Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 64
Filtrer
1.
Front Immunol ; 15: 1256766, 2024.
Article de Anglais | MEDLINE | ID: mdl-38487537

RÉSUMÉ

CD5 is a member of the scavenger receptor cysteine-rich superfamily that is expressed on T cells and a subset of B cells (B1a) cell and can regulate the T cell receptor signaling pathway. Blocking CD5 function may have therapeutic potential in treatment of cancer by enhancing cytotoxic T lymphocyte recognition and ablation of tumour cells. The effect of administering an anti-CD5 antibody to block or reduce CD5 function as an immune checkpoint blockade to enhance T cell anti-tumour activation and function in vivo has not been explored. Here we challenged mice with poorly immunogenic 4T1 breast tumour cells and tested whether treatment with anti-CD5 monoclonal antibodies (MAb) in vivo could enhance non-malignant T cell anti-tumour immunity and reduce tumour growth. Treatment with anti-CD5 MAb resulted in an increased fraction of CD8+ T cells compared to CD4+ T cell in draining lymph nodes and the tumour microenvironment. In addition, it increased activation and effector function of T cells isolated from spleens, draining lymph nodes, and 4T1 tumours. Furthermore, tumour growth was delayed in mice treated with anti-CD5 MAb. These data suggest that use of anti-CD5 MAb as an immune checkpoint blockade can both enhance activation of T cells in response to poorly immunogenic antigens and reduce tumour growth in vivo. Exploration of anti-CD5 therapies in treatment of cancer, alone and in combination with other immune therapeutic drugs, is warranted.


Sujet(s)
Tumeurs du sein , Lymphocytes T CD8+ , Souris , Animaux , Humains , Femelle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Lymphocytes T cytotoxiques , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Allogreffes , Microenvironnement tumoral
2.
Mol Ther ; 31(2): 535-551, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36068918

RÉSUMÉ

Immune checkpoint blockade can induce potent and durable responses in patients with highly immunogenic mismatch repair-deficient tumors; however, these drugs are ineffective against immune-cold neuroblastoma tumors. To establish a role for a T cell-based therapy against neuroblastoma, we show that T cell and memory T cell-dependent gene expression are associated with improved survival in high-risk neuroblastoma patients. To stimulate anti-tumor immunity and reproduce this immune phenotype in neuroblastoma tumors, we used CRISPR-Cas9 to knockout MLH1-a crucial molecule in the DNA mismatch repair pathway-to induce mismatch repair deficiency in a poorly immunogenic murine neuroblastoma model. Induced mismatch repair deficiency increased the expression of proinflammatory genes and stimulated T cell infiltration into neuroblastoma tumors. In contrast to adult cancers with induced mismatch repair deficiency, neuroblastoma tumors remained unresponsive to anti-PD1 treatment. However, anti-CTLA4 therapy was highly effective against these tumors. Anti-CTLA4 therapy promoted immune memory and T cell epitope spreading in cured animals. Mechanistically, the effect of anti-CTLA4 therapy against neuroblastoma tumors with induced mismatch repair deficiency is CD4+ T cell dependent, as depletion of these cells abolished the effect. Therefore, a therapeutic strategy involving mismatch repair deficiency-based T cell infiltration of neuroblastoma tumors combined with anti-CTLA4 can serve as a novel T cell-based treatment strategy for neuroblastoma.


Sujet(s)
Tumeurs du cerveau , Tumeurs colorectales , Neuroblastome , Souris , Animaux , Mémoire immunologique , Tumeurs colorectales/anatomopathologie , Neuroblastome/génétique , Neuroblastome/thérapie
3.
Int J Mol Sci ; 23(1)2021 12 27.
Article de Anglais | MEDLINE | ID: mdl-35008676

RÉSUMÉ

We have previously found that TdT-interacting factor 1 (TdIF1) is a potential oncogene expressed in non-small cell lung cancer (NSCLC) and is associated with poor prognosis. However, its exact mechanism is still unclear. The lysine-specific demethylase 1 (LSD1) is a crucial mediator of the epithelial-mesenchymal transition (EMT), an important process triggered during cancer metastasis. Here, we confirm that TdIF1 is highly expressed in NSCLC and related to lymph node metastasis through The Cancer Genome Atlas (TCGA) analysis of clinical samples. Silencing TdIF1 can regulate the expression of EMT-related factors and impair the migration and invasion ability of cancer cells in vitro. An analysis of tumor xenografts in nude mice confirmed that silencing TdIF1 inhibits tumor growth. Furthermore, we determined the interaction between TdIF1 and LSD1 using immunoprecipitation. Chromatin immunoprecipitation (ChIP) revealed that TdIF1 was enriched in the E-cadherin promoter region. The knockdown of TdIF1 repressed the enrichment of LSD1 at the E-cadherin promoter region, thereby regulating the level of promoter histone methylation and modulating E-cadherin transcription activity, ultimately leading to changes in EMT factors and cancer cell migration and invasion ability. The LSD1 inhibitor and TdIF1 knockdown combination showed a synergistic effect in inhibiting the growth, migration, and invasion of NSCLC cells. Taken together, this is the first demonstration that TdIF1 regulates E-cadherin transcription by recruiting LSD1 to the promoter region, thereby promoting EMT and tumor metastasis and highlighting the potential of TdIF1 as a therapeutic target for NSCLC.


Sujet(s)
Cadhérines/génétique , Protéines de liaison à l'ADN/métabolisme , Déméthylation , Transition épithélio-mésenchymateuse/génétique , Histone Demethylases/métabolisme , Histone/métabolisme , Tumeurs du poumon/génétique , Régions promotrices (génétique) , Facteurs de transcription/métabolisme , Animaux , Cadhérines/métabolisme , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation négative/génétique , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Tumeurs du poumon/anatomopathologie , Lysine/métabolisme , Méthylation , Souris de lignée BALB C , Souris nude , Invasion tumorale , Métastase tumorale , Liaison aux protéines
4.
Methods Enzymol ; 636: 49-76, 2020.
Article de Anglais | MEDLINE | ID: mdl-32178827

RÉSUMÉ

The importance of the immune system in combating many different types of malignancies has been firmly established. Indeed, the success of immune checkpoint-blocking antibodies in cancer treatment has focused the attention of many researchers on the tumor immune microenvironment. As one consequence, the analysis of the number, type, and activation status of tumor-infiltrating leukocytes (TILs) has become an area of great interest. Detailed knowledge of the immunological profile of a given tumor has the potential to predict patient response to therapy and overall survival. This chapter is intended to summarize the steps necessary for basic comparative analysis of the infiltration and activation status of immune cells in various human solid tumors, using freely available data obtained from The Cancer Genome Atlas (TCGA).


Sujet(s)
Tumeurs , Transcriptome , Épigénome , Humains , Lymphocytes TIL , Tumeurs/génétique , Microenvironnement tumoral/génétique
5.
Cancer Immunol Immunother ; 69(6): 951-967, 2020 Jun.
Article de Anglais | MEDLINE | ID: mdl-32076794

RÉSUMÉ

Dendritic cell (DC) based immunotherapy is a promising approach to clinical cancer treatment. miRNAs are a class of small non-coding RNA molecules that bind to RNAs to mediate multiple events which are important in diverse biological processes. miRNA mimics and antagomirs may be potent agents to enhance DC-based immunotherapy against cancers. miRNA array analysis was used to identify a representative miR-5119 potentially regulating PD-L1 in DCs. We evaluated levels of ligands of immune cell inhibitory receptors (IRs) and miR-5119 in DCs from immunocompetent mouse breast tumor-bearing mice, and examined the molecular targets of miR-5119. We report that miRNA-5119 was downregulated in spleen DCs from mouse breast cancer-bearing mice. In silico analysis and qPCR data showed that miRNA-5119 targeted mRNAs encoding multiple negative immune regulatory molecules, including ligands of IRs such as PD-L1 and IDO2. DCs engineered to express a miR-5119 mimic downregulated PD-L1 and prevented T cell exhaustion in mice with breast cancer homografts. Moreover, miR-5119 mimic-engineered DCs effectively restored function to exhausted CD8+ T cells in vitro and in vivo, resulting in robust anti-tumor cell immune response, upregulated cytokine production, reduced T cell apoptosis, and exhaustion. Treatment of 4T1 breast tumor-bearing mice with miR-5119 mimic-engineered DC vaccine reduced T cell exhaustion and suppressed mouse breast tumor homograft growth. This study provides evidence supporting a novel therapeutic approach using miRNA-5119 mimic-engineered DC vaccines to regulate inhibitory receptors and enhance anti-tumor immune response in a mouse model of breast cancer. miRNA/DC-based immunotherapy has potential for advancement to the clinic as a new strategy for DC-based anti-breast cancer immunotherapy.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Vaccins anticancéreux/immunologie , Cellules dendritiques/immunologie , Immunothérapie/méthodes , microARN/métabolisme , Animaux , Tumeurs du sein/immunologie , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Transfection
6.
Eur J Immunol ; 50(5): 695-704, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-31943150

RÉSUMÉ

CD5 is expressed on T cells and a subset of B cells (B1a). It can attenuate TCR signalling and impair CTL activation and is a therapeutic targetable tumour antigen expressed on leukemic T and B cells. However, the potential therapeutic effect of functionally blocking CD5 to increase T cell anti-tumour activity against tumours (including solid tumours) has not been explored. CD5 knockout mice show increased anti-tumour immunity: reducing CD5 on CTLs may be therapeutically beneficial to enhance the anti-tumour response. Here, we show that ex vivo administration of a function-blocking anti-CD5 MAb to primary mouse CTLs of both tumour-naïve mice and mice bearing murine 4T1 breast tumour homografts enhanced their capacity to respond to activation by treatment with anti-CD3/anti-CD28 MAbs or 4T1 tumour cell lysates. Furthermore, it enhanced TCR signalling (ERK activation) and increased markers of T cell activation, including proliferation, CD69 levels, IFN-γ production, apoptosis and Fas receptor and Fas ligand levels. Finally, CD5 function-blocking MAb treatment enhanced the capacity of CD8+ T cells to kill 4T1-mouse tumour cells in an ex vivo assay. These data support the potential of blockade of CD5 function to enhance T cell-mediated anti-tumour immunity.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Anticorps neutralisants/pharmacologie , Antigène CD28/immunologie , Antigènes CD5/immunologie , Tumeurs expérimentales de la mamelle/traitement médicamenteux , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques immunologiques , Antigène CD28/antagonistes et inhibiteurs , Antigène CD28/génétique , Antigènes CD5/antagonistes et inhibiteurs , Antigènes CD5/génétique , Extrait cellulaire/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire , Ligand de Fas/génétique , Ligand de Fas/immunologie , Femelle , Régulation de l'expression des gènes , Interféron gamma/génétique , Interféron gamma/immunologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Glandes mammaires animales/effets des médicaments et des substances chimiques , Glandes mammaires animales/immunologie , Glandes mammaires animales/anatomopathologie , Tumeurs expérimentales de la mamelle/génétique , Tumeurs expérimentales de la mamelle/immunologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris knockout , Transduction du signal , Lymphocytes T cytotoxiques/immunologie , Antigènes CD95/génétique , Antigènes CD95/immunologie
7.
Front Immunol ; 11: 584937, 2020.
Article de Anglais | MEDLINE | ID: mdl-33584650

RÉSUMÉ

CD5, a member of the scavenger receptor cysteine-rich superfamily, is a marker for T cells and a subset of B cells (B1a). CD5 associates with T-cell and B-cell receptors and increased CD5 is an indication of B cell activation. In tumor-infiltrating lymphocytes (TILs) isolated from lung cancer patients, CD5 levels were negatively correlated with anti-tumor activity and tumor-mediated activation-induced T cell death, suggesting that CD5 could impair activation of anti-tumor T cells. We determined CD5 levels in T cell subsets in different organs in mice bearing syngeneic 4T1 breast tumor homografts and assessed the relationship between CD5 and increased T cell activation and effector function by flow cytometry. We report that T cell CD5 levels were higher in CD4+ T cells than in CD8+ T cells in 4T1 tumor-bearing mice, and that high CD5 levels on CD4+ T cells were maintained in peripheral organs (spleen and lymph nodes). However, both CD4+ and CD8+ T cells recruited to tumors had reduced CD5 compared to CD4+ and CD8+ T cells in peripheral organs. In addition, CD5high/CD4+ T cells and CD5high/CD8+ T cells from peripheral organs exhibited higher levels of activation and associated effector function compared to CD5low/CD4+ T cell and CD5low/CD8+ T cell from the same organs. Interestingly, CD8+ T cells among TILs and downregulated CD5 were activated to a higher level, with concomitantly increased effector function markers, than CD8+/CD5high TILs. Thus, differential CD5 levels among T cells in tumors and lymphoid organs can be associated with different levels of T cell activation and effector function, suggesting that CD5 may be a therapeutic target for immunotherapeutic activation in cancer therapy.


Sujet(s)
Antigènes CD5/immunologie , Lymphocytes T CD8+/immunologie , Tumeurs du poumon/immunologie , Activation des lymphocytes/immunologie , Animaux , Femelle , Lymphocytes TIL/immunologie , Souris , Souris de lignée BALB C , Sous-populations de lymphocytes T/immunologie
8.
Open Biol ; 9(10): 190061, 2019 10 31.
Article de Anglais | MEDLINE | ID: mdl-31594465

RÉSUMÉ

Blockade of inhibitory receptors (IRs) is one of the most effective immunotherapeutic approaches to treat cancer. Dysfunction of miRNAs is a major cause of aberrant expression of IRs and contributes to the immune escape of cancer cells. How miRNAs regulate immune checkpoint proteins in breast cancer remains largely unknown. In this study, downregulation of miRNAs was observed in PD-1-overexpressing CD8+ T cells using miRNA array analysis of mouse breast cancer homografts. The data reveal that miR-149-3p was predicted to bind the 3'UTRs of mRNAs encoding T-cell inhibitor receptors PD-1, TIM-3, BTLA and Foxp1. Treatment of CD8+ T cells with an miR-149-3p mimic reduced apoptosis, attenuated changes in mRNA markers of T-cell exhaustion and downregulated mRNAs encoding PD-1, TIM-3, BTLA and Foxp1. On the other hand, T-cell proliferation and secretion of effector cytokines indicative of increased T-cell activation (IL-2, TNF-α, IFN-γ) were upregulated after miR-149-3p mimic treatment. Moreover, the treatment with a miR-149-3p mimic promoted the capacity of CD8+ T cells to kill targeted 4T1 mouse breast tumour cells. Collectively, these data show that miR-149-3p can reverse CD8+ T-cell exhaustion and reveal it to be a potential antitumour immunotherapeutic agent in breast cancer.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Cytokines/métabolisme , Tumeurs expérimentales de la mamelle/génétique , microARN/métabolisme , Régions 3' non traduites , Animaux , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Cellules cultivées , Cytokines/génétique , Femelle , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Immunothérapie/méthodes , Tumeurs expérimentales de la mamelle/métabolisme , Tumeurs expérimentales de la mamelle/thérapie , Souris , Souris de lignée BALB C , microARN/génétique , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme
9.
Oncoimmunology ; 7(10): e1498439, 2018.
Article de Anglais | MEDLINE | ID: mdl-30288365

RÉSUMÉ

Cancers progress when the immune system fails to identify and eliminate malignant cells. Recognition of this, combined with advances in tumor immunology, has allowed development of therapies that induce effective anti-tumor immune responses. For incompletely-understood reasons, effective responses to immunotherapy occur in some patients and not others. Head and neck squamous cell carcinomas (HNSCC) are a common cancer type that can be divided into two subsets based on human papillomavirus (HPV) status. HPV status is a strong predictor of positive clinical outcome. Expression of exogenous viral antigens by HPV+, but not HPV-, HNSCC allows direct comparison of the immune status (immune cell presence and characteristics) between these two otherwise anatomically-similar tumors. Using TCGA data, we compared the immune landscape between HPV+ and HPV- treatment-naïve HNSCC. As compared to HPV- samples, HPV+ HNSCC exhibited a strong Th1 response characterized by increased infiltration with multiple types of immune cells and expression of their effector molecules. HPV+ HNSCC also expressed higher levels of CD39 and multiple T-cell exhaustion markers including LAG3, PD1, TIGIT, and TIM3 compared to HPV- HNSCC. Importantly, patients with higher expression of these exhaustion markers-indicative of a T-cell-inflamed tumor-correlated with markedly improved survival in HPV+, but not HPV-, HNSCC. Thus, profound differences exist between the immune landscape of HPV+ and HPV- HNSCC. These results suggest that immune checkpoint inhibitor therapy is a promising treatment strategy for HPV+ HNSCC, and that expression of immune checkpoint molecules could serve as a predictive biomarker of patient outcome in HPV+ HNSCC.

10.
Article de Anglais | MEDLINE | ID: mdl-30345081

RÉSUMÉ

TdT-interacting factor 1 (TdIF1) is a ubiquitously expressed DNA- and protein-binding protein that directly binds to terminal deoxynucleotidyl transferase (TdT) polymerase. Little is known about the functional role of TdIF1 in cancer cellular signaling, nor has it previously been identified as aberrant in any type of cancer. We report here for the first time that TdIF1 is abundantly expressed in clinical lung cancer patients and that high expression of TdIF1 is associated with poor patient prognosis. We further established that TdIF1 is highly expressed in human non-small cell lung cancer (NSCLC) cell lines compared to a normal lung cell line. shRNA-mediated gene silencing of TdIF1 resulted in the suppression of proliferation and anchorage-independent colony formation of the A549 adenocarcinoma cell line. Moreover, when these TdIF1-silenced cells were used to establish a mouse xenograft model of human NSCLC, tumor size was greatly reduced. These data suggest that TdIF1 is a potent regulator of lung tumor development. Several cell cycle-related and tumor growth signaling pathways, including the p53 and HDAC1/2 pathways, were identified as participating in the TdIF1 signaling network by in silico analysis. Microarray, transcriptome and protein-level analyses validated p53 and HDAC1/2 modulation upon TdIF1 downregulation in an NSCLC cellular model. Moreover, several other cell cycle regulators were affected at the transcript level by TdIF1 silencing, including an increase in CDKN1A/p21 transcripts. Taken together, these results indicate that TdIF1 is a bona fide tumor-promoting factor in NSCLC and a potential target for therapy.

11.
PLoS One ; 13(1): e0191766, 2018.
Article de Anglais | MEDLINE | ID: mdl-29364966

RÉSUMÉ

Emerging drug-resistance and drug-associated toxicities are two major factors limiting successful cancer therapy. Combinations of chemotherapeutic drugs have been used in the clinic to improve patient outcome. However, cancer cells can acquire resistance to drugs, alone or in combination. Resistant tumors can also exhibit cross-resistance to other chemotherapeutic agents, resulting in sub-optimal treatment and/or treatment failure. Therefore, developing novel oncology drugs that induce no or little acquired resistance and with a favorable safety profile is essential. We show here that combining COTI-2, a novel clinical stage agent, with multiple chemotherapeutic and targeted agents enhances the activity of these drugs in vitro and in vivo. Importantly, no overt toxicity was observed in the combination treatment groups in vivo. Furthermore, unlike the tested chemotherapeutic drugs, cancer cells did not develop resistance to COTI-2. Finally, some chemo-resistant tumor cell lines only showed mild cross-resistance to COTI-2 while most remained sensitive to it.


Sujet(s)
Aminoquinoléines/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Thiosemicarbazones/usage thérapeutique , Animaux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Cisplatine/administration et posologie , Désoxycytidine/administration et posologie , Désoxycytidine/analogues et dérivés , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon/anatomopathologie , Souris , Paclitaxel/administration et posologie , Vinblastine/administration et posologie , Vinblastine/analogues et dérivés , Vinorelbine , Tests d'activité antitumorale sur modèle de xénogreffe ,
12.
J Pharmacol Exp Ther ; 364(1): 46-54, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29061656

RÉSUMÉ

Although cancer cell genetic instability contributes to characteristics that mediate tumorigenicity, it also contributes to the tumor-selective toxicity of some chemotherapy drugs. This synthetic lethality can be enhanced by inhibitors of DNA repair. To exploit this potential Achilles heel, we tested the ability of a RAD51 inhibitor to potentiate the cytotoxicity of chemotherapy drugs. 2-(Benzylsulfonyl)-1-(1H-indol-3-yl)-1,2-dihydroisoquinoline (IBR2) inhibits RAD51-mediated DNA double-strand break repair but also enhances cytotoxicity of the Bcr-Abl inhibitor imatinib. The potential for synergy between IBR2 and more drugs was examined in vitro across a spectrum of cancer cell lines from various tissues. Cells were exposed to IBR2 simultaneously with inhibitors of receptor tyrosine kinases, DNA-damaging agents, or microtubule disruptors. IBR2, at concentrations that inhibited proliferation between 0% and 75%, enhanced toxicity by up to 80% of imatinib and regorafenib (targets RAF and kit); epidermal growth factor receptor inhibitors erlotinib, gefitinib, afatinib, and osimertinib; and vincristine, an inhibitor of microtubule function. However, IBR2 antagonized the action of olaparib, cisplatin, melphalan, and irinotecan. A vincristine-resistant squamous cell line was not cross resistant to imatinib, but IBR2 and another RAD51 inhibitor (B02) enhanced imatinib toxicity in this cell line, its HN-5a parent, and the colon cancer line HT-29 by up to 60% and much better than verapamil, a P-glycoprotein inhibitor (P < 0.05). Given the disparate agents the functions of which are enhanced by IBR2, the mechanisms of enhancement may be multimodal. Whether RAD51 is common to these mechanisms remains to be elucidated, but it provides the potential for selectivity to tumor cells.


Sujet(s)
Prolifération cellulaire/effets des médicaments et des substances chimiques , Indoles/administration et posologie , Protéines microtubulaires/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/administration et posologie , Rad51 Recombinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Tétrahydroisoquinoléines/administration et posologie , Cellules A549 , Antinéoplasiques/administration et posologie , Prolifération cellulaire/physiologie , Relation dose-effet des médicaments , Synergie des médicaments , Cellules HEK293 , Cellules HT29 , Humains , Cellules K562 , Cellules MCF-7 , Protéines microtubulaires/métabolisme , Rad51 Recombinase/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme
13.
Oncotarget ; 8(36): 60724, 2017 09 01.
Article de Anglais | MEDLINE | ID: mdl-29062467

RÉSUMÉ

[This corrects the article DOI: 10.18632/oncotarget.9133.].

14.
Crit Care ; 21(1): 210, 2017 08 08.
Article de Anglais | MEDLINE | ID: mdl-28789683

RÉSUMÉ

BACKGROUND: Despite many animal studies and clinical trials, mortality in sepsis remains high. This may be due to the fact that most experimental studies of sepsis employ young animals, whereas the majority of septic patients are elderly (60 - 70 years). The objective of the present study was to examine the sepsis-induced inflammatory and pro-coagulant responses in aged mice. Since running exercise protects against a variety of diseases, we also examined the effect of voluntary running on septic responses in aged mice. METHODS: Male C57BL/6 mice were housed in our institute from 2-3 to 22 months (an age mimicking that of the elderly). Mice were prevented from becoming obese by food restriction (given 70-90% of ad libitum consumption amount). Between 20 and 22 months, a subgroup of mice ran voluntarily on wheels, alternating 1-3 days of running with 1-2 days of rest. At 22 months, mice were intraperitoneally injected with sterile saline (control) or 3.75 g/kg fecal slurry (septic). At 7 h post injection, we examined (1) neutrophil influx in the lung and liver by measuring myeloperoxidase and/or neutrophil elastase in the tissue homogenates by spectrophotometry, (2) interleukin 6 (IL6) and KC in the lung lavage by ELISA, (3) pulmonary surfactant function by measuring percentage of large aggregates, (4) capillary plugging (pro-coagulant response) in skeletal muscle by intravital microscopy, (5) endothelial nitric oxide synthase (eNOS) protein in skeletal muscle (eNOS-derived NO is putative inhibitor of capillary plugging) by immunoblotting, and (6) systemic blood platelet counts by hemocytometry. RESULTS: Sepsis caused high levels of pulmonary myeloperoxidase, elastase, IL6, KC, liver myeloperoxidase, and capillary plugging. Sepsis also caused low levels of surfactant function and platelet counts. Running exercise increased eNOS protein and attenuated the septic responses. CONCLUSIONS: Voluntary running protects against exacerbated sepsis-induced inflammatory and pro-coagulant responses in aged mice. Protection against pro-coagulant responses may involve eNOS upregulation. The present discovery in aged mice calls for clinical investigation into potential beneficial effects of exercise on septic outcomes in the elderly.


Sujet(s)
Course à pied/physiologie , Sepsie/physiopathologie , Vieillissement/métabolisme , Vieillissement/physiologie , Analyse de variance , Animaux , Test ELISA/méthodes , Interleukine-6/analyse , Interleukine-6/sang , Leukocyte elastase/analyse , Leukocyte elastase/sang , Mâle , Souris , Souris de lignée C57BL , Myeloperoxidase/analyse , Myeloperoxidase/sang
15.
Clin Cancer Res ; 23(4): 1068-1079, 2017 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-27856600

RÉSUMÉ

Purpose: The ubiquitously expressed transmembrane glycoprotein CD47 delivers an anti-phagocytic (do not eat) signal by binding signal-regulatory protein α (SIRPα) on macrophages. CD47 is overexpressed in cancer cells and its expression is associated with poor clinical outcomes. TTI-621 (SIRPαFc) is a fully human recombinant fusion protein that blocks the CD47-SIRPα axis by binding to human CD47 and enhancing phagocytosis of malignant cells. Blockade of this inhibitory axis using TTI-621 has emerged as a promising therapeutic strategy to promote tumor cell eradication.Experimental Design: The ability of TTI-621 to promote macrophage-mediated phagocytosis of human tumor cells was assessed using both confocal microscopy and flow cytometry. In vivo antitumor efficacy was evaluated in xenograft and syngeneic models and the role of the Fc region in antitumor activity was evaluated using SIRPαFc constructs with different Fc tails.Results: TTI-621 enhanced macrophage-mediated phagocytosis of both hematologic and solid tumor cells, while sparing normal cells. In vivo, TTI-621 effectively controlled the growth of aggressive AML and B lymphoma xenografts and was efficacious in a syngeneic B lymphoma model. The IgG1 Fc tail of TTI-621 plays a critical role in its antitumor activity, presumably by engaging activating Fcγ receptors on macrophages. Finally, TTI-621 exhibits minimal binding to human erythrocytes, thereby differentiating it from CD47 blocking antibodies.Conclusions: These data indicate that TTI-621 is active across a broad range of human tumors. These results further establish CD47 as a critical regulator of innate immune surveillance and form the basis for clinical development of TTI-621 in multiple oncology indications. Clin Cancer Res; 23(4); 1068-79. ©2016 AACR.


Sujet(s)
Antigènes de différenciation/génétique , Antigènes CD47/génétique , Immunoglobuline G/immunologie , Tumeurs/traitement médicamenteux , Récepteurs immunologiques/génétique , Protéines de fusion recombinantes/administration et posologie , Animaux , Anticorps bloquants/administration et posologie , Anticorps bloquants/immunologie , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/immunologie , Antigènes CD47/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Humains , Souris , Tumeurs/immunologie , Tumeurs/anatomopathologie , Phagocytose/effets des médicaments et des substances chimiques , Liaison aux protéines , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Récepteurs immunologiques/antagonistes et inhibiteurs , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Oncotarget ; 7(27): 41363-41379, 2016 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-27150056

RÉSUMÉ

Identification of novel anti-cancer compounds with high efficacy and low toxicity is critical in drug development. High-throughput screening and other such strategies are generally resource-intensive. Therefore, in silico computer-aided drug design has gained rapid acceptance and popularity. We employed our proprietary computational platform (CHEMSAS®), which uses a unique combination of traditional and modern pharmacology principles, statistical modeling, medicinal chemistry, and machine-learning technologies to discover and optimize novel compounds that could target various cancers. COTI-2 is a small molecule candidate anti-cancer drug identified using CHEMSAS. This study describes the in vitro and in vivo evaluation of COTI-2. Our data demonstrate that COTI-2 is effective against a diverse group of human cancer cell lines regardless of their tissue of origin or genetic makeup. Most treated cancer cell lines were sensitive to COTI-2 at nanomolar concentrations. When compared to traditional chemotherapy or targeted-therapy agents, COTI-2 showed superior activity against tumor cells, in vitro and in vivo. Despite its potent anti-tumor efficacy, COTI-2 was safe and well-tolerated in vivo. Although the mechanism of action of COTI-2 is still under investigation, preliminary results indicate that it is not a traditional kinase or an Hsp90 inhibitor.


Sujet(s)
Aminoquinoléines/usage thérapeutique , Antinéoplasiques/isolement et purification , Antinéoplasiques/usage thérapeutique , Tumeurs/traitement médicamenteux , Thiosemicarbazones/usage thérapeutique , Adenosine triphosphatases/antagonistes et inhibiteurs , Aminoquinoléines/isolement et purification , Animaux , Lignée cellulaire tumorale , Chimie pharmaceutique , Découverte de médicament , Femelle , Cellules HCT116 , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Cellules HT29 , Humains , Cellules MCF-7 , Souris , Souris nude , Tumeurs/anatomopathologie , Thiosemicarbazones/isolement et purification , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Oncotarget ; 7(15): 20825-39, 2016 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-26959114

RÉSUMÉ

Human tumor heterogeneity promotes therapeutic failure by increasing the likelihood of resistant cell subpopulations. The PARP-1 inhibitor olaparib is approved for use in BRCA-mutated ovarian cancers but BRCA2-reversion mutations lead to functional homologous recombination repair (HRR) and olaparib resistance. To overcome that resistance and expand use of PARP1 inhibition to cancers with functional HRR, we developed an antisense strategy to render the majority of tumor cells in a population BRCA2-deficient. We predicted that this strategy would render HRR-proficient tumor cells sensitive to olaparib and prevent emergence of resistance in a tumor cell population heterogeneous for HRR proficiency. We report that BRCA2 downregulation sensitized multiple human tumor cell lines (but not non-cancer human kidney cells) to olaparib and, combined with olaparib, increased aneuploidy and chromosomal translocations in human tumor cells. In a mixed HRR-proficient and HRR-deficient cell population, olaparib monotherapy allowed outgrowth of HRR-proficient cells resistant to subsequent olaparib treatment. Combined BRCA2 inhibition and olaparib treatment prevented selection of HRR-proficient cells and inhibited proliferation of the entire population. Treatment with BRCA2 siRNA and olaparib decreased ovarian xenograft growth in mice more effectively than either treatment alone. In vivo use of BRCA2 antisense oligonucleotides may be a viable option to expand clinical use of olaparib and prevent resistance.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéine BRCA2/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Phtalazines/pharmacologie , Pipérazines/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Protéine BRCA2/génétique , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Souris , Souris nude , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Petit ARN interférent/génétique , Réparation de l'ADN par recombinaison/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Tumori ; 102(1): 31-9, 2016.
Article de Anglais | MEDLINE | ID: mdl-26429639

RÉSUMÉ

AIMS AND BACKGROUND: In light of the need for more selective anticancer therapy, much work has been directed at developing compounds or biological agents that target functions specific to cancer cells. To this end, numerous viruses have been engineered to exploit the dependence of cancer cells on particular anomalies that contribute to their rogue proliferative activity, such as dysfunctional p53, overactive mitogenic signaling, or a defective interferon response. The oncolytic human adenovirus dl1520 (ONYX-015) was engineered to propagate specifically in p53-deficient tumors, which comprise over half of all tumors. Based on successes in clinical trials, the full potential of dl1520 and other oncolytic viruses may be even better realized by using them in combination with conventional chemotherapy drugs. METHODS: As a model system in which to test this potential, representative cell lines from 2 common cancer types, oral squamous cell carcinoma (HN-5a) and colon adenocarcinoma (HT-29), were chosen, as well as platinum-drug-resistant variants of each. RESULTS: Following preliminary screening of virus and drug combinations, dl1520 and melphalan were found to synergistically inhibit proliferation of all the cancer cell lines. Melphalan pretreatment or cotreatment with dl1520 enhanced inhibition of proliferation by dl1520 by up to 60% and increased apoptosis by up to 25%. The tight-junction protein CAR (coxsackie and adenovirus receptor), via which adenovirus enters cells, was not upregulated by treatment with melphalan, suggesting that other mechanisms contribute to synergy. CONCLUSIONS: The synergy between melphalan and dl1520 suggests that tumor-selective cell killing by oncolytic viruses may be augmented by combining with cytotoxic drugs.


Sujet(s)
Adenoviridae , Antinéoplasiques/pharmacologie , Melphalan/pharmacologie , Tumeurs/traitement médicamenteux , Virus oncolytiques , Vaccins antiviraux/pharmacologie , Adénocarcinome/traitement médicamenteux , Antinéoplasiques alcoylants/pharmacologie , Carcinome épidermoïde/traitement médicamenteux , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs du côlon/traitement médicamenteux , Synergie des médicaments , Humains , Tumeurs de la bouche/traitement médicamenteux
20.
Methods Mol Biol ; 1364: 151-63, 2016.
Article de Anglais | MEDLINE | ID: mdl-26472449

RÉSUMÉ

The expression of a gene can be specifically downregulated by small interfering RNA (SiRNA). Modified carbon nanotubes (CNT) can be used to protect SiRNA and facilitate its entry into cells. Regardless of that, simple and efficient functionalization of CNT is lacking. Effective SiRNA delivery can be carried out using non-covalently functionalized CNT, where non-covalent (versus covalent) functionalization is simpler and more expeditious. Non-covalently functionalized single walled carbon nanotubes (SWCNT) that include a lipopolymer are described here. Polyethylenimine (PEI) conjugated to 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPE-PEG) was generated and the products used to disperse CNT to form DSPE-PEG-PEI/CNT (DGI/C), an agent capable of facilitating SiRNA delivery to cells in vitro and organs and cells in vivo.


Sujet(s)
Vecteurs de médicaments/composition chimique , Nanotubes de carbone/composition chimique , Phosphatidyléthanolamine/composition chimique , Polyéthylène glycols/composition chimique , Polyéthylèneimine/composition chimique , Petit ARN interférent/composition chimique , Animaux , Vecteurs de médicaments/synthèse chimique , Petit ARN interférent/génétique , Transfection
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...