induced oral mucositis (RIOM) is one of the most common side effects of radiotherapy in cancer patients, especially in almost all head and neck cancer patients. It presents as severe pain and ulceration. The development of RIOM is composed of five stages: initiation, primary damage response, signal amplification, ulceration, and healing. However, the key regulators involved in the RIOM pathogenesis remain largely unknown. In this study, we reveal a novel role of miR-200c, a member of the miR-200 family, in modulating RIOM pathogenesis. Using a mouse model mimicking RIOM, we found that the miR-200 family numbers (miR-141, miR-200a, miR-200b, and miR-200c) except miR-429 were significantly induced during the RIOM formation. Besides, in RIOM mice, miR-200c expression level was also increased dramatically in the normal human keratinocytes (NHKs) after irradiation. Knockdown of miR-200c expression with miR-200c-3p-shRNA significantly reduced senescence phenotype and enhanced cell proliferation in NHKs after irradiation. The generation of reactive oxygen species (ROS) and p47 enzyme involved in ROS production was increased after irradiation but both were markedly reduced in NHKs by miR-200c inhibition. Knockdown of miR-200c expression in NHKs increased DNA double-strand break repair after irradiation compared with control NHKs. Furthermore, miR-200c inhibition repressed the production of proinflammatory cytokines (TGF-ß, TNF-α, and IL-1α) via inhibiting NF-κB and Smad2 activation in NHKs exposed to IR. Additionally, miR-200c inhibition promoted NHK migration and increased the expression of molecules that regulate epithelial to mesenchymal transition, including Snail, Vimentin, Zeb1, and Bmi-1. These results not only identify the key role of miR-200c in the pathogenesis of RIOM but also provide a novel therapeutic target to treat RIOM.


Sujet(s)
microARN/métabolisme , Stomatite/étiologie , Animaux , Mouvement cellulaire/effets des radiations , Prolifération cellulaire/effets des radiations , Vieillissement de la cellule/effets des radiations , Cytokines/biosynthèse , Réparation de l'ADN , Humains , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Kératinocytes/effets des radiations , Souris , microARN/génétique , Lésions radiques expérimentales/étiologie , Lésions radiques expérimentales/génétique , Lésions radiques expérimentales/métabolisme , Espèces réactives de l'oxygène/métabolisme , Stomatite/génétique , Stomatite/métabolisme , Stomatite/anatomopathologie
7.
Cancer Res ; 79(19): 5022-5033, 2019 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-31142513

RÉSUMÉ

Epithelial cells in the field of lung injury can give rise to distinct premalignant lesions that may bear unique genetic aberrations. A subset of these lesions may escape immune surveillance and progress to invasive cancer; however, the mutational landscape that may predict progression has not been determined. Knowledge of premalignant lesion composition and the associated microenvironment is critical for understanding tumorigenesis and the development of effective preventive and interception strategies. To identify somatic mutations and the extent of immune cell infiltration in adenomatous premalignancy and associated lung adenocarcinomas, we sequenced exomes from 41 lung cancer resection specimens, including 89 premalignant atypical adenomatous hyperplasia lesions, 15 adenocarcinomas in situ, and 55 invasive adenocarcinomas and their adjacent normal lung tissues. We defined nonsynonymous somatic mutations occurring in both premalignancy and the associated tumor as progression-associated mutations whose predicted neoantigens were highly correlated with infiltration of CD8+ and CD4+ T cells as well as upregulation of PD-L1 in premalignant lesions, suggesting the presence of an adaptive immune response to these neoantigens. Each patient had a unique repertoire of somatic mutations and associated neoantigens. Collectively, these results provide evidence for mutational heterogeneity, pathway dysregulation, and immune recognition in pulmonary premalignancy.Significance: These findings identify progression-associated somatic mutations, oncogenic pathways, and association between the mutational landscape and adaptive immune responses in adenomatous premalignancy.See related commentary by Merrick, p. 4811.


Sujet(s)
Adénocarcinome , Adénomes , Tumeurs du poumon , États précancéreux , Génomique , Humains , Microenvironnement tumoral
8.
Cancer Res ; 78(8): 1986-1999, 2018 04 15.
Article de Anglais | MEDLINE | ID: mdl-29431637

RÉSUMÉ

Epithelial-to-mesenchymal transition (EMT) is organized in cancer cells by a set of key transcription factors, but the significance of this process is still debated, including in non-small cell lung cancer (NSCLC). Here, we report increased expression of the EMT-inducing transcription factor Snail in premalignant pulmonary lesions, relative to histologically normal pulmonary epithelium. In immortalized human pulmonary epithelial cells and isogenic derivatives, we documented Snail-dependent anchorage-independent growth in vitro and primary tumor growth and metastatic behavior in vivo Snail-mediated transformation relied upon silencing of the tumor-suppressive RNA splicing regulatory protein ESRP1. In clinical specimens of NSCLC, ESRP1 loss was documented in Snail-expressing premalignant pulmonary lesions. Mechanistic investigations showed that Snail drives malignant progression in an ALDH+CD44+CD24- pulmonary stem cell subset in which ESRP1 and stemness-repressing microRNAs are inhibited. Collectively, our results show how ESRP1 loss is a critical event in lung carcinogenesis, and they identify new candidate directions for targeted therapy of NSCLC.Significance: This study defines a Snail-ESRP1 cancer axis that is crucial for human lung carcinogenesis, with implications for new intervention strategies and translational opportunities. Cancer Res; 78(8); 1986-99. ©2018 AACR.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Extinction de l'expression des gènes , Poumon/anatomopathologie , Protéines de liaison à l'ARN/génétique , Facteurs de transcription de la famille Snail/physiologie , Animaux , Lignée de cellules transformées , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Humains , Poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Modèles animaux
9.
Mol Nutr Food Res ; 62(6): e1700844, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29418071

RÉSUMÉ

SCOPE: Short-chain fatty acid sodium butyrate (NaB) is the byproduct of bacterial anaerobic fermentation of dietary fiber in the colon, and has been shown to have an antitumor effect on colorectal cancer (CRC). The miR-200 family is a key regulator of the epithelial-mesenchymal transition (EMT). We investigate the role of miR-200s expression on cell migration in NaB-treated CRC cells. METHODS AND RESULTS: HCT116 and LOVO CRC cells treated with NaB depicted reduced cell proliferation, enhanced apoptosis, and cell cycle arrest. NaB inhibited cell migration in the wound healing and transwell assays, and in spheriod cultures while regulating EMT-related protein expression. NaB reciprocally increased miR-200s but reduced expression of their target genes (Bmi-1, Zeb1, EZH2). Cells transfected with miR-200c shRNA displayed a significant blockade of NaB-induced anti-invasive activity. Upregulation of Bmi-1 expression partially reversed the effect of NaB. In addition to inhibition of tumor growth in vivo, qRT-PCR results showed that NaB increased miR-200c/200b/492 expression in the tumor tissues. Immunohistochemistry and Western blotting results demonstrated that NaB decreased Bmi-1 expression in vivo. CONCLUSION: NaB inhibits CRC cell migration by enhancing miR-200c expression-mediated downregulation of Bmi-1. These findings support the utility of NaB in colorectal cancer therapy.


Sujet(s)
Acide butyrique/pharmacologie , Tumeurs colorectales/traitement médicamenteux , microARN/physiologie , Complexe répresseur Polycomb-1/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Acide butyrique/usage thérapeutique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/anatomopathologie , Régulation négative , Femelle , Cellules HCT116 , Humains , Souris , Facteur de transcription Zeb1/génétique
10.
Otolaryngol Head Neck Surg ; 158(1): 110-117, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-28895464

RÉSUMÉ

Objective To evaluate the adverse effects and therapeutic efficacy of our biocompatible polymer platform delivering targeted local therapy of cytokine CCL21 and cisplatin in a partially resected xenograft animal model of head and neck squamous cell carcinoma. In addition, to evaluate the efficacy of cotreatment with radiotherapy and assess the biocompatibility of the cisplatin-eluting polymer in the murine neck. Study Design Experimental animal study. Setting Academic research laboratory. Subjects and Methods SCCVII/SF cell injection established head and neck squamous cell carcinoma tumors in C3H/HeJ mice. Subjects underwent surgery, and a chemokine-eluting polymer was implanted into the resected site. Subjects treated with cisplatin received radiation or no radiation, and tissue was harvested after 8 weeks to assess polymer biocompatibility. Results Our results with the polymer platform significantly ( P < .05) reduced SCCVII/SF tumor size in C3H/HeJ mice with cisplatin (49% ± 8.7%, Δ3.4 ± 0.6 cm3 [95% CI]), CCL21 (42% ± 4.8%, Δ3.5 ± 0.4 cm3), and cisplatin/CCL21 dual-agent polymer (82% ± 4.4%, Δ8.0 ± 0.4 cm3) as compared with controls. Cisplatin polymer with high-dose (16 Gy) and low-dose (4 Gy) radiation reduced tumor mass (respectively, 92% ± 7.2%, Δ6.1 ± 0.5 cm3; 85% ± 7.4%, Δ5.7 ± 0.5 cm3) as compared with the reduction from high-dose radiotherapy alone (70% ± 7.9%, Δ4.7 ± 0.5 cm3). No significant toxicity or inflammation was noted on histopathology after radiotherapy and cisplatin-eluting polymer treatment. Conclusion Cisplatin, CCL21, and cisplatin/CCL21 dual-agent polymer all exhibit significant antitumor effects and decrease tumor burden. Moreover, combination cisplatin polymer with radiotherapy may permit a decrease in intensity of radiation therapy in patients having received the cisplatin polymer. Histopathologic analysis suggests that the polymer is free from significant adverse effects in this model and warrants clinical trial.


Sujet(s)
Carcinome épidermoïde/traitement médicamenteux , Chimiokine CCL21/administration et posologie , Cisplatine/administration et posologie , Systèmes de délivrance de médicaments , Tumeurs de la tête et du cou/traitement médicamenteux , Polymères/pharmacologie , Animaux , Matériaux biocompatibles/pharmacologie , Modèles animaux de maladie humaine , Test ELISA , Souris , Polyesters/pharmacologie
11.
BMC Cancer ; 17(1): 805, 2017 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-29191172

RÉSUMÉ

BACKGROUND: MicroRNAs (miRNAs) exhibit essential regulatory functions related to cell growth, apoptosis, development and differentiation. Dysregulated expression of miRNAs is associated with a wide variety of human diseases. As such miRNA signatures are valuable as biomarkers for disease and for making treatment decisions. Hepatitis B virus (HBV) is a major risk factor for hepatocellular carcinoma (HCC). Here we screened for miRNAs in chronic HBV associated HCC. METHODS: To determine the miRNAs in HCC occurrence associated with HBV infection, we analyzed global miRNA expression profiles in 12 pairs of HCC and adjacent matched non-HCC tissues from HBV-positive and HBV-negative patients using microarray analyses. The microarray result was validated by real-time PCR in 32 HBV-positive and 24 HBV-negative patient HCC samples. The potential candidate target genes of the miRNAs were predicted by miRWalk software. Genes simultaneously predicted as targets by two or more miRNAs were subjected to GO and KEGG pathway analysis. The miRNA regulatory network analysis was performed using the Ingenuity Pathway Analysis (IPA) software. RESULTS: Eight miRNAs (miR-223, miR-98, miR-15b, miR-199a-5p, miR-19b, miR-22, miR-451, and miR-101) were involved in HBV-unrelated HCC, 5 miRNAs (miR-98, miR-375, miR-335, miR-199a-5p, and miR-22) were involved in HBV infection, and 7 miRNAs (miR-150, miR-342-3p, miR-663, miR-20b, miR-92a-3p, miR-376c-3p and miR-92b) were specifically altered in HBV-related HCC. Gene Ontology and KEGG analyses predict that these HBV-related HCC miRNAs are involved in the regulation of: transcription, RNA polymerase II promoter, phosphorylation of proteins through MAPK signaling pathway, focal adhesion, and actin cytoskeleton. IPA analysis also suggest that these miRNAs act on AGO2, TP53, CCND1, and 11 other genes that significantly influence HCC occurrence and HBV infection. CONCLUSION: Our data indicates that the unique 7 miRNAs expression signature could be involved in the development HBV- related HCC.


Sujet(s)
Carcinome hépatocellulaire/génétique , Analyse de profil d'expression de gènes/méthodes , Hépatite B/génétique , Tumeurs du foie/génétique , microARN/génétique , Carcinome hépatocellulaire/virologie , Biologie informatique , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Humains , Tumeurs du foie/virologie , Séquençage par oligonucléotides en batterie , Logiciel
12.
Sci Rep ; 7(1): 9192, 2017 08 23.
Article de Anglais | MEDLINE | ID: mdl-28835699

RÉSUMÉ

Casitas B-lineage lymphoma (CBL) is an E3 ubiquitin ligase and a molecule of adaptor that we have shown is important for non-small-cell lung cancer (NSCLC). We investigated if MET is a target of CBL and if enhanced in CBL-altered NSCLC. We showed that CBL wildtype cells have lower MET expression than CBL mutant cells. Ubiquitination of MET was also decreased in CBL mutant cells compared to wildtype cells. Mutant cells were also more sensitive to MET inhibitor SU11274 than wild-type cells. sh-RNA-mediated knockdown of CBL enhanced cell motility and colony formation in NSCLC cells, and these activities were inhibited by SU11274. Assessment of the phospho-kinome showed decreased phosphorylation of pathways involving MET, paxillin, EPHA2, and VEGFR. When CBL was knocked down in the mutant cell line H1975 (erlotinib-resistant), it became sensitive to MET inhibition. Our findings suggest that CBL status is a potential positive indicator for MET-targeted therapeutics in NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines proto-oncogènes c-cbl/génétique , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs ErbB/génétique , Expression des gènes , Techniques de knock-down de gènes , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Souris , Mutation , Métastase tumorale , Phosphorylation , Inhibiteurs de protéines kinases/pharmacologie , Protéolyse , Protéines proto-oncogènes c-cbl/métabolisme , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Interférence par ARN , Petit ARN interférent/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Clin Cancer Res ; 23(16): 4556-4568, 2017 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-28468947

RÉSUMÉ

Purpose: A phase I study was conducted to determine safety, clinical efficacy, and antitumor immune responses in patients with advanced non-small cell lung carcinoma (NSCLC) following intratumoral administration of autologous dendritic cells (DC) transduced with an adenoviral (Ad) vector expressing the CCL21 gene (Ad-CCL21-DC). We evaluated safety and tumor antigen-specific immune responses following in situ vaccination (ClinicalTrials.gov: NCT01574222).Experimental Design: Sixteen stage IIIB/IV NSCLC subjects received two vaccinations (1 × 106, 5 × 106, 1 × 107, or 3 × 107 DCs/injection) by CT- or bronchoscopic-guided intratumoral injections (days 0 and 7). Immune responses were assessed by tumor antigen-specific peripheral blood lymphocyte induction of IFNγ in ELISPOT assays. Tumor biopsies were evaluated for CD8+ T cells by IHC and for PD-L1 expression by IHC and real-time PCR (RT-PCR).Results: Twenty-five percent (4/16) of patients had stable disease at day 56. Median survival was 3.9 months. ELISPOT assays revealed 6 of 16 patients had systemic responses against tumor-associated antigens (TAA). Tumor CD8+ T-cell infiltration was induced in 54% of subjects (7/13; 3.4-fold average increase in the number of CD8+ T cells per mm2). Patients with increased CD8+ T cells following vaccination showed significantly increased PD-L1 mRNA expression.Conclusions: Intratumoral vaccination with Ad-CCL21-DC resulted in (i) induction of systemic tumor antigen-specific immune responses; (ii) enhanced tumor CD8+ T-cell infiltration; and (iii) increased tumor PD-L1 expression. Future studies will evaluate the role of combination therapies with PD-1/PD-L1 checkpoint inhibition combined with DC-CCL21 in situ vaccination. Clin Cancer Res; 23(16); 4556-68. ©2017 AACR.


Sujet(s)
Lymphocytes T CD8+/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Chimiokine CCL21/immunologie , Cellules dendritiques/immunologie , Immunothérapie adoptive/méthodes , Tumeurs du poumon/thérapie , Adulte , Sujet âgé , Antigène CD274/génétique , Antigène CD274/immunologie , Antigène CD274/métabolisme , Lymphocytes T CD8+/métabolisme , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/immunologie , Chimiokine CCL21/génétique , Études de cohortes , Cellules dendritiques/métabolisme , Cellules dendritiques/transplantation , Dyspnée/étiologie , Femelle , Humains , Immunothérapie adoptive/effets indésirables , Injections intralésionnelles , Interféron gamma/immunologie , Interféron gamma/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Mâle , Adulte d'âge moyen , Faiblesse musculaire/étiologie , Douleur/étiologie
14.
Oral Oncol ; 60: 81-9, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27531877

RÉSUMÉ

BACKGROUND: In the present study, we investigated the role of p38-p38IP signaling in the inflammation-induced promotion of epithelial-to-mesenchymal transition (EMT) in Head and Neck Squamous Cell Carcinoma (HNSCC). METHODS: Quantitative RT-PCR, western blot analysis, spheroid modeling and immunohistochemical staining of human HNSCC tissue sections were used. RESULTS: p38 inhibitor treated and p38 shRNA HNSCC cell lines demonstrate a significant upregulation in E-cadherin mRNA and a decrease in the mRNA expression of Snail. p38 binds to and stabilizes p38IP, a subunit of histone SPT3-TAF9-GCN5 acetyltransferase (STAGA), resulting in enhanced transcription of Snail. p38 shRNA HNSCC cell lines show a less invasive phenotype in a spheroid model. In clinical HNSCC samples, p38 interacting protein (p38IP) is significantly increased compared to adjacent normal tissue. An inverse relationship between p38, p38IP and E-cadherin is demonstrated. CONCLUSIONS: Herein we provide the first report that p38-p38IP is required for the Snail-induced E-cadherin down-regulation and cell invasion in HNSCC.


Sujet(s)
Carcinome épidermoïde/anatomopathologie , Transition épithélio-mésenchymateuse , Tumeurs de la tête et du cou/anatomopathologie , Facteurs de transcription de la famille Snail/métabolisme , Facteurs de transcription/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Cadhérines/métabolisme , Carcinome épidermoïde/enzymologie , Carcinome épidermoïde/métabolisme , Lignée cellulaire tumorale , Tumeurs de la tête et du cou/enzymologie , Tumeurs de la tête et du cou/métabolisme , Humains , Invasion tumorale , Métastase tumorale , Facteurs de transcription de la famille Snail/génétique , Carcinome épidermoïde de la tête et du cou , Facteurs de transcription/isolement et purification , p38 Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs
15.
Oral Oncol ; 51(10): 921-8, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26306422

RÉSUMÉ

OBJECTIVES: To investigate the role of p53 in NF-κB mediated epithelial-to-mesenchymal (EMT) in head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS: We utilized HNSCC and normal oral epithelial cell lines as our model system. We used a lentiviral shRNA system to silence the expression of p65 and p53 in these cell lines. Mutant and wild-type (WT) p53 background genotypes were analyzed. The expression of epithelial and mesenchymal markers was determined using western blotting and quantitative PCR assays. Cell morphology, growth, and invasion were determined using a 3-dimensional spheroid culture and anchorage independent growth (AIG) assays. RESULTS: In HNSCC cells with mutant p53 we found that silencing p65 expression promoted EMT. In contrast, in the context of WT p53, ectopic p65 over-expression promoted EMT. Ablation of WT p53 in normal oral epithelial cells blocked EMT induced by p65 over-expression. We demonstrate that AIG and apoptosis induced by NF-κB activation is regulated by p53. CONCLUSION: Our data demonstrates that p53 mutational status is critical in determining the outcome of NF-κB activation in HNSCC. In the presence of WT p53, excess p65 signal can promote EMT. Conversely, ablation of p65 in the context of mutant p53 drives EMT. These results demonstrate that p53 mutational status alters the outcome of NF-κB signaling. These results, though preliminary, demonstrate the critical role of p53 mutational status in determining the outcome of NF-κB signaling and suggest that monitoring p53 status may inform the utility of NF-κB inhibitor treatment in HNSCC.


Sujet(s)
Carcinome épidermoïde/anatomopathologie , Transition épithélio-mésenchymateuse/physiologie , Tumeurs de la tête et du cou/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Protéine p53 suppresseur de tumeur/physiologie , Apoptose , Marqueurs biologiques tumoraux/métabolisme , Carcinome épidermoïde/métabolisme , Lignée cellulaire tumorale , Tumeurs de la tête et du cou/métabolisme , Humains
16.
Oncoimmunology ; 4(4): e992237, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-26137407

RÉSUMÉ

An immune tolerant tumor microenvironment promotes immune evasion of lung cancer. Agents that antagonize immune tolerance will thus aid the fight against this devastating disease. Members of the tumor necrosis factor receptor (TNFR) family modulate the magnitude, duration and phenotype of immune responsiveness to antigens. Among these, GITR expressed on immune cells functions as a key regulator in inflammatory and immune responses. Here, we evaluate the GITR agonistic antibody (DTA-1) as a mono-therapy and in combination with therapeutic vaccination in murine lung cancer models. We found that DTA-1 treatment of tumor-bearing mice increased: (i) the frequency and activation of intratumoral natural killer (NK) cells and T lymphocytes, (ii) the antigen presenting cell (APC) activity in the tumor, and (iii) systemic T-cell specific tumor cell cytolysis. DTA-1 treatment enhanced tumor cell apoptosis as quantified by cleaved caspase-3 staining in the tumors. DTA-1 treatment increased expression of IFNγ, TNFα and IL-12 but reduced IL-10 levels in tumors. Furthermore, increased anti-angiogenic chemokines corresponding with decreased pro-angiogenic chemokine levels correlated with reduced expression of the endothelial cell marker Meca 32 in the tumors of DTA-1 treated mice. In accordance, there was reduced tumor growth (8-fold by weight) in the DTA-1 treatment group. NK cell depletion markedly inhibited the antitumor response elicited by DTA-1. DTA-1 combined with therapeutic vaccination caused tumor rejection in 38% of mice and a 20-fold reduction in tumor burden in the remaining mice relative to control. Mice that rejected tumors following therapy developed immunological memory against subsequent re-challenge. Our data demonstrates GITR agonist antibody activated NK cell and T lymphocyte activity, and enhanced therapeutic vaccination responses against lung cancer.

17.
Autoimmun Rev ; 13(12): 1174-81, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25151974

RÉSUMÉ

T helper 17 (Th17), a distinct subset of CD4(+) T cells with IL-17 as their major cytokine, orchestrate the pathogenesis of inflammatory and autoimmune diseases. Dysregulated Th17 cells contribute to inflammatory and autoimmune diseases. Candidate biologics are in development for targeting IL-17, IL-17 receptors or IL-17 pathways. Several drugs that impact the IL-17 pathway are already in clinical trials for the treatment of autoimmune diseases. In this review we provide evidence for the role of Th17 cells in immune-mediated diseases. An understanding of the role of Th17 in these conditions will provide important insights and unravel novel targets for therapeutic intervention.


Sujet(s)
Auto-immunité , Cellules Th17/immunologie , Animaux , Maladies auto-immunes/immunologie , Hormones sexuelles stéroïdiennes/immunologie , Humains , Inflammation/immunologie , Interleukine-17/immunologie , Cellules souches mésenchymateuses/immunologie
18.
Br J Pharmacol ; 171(19): 4440-54, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24902966

RÉSUMÉ

BACKGROUND AND PURPOSE: Doxorubicin-based chemotherapy induces cardiotoxicity, which limits its clinical application. We previously reported the protective effects of quercetin against doxorubicin-induced hepatotoxicity. In this study, we tested the effects of quercetin on the expression of Bmi-1, a protein regulating mitochondrial function and ROS generation, as a mechanism underlying quercetin-mediated protection against doxorubicin-induced cardiotoxicity. EXPERIMENTAL APPROACH: Effects of quercetin on doxorubicin-induced cardiotoxicity was evaluated using H9c2 cardiomyocytes and C57BL/6 mice. Changes in apoptosis, mitochondrial function, oxidative stress and related signalling were evaluated in H9c2 cells. Cardiac function, serum enzyme activity and reactive oxygen species (ROS) generation were measured in mice after a single injection of doxorubicin with or without quercetin pre-treatment. KEY RESULTS: In H9c2 cells, quercetin reduced doxorubicin-induced apoptosis, mitochondrial dysfunction, ROS generation and DNA double-strand breaks. The quercetin-mediated protection against doxorubicin toxicity was characterized by decreased expression of Bid, p53 and oxidase (p47 and Nox1) and by increased expression of Bcl-2 and Bmi-1. Bmi-1 siRNA abolished the protective effect of quercetin against doxorubicin-induced toxicity in H9c2 cells. Furthermore, quercetin protected mice from doxorubicin-induced cardiac dysfunction that was accompanied by reduced ROS levels and lipid peroxidation, but enhanced the expression of Bmi-1 and anti-oxidative superoxide dismutase. CONCLUSIONS AND IMPLICATIONS: Our results demonstrate that quercetin decreased doxorubicin-induced cardiotoxicity in vitro and in vivo by reducing oxidative stress by up-regulation of Bmi-1 expression. The findings presented in this study have potential applications in preventing doxorubicin-induced cardiomyopathy.


Sujet(s)
Antioxydants/pharmacologie , Cardiotoniques/pharmacologie , Cardiotoxicité/métabolisme , Complexe répresseur Polycomb-1/métabolisme , Protéines proto-oncogènes/métabolisme , Quercétine/pharmacologie , Animaux , Antibiotiques antinéoplasiques , Apoptose/effets des médicaments et des substances chimiques , Cardiotoxicité/anatomopathologie , Lignée cellulaire , Doxorubicine , Femelle , Peroxydation lipidique/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/physiologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Complexe répresseur Polycomb-1/génétique , Protéines proto-oncogènes/génétique , Rats , Espèces réactives de l'oxygène/métabolisme
19.
Cancers (Basel) ; 6(2): 1098-110, 2014 May 07.
Article de Anglais | MEDLINE | ID: mdl-24810425

RÉSUMÉ

Cancer, a major health problem, affects 12 million people worldwide every year. With surgery and chemo-radiation the long term survival rate for the majority of cancer patients is dismal. Thus novel treatments are urgently needed. Immunotherapy, the harnessing of the immune system to destroy cancer cells is an attractive option with potential for long term anti-tumor benefit. Cytokines are biological response modifiers that stimulate anti-tumor immune responses. In this review, we discuss the anti-tumor efficacy of the chemotactic cytokine CCL21 and its pre-clinical and clinical application in cancer.

20.
Otolaryngol Head Neck Surg ; 151(3): 447-53, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-24825873

RÉSUMÉ

OBJECTIVE: This study aimed to evaluate the therapeutic efficacy of a novel polymer platform delivering cisplatin and cytokines in the treatment of head and neck squamous cell carcinoma (HNSCC). STUDY DESIGN: In vivo study. SETTING: Academic research laboratory. SUBJECTS AND METHODS: Mice were randomized to receive implantation of (1) no polymer, (2) plain polymer, (3) plain polymer with local cisplatin injection, or (4) cisplatin polymer. The 2 groups of mice implanted with cisplatin polymer or no polymer were further randomized to receive (1) 4 Grays external beam radiation for 4 days or (2) no radiation. For cytokine studies, mice were grouped into (1) no polymer, (2) plain polymer, (3) plain polymer with intratumoral injection of recombinant CCL21 twice a week, (4) polymer containing parental dendritic cells, or (5) polymer containing dendritic cells secreting CCL21 (DC-CCL21). RESULTS: The cisplatin-secreting polymer effectively reduced tumors in the mice by more than 16-fold (P < .01). We also observed a statistically significant lower tumor weight among mice treated with cisplatin polymer and concomitant radiation compared to control groups. The DC-CCL21 polymer reduced SCCVII/SF tumors in the C3H/HeJ mice by more than 41% (P < .01). CONCLUSION: Herein, we demonstrate the efficacy of a novel polymer platform in delivering cisplatin and cytokines. We also demonstrate that we can effectively grow dendritic cells in the polymer that can actively secrete CCL21 for a minimum of 5 days. This polymer may represent a new therapeutic modality for patients with HNSCC. Once this polymer platform is optimized, we will plan to pursue prospective trials in patients with HNSCC.


Sujet(s)
Carcinome épidermoïde/traitement médicamenteux , Cisplatine/administration et posologie , Cytokines/pharmacologie , Implant pharmaceutique , Tumeurs de la tête et du cou/traitement médicamenteux , Polymères/pharmacologie , Analyse de variance , Animaux , Technique de Western , Carcinome épidermoïde/mortalité , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/radiothérapie , Modèles animaux de maladie humaine , Systèmes de délivrance de médicaments , Test ELISA , Femelle , Cytométrie en flux , Tumeurs de la tête et du cou/mortalité , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/radiothérapie , Immunohistochimie , Mâle , Souris , Souris de lignée C3H , Répartition aléatoire , Taux de survie , Résultat thérapeutique , Charge tumorale/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...