Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 422
Filtrer
1.
Oncoimmunology ; 13(1): 2395067, 2024.
Article de Anglais | MEDLINE | ID: mdl-39188754

RÉSUMÉ

Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.


Sujet(s)
Imidazoles , Inhibiteurs de points de contrôle immunitaires , Poly I-C , Animaux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Poly I-C/administration et posologie , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Imidazoles/pharmacologie , Imidazoles/administration et posologie , Imidazoles/usage thérapeutique , Immunothérapie/méthodes , Humains , Récepteurs de type Toll/agonistes , Lignée cellulaire tumorale , Femelle , Tumeurs du côlon/immunologie , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/thérapie , Souris de lignée C57BL , Hydrogels/administration et posologie , Hydrogels/composition chimique ,
2.
Int J Med Sci ; 21(1): 107-122, 2024.
Article de Anglais | MEDLINE | ID: mdl-38164360

RÉSUMÉ

NF-κB activation is pivotal for the excess inflammation causing the critical condition and mortality of respiratory viral infection patients. This study was aimed to evaluate the effect of a banana plant extract (BPE) on suppressing NF-κB activity and acute lung inflammatory responses in mice induced by a synthetic double-stranded RNA viral mimetic, polyinosinic-polycytidylic acid (poly (I:C)). The inflammatory responses were analyzed by immunohistochemistry and HE stains and ELISA. The NF-κB activities were detected by immunohistochemistry in vivo and immunofluorescence and Western blot in vitro. Results showed that BPE significantly decreased influx of immune cells (neutrophils, lymphocytes, and total WBC), markedly suppressed the elevation of pro-inflammatory cytokines and chemokines (IL-6, RANTES, IFN-γ, MCP-1, keratinocyte-derived chemokine, and IL-17), and restored the diminished anti-inflammatory IL-10 in the bronchoalveolar lavage fluid (BALF) of poly (I:C)-stimulated mice. Accordingly, HE staining revealed that BPE treatment alleviated poly (I:C)-induced inflammatory cell infiltration and histopathologic changes in mice lungs. Moreover, immunohistochemical analysis showed that BPE reduced the pulmonary IL-6, CD11b (macrophage marker), and nuclear NF-κB p65 staining intensities, whilst restored that of IL-10 in poly (I:C)-stimulated mice. In vitro, BPE antagonized poly(I:C)-induced elevation of IL-6, nitric oxide, reactive oxygen species, NF-κB p65 signaling, and transient activation of p38 MAPK in human lung epithelial-like A549 cells. Taken together, BPE ameliorated viral mimic poly(I:C)-induced acute pulmonary inflammation in mice, evidenced by reduced inflammatory cell infiltration and regulation of both pro- and anti-inflammatory cytokines. The mechanism of action might closely associate with NF-κB signaling inhibition.


Sujet(s)
Musa , Pneumopathie infectieuse , Souris , Humains , Animaux , Facteur de transcription NF-kappa B , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Interleukine-10 , Interleukine-6 , Extraits de plantes/pharmacologie , Extraits de plantes/usage thérapeutique , Cytokines , Inflammation/induit chimiquement , Inflammation/traitement médicamenteux , Chimiokines , Anti-inflammatoires/usage thérapeutique
3.
Ann Anat ; 250: 152158, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37666464

RÉSUMÉ

BACKGROUND: Polyinosinic-polycytidylic acid (pIC) is a synthetic analog of double-stranded RNA. It is used as a synthetic adjuvant to induce an adaptive immune response. However, the effect of pIC on the development of mediastinal fat-associated lymphoid clusters (MFALCs) that regulate intrathoracic hemostasis has remained unidentified. METHODS: We investigated the impact of intranasal (i.n.) administration (pIC i.n. group) and intravenous (i.v.) administration (pIC i.v. group) of pIC on both MFALCs and lung tissue. RESULTS: Compared with the control phosphate-buffered saline (PBS) groups, both pIC-administered groups displayed a significant increase in the MFALC size (particularly in the pIC i.n. group), area of MFALC high endothelial venules (HEVs), area of lymphatic vessels (LVs), number of proliferating cells (particularly in the pIC i.v. group), and number of immune cells (B220+ B-lymphocytes, CD3+ T-lymphocytes, Iba1+ macrophages, and Gr-1+ granulocytes) in both MFALCs and lung tissues. In addition, a positive correlation was detected between MFALC size and proliferating cells, immune cell population, LVs, and HEVs within MFALCs in both groups. Except for the proliferating cell and B-lymphocyte populations in the i.n. administered group and granulocyte populations in both i.n. and i.v. administered routes, such correlations were significant. CONCLUSION: In all, our data indicate that local or systemic administration of pIC induces the development of MFALCs and can be used as an immunostimulant therapeutic strategy.


Sujet(s)
Vaisseaux lymphatiques , Poly I-C , Souris , Animaux , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Poumon , Lymphocytes T , Lymphocytes B
4.
J Control Release ; 361: 443-454, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37558053

RÉSUMÉ

Triple-negative breast cancer (TNBC) is highly aggressive and has no standard treatment. Although being considered as an alternative to conventional treatments for TNBC, immunotherapy has to deal with many challenges that hinder its efficacy, particularly the poor immunogenic condition of the tumor microenvironment (TME). Herein, we designed a liposomal nanoparticle (LN) platform that delivers simultaneously toll-like receptor 7 (imiquimod, IQ) and toll-like receptor 3 (poly(I:C), IC) agonists to take advantage of the different toll-like receptor (TLR) signaling pathways, which enhances the condition of TME from a "cold" to a "hot" immunogenic state. The optimized IQ/IC-loaded LN (IQ/IC-LN) was effectively internalized by cancer cells, macrophages, and dendritic cells, followed by the release of the delivered drugs and subsequent stimulation of the TLR3 and TLR7 signaling pathways. This stimulation encouraged the secretion of type I interferon (IFN-α, IFN-ß) and CXCLl0, a T-cell and antigen-presenting cells (APCs) recruitment chemokine, from both cancer cells and macrophages and polarized macrophages to the M1 subtype in in vitro studies. Notably, systemic administration of IQ/IC-LN allowed for the high accumulation of drug content in the tumor, followed by the effective uptake by immune cells in the TME. IQ/IC-LN treatment comprehensively enhanced the immunogenic condition in the TME, which robustly inhibited tumor growth in tumor-bearing mice. Furthermore, synergistic antitumor efficacy was obtained when the IQ/IC-LN-induced immunogenic state in TME was combined with anti-PD1 antibody therapy. Thus, our results suggest the potential of combining 2 TLR agonists to reform the TME from a "cold" to a "hot" state, supporting the therapeutic efficacy of immune checkpoint inhibitors.


Sujet(s)
Récepteur de type Toll-3 , Tumeurs du sein triple-négatives , Humains , Animaux , Souris , Tumeurs du sein triple-négatives/traitement médicamenteux , Adjuvants immunologiques , Liposomes , Poly I-C/usage thérapeutique , Immunothérapie/méthodes , Microenvironnement tumoral
5.
Environ Sci Pollut Res Int ; 30(10): 28118-28132, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36394807

RÉSUMÉ

Rhinovirus infection frequently causes COPD and asthma exacerbations. Impaired anti-viral signaling and reduced viral clearance have both been seen in sick bronchial epithelium, potentially increasing exacerbations. Polyinosinic:polycytidylic acid (Poly(I:C)), a Toll-like receptor-3 (TLR3) ligand, has been shown to cause a viral exacerbation of severe asthma by detecting double-stranded RNA (dsRNA). The purpose of this work was to determine the effect of a TLR3/dsRNA complex inhibitor-Calbiochem drug in the prevention of Poly(I:C)-induced airway inflammation following TLR3 activation and to uncover a potential pathway for the cure of asthma through TLR3 inhibition. Mice were sensitized with Poly(I:C) as an asthma model before being challenged by PBS and ovalbumin (OVA) chemicals. The mice were administered a TLR3/dsRNA complex inhibitor. Throughout the trial, the mice's body weight was measured after each dosage. Biochemical methods are used to analyze the protein as well as enzyme composition in airway tissues. BALF specimens are stained using Giemsa to identify inflammatory cells and lung histopathology to determine morphological abnormalities in lung tissues. By using the ELISA approach, cytokine levels such as TNF-α, IL-13, IL-6, IL-5, and IgE antibody expression in lung tissue and blood serum were assessed. TLR3/dsRNA complex inhibitor drug significantly lowered the number of cells in BALF and also on Giemsa staining slides. It also downregulated the level of TNF-α and IL-6 in contrast to OVA and Poly(I:C) administered in animals. A TLR3/dsRNA complex inhibitor decreased the fraction of oxidative stress markers (MDA, GSH, GPx, and CAT) in lung tissues while keeping the mice's body weight constant during the treatment period. By decreasing alveoli, bronchial narrowing, smooth muscle hypertrophy, and granulocyte levels, the TLR3/dsRNA complex blocker significantly reduced the histopathological damage caused by OVA and Poly(I:C) compounds. In an animal model utilizing ovalbumin, TLR3/dsRNA complex inhibitors similarly reduced the bronchial damage produced by Poly(I:C). A novel TLR3/dsRNA complex inhibitor is expected to be employed in clinical studies since it suppresses airway inflammation without inducing antiviral approach resistance.


Sujet(s)
Asthme , Facteur de nécrose tumorale alpha , Souris , Animaux , Facteur de nécrose tumorale alpha/métabolisme , Ovalbumine , ARN double brin/métabolisme , ARN double brin/usage thérapeutique , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/usage thérapeutique , Interleukine-6/métabolisme , Modèles animaux de maladie humaine , Asthme/induit chimiquement , Poumon/anatomopathologie , Inflammation/induit chimiquement , Inflammation/métabolisme , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Liquide de lavage bronchoalvéolaire
6.
Front Immunol ; 13: 882277, 2022.
Article de Anglais | MEDLINE | ID: mdl-35655783

RÉSUMÉ

Major Histocompatibility Complex (MHC)-I and -II genes are upregulated in intestinal epithelial cells (IECs) during active inflammatory bowel diseases (IBD), but little is known about how IBD-relevant pro-inflammatory signals and IBD drugs can regulate their expression. We have previously shown that the synthetic analog of double-stranded RNA (dsRNA) Polyinosinic:polycytidylic acid (Poly(I:C)), induces interferon stimulated genes (ISGs) in colon organoids (colonoids). These ISGs may be involved in the induction of antigen presentation. In the present study, we applied colonoids derived from non-IBD controls and ulcerative colitis patients to identify induction and effects of IBD-drugs on antigen presentation in IECs in the context of Tumor Necrosis Factor (TNF)-driven inflammation. By RNA sequencing, we show that a combination of TNF and Poly(I:C) strongly induced antigen-presentation gene signatures in colonoids, including expression of MHC-II genes. MHC-I and -II protein expression was confirmed by immunoblotting and immunofluorescence. TNF+Poly(I:C)-dependent upregulation of MHC-II expression was associated with increased expression of Janus Kinases JAK1/2 as well as increased activation of transcription factor Signal transducer and activator of transcription 1 (STAT1). Accordingly, pre-treatment of colonoids with IBD-approved pan-Janus Kinase (JAK) inhibitor Tofacitinib led to the downregulation of TNF+Poly(I:C)-dependent MHC-II expression associated with the abrogation of STAT1 activation. Pre-treatment with corticosteroid Budesonide, commonly used in IBD, did not alter MHC-II expression. Collectively, our results identify a regulatory role for IBD-relevant pro-inflammatory signals on MHC-II expression that is influenced by Tofacitinib.


Sujet(s)
Maladies inflammatoires intestinales , Inhibiteurs des Janus kinases , Côlon/anatomopathologie , Épithélium/métabolisme , Humains , Maladies inflammatoires intestinales/métabolisme , Inhibiteurs des Janus kinases/usage thérapeutique , Complexe majeur d'histocompatibilité , Pipéridines , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Pyrimidines , Facteur de nécrose tumorale alpha/usage thérapeutique
7.
Viruses ; 14(2)2022 01 19.
Article de Anglais | MEDLINE | ID: mdl-35215785

RÉSUMÉ

SARS-CoV-2, a member of the coronavirus family, is the causative agent of the COVID-19 pandemic. Currently, there is still an urgent need in developing an efficient therapeutic intervention. In this study, we aimed at evaluating the therapeutic effect of a single intranasal treatment of the TLR3/MDA5 synthetic agonist Poly(I:C) against a lethal dose of SARS-CoV-2 in K18-hACE2 transgenic mice. We demonstrate here that early Poly(I:C) treatment acts synergistically with SARS-CoV-2 to induce an intense, immediate and transient upregulation of innate immunity-related genes in lungs. This effect is accompanied by viral load reduction, lung and brain cytokine storms prevention and increased levels of macrophages and NK cells, resulting in 83% mice survival, concomitantly with long-term immunization. Thus, priming the lung innate immunity by Poly(I:C) or alike may provide an immediate, efficient and safe protective measure against SARS-CoV-2 infection.


Sujet(s)
COVID-19/immunologie , COVID-19/prévention et contrôle , Immunité innée , Poly I-C/immunologie , Poly I-C/usage thérapeutique , SARS-CoV-2/effets des médicaments et des substances chimiques , Récepteur de type Toll-3/agonistes , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Animaux , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/prévention et contrôle , Modèles animaux de maladie humaine , Femelle , Humains , Poumon/immunologie , Poumon/virologie , Souris , Souris transgéniques , SARS-CoV-2/immunologie , Récepteur de type Toll-3/immunologie , Charge virale/effets des médicaments et des substances chimiques , Traitements médicamenteux de la COVID-19
8.
Hepatology ; 76(3): 630-645, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-34860431

RÉSUMÉ

BACKGROUND AND AIMS: Immunotherapy with programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) blockade has shown low response rates in liver cancer patients, with the underlying mechanisms unclear. To decipher a specific impact of the liver microenvironment, we compared the effects of anti-PD-L1 antibody (αPD-L1) blockade on the same tumor grown s.c. or in the liver. APPROACH AND RESULTS: We generated s.c. tumors in mice by inoculating MC38 colorectal cancer (CRC) cells under the skin and metastatic liver tumors by portal vein or splenic injection of CRC cells. Tumor-bearing mice were treated by i.p. injection of αPD-L1, polyinosinic:polycytidylic acid (poly[I:C]), or both. αPD-L1 monotherapy significantly suppressed s.c. tumor growth, but showed no effect on metastatic liver tumors. However, the combination of αPD-L1 with poly(I:C), an innate immunity-stimulating reagent, robustly inhibited tumor progression in liver. The combination therapy effectively down-regulated myeloid-derived suppressor cells (MDSCs), but up-regulated ratios of M1/M2 macrophages, CD8/CD4, and CD8/regulatory T (Treg) cells infiltrated into liver tumors and whole liver. A group of long-lasting T-bet+ Eomes- PD-1- cytotoxic T cells was maintained in the combo-treated liver, leading to resistance to tumor recurrence. Depleting macrophages or blocking type Ⅰ interferon signaling abrogated the synergistic antitumor effect of αPD-L1 and poly(I:C), indicating a requirement of boosting innate immunity for optimized activation of cytotoxic T cells by PD-1/PD-L1 blockade. CONCLUSIONS: The poor response of liver cancers to αPD-L1 therapy is largely attributable to a unique hepatic immunotolerant microenvironment, independent of tumor origins or types. The success of a combinatorial immunotherapy relies on coordinated inhibition or activation of various innate and adaptive immune cell activities.


Sujet(s)
Antigène CD274 , Tumeurs du foie , Animaux , Antigène CD274/métabolisme , Lignée cellulaire tumorale , Facteurs immunologiques/pharmacologie , Tumeurs du foie/traitement médicamenteux , Souris , Récidive tumorale locale , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Récepteur-1 de mort cellulaire programmée , Microenvironnement tumoral
9.
J Immunother Cancer ; 9(9)2021 09.
Article de Anglais | MEDLINE | ID: mdl-34531246

RÉSUMÉ

BACKGROUND: Tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer and hinder the antitumoral efficacy of most treatments currently applied in the clinic. Previous studies have evaluated the antitumoral immune response triggered by (TLR) agonists, such as poly(I:C), imiquimod (R837) or resiquimod (R848) as monotherapies; however, their combination for the treatment of cancer has not been explored. This study investigates the antitumoral efficacy and the macrophage reprogramming triggered by poly(I:C) combined with R848 or with R837, versus single treatments. METHODS: TLR agonist treatments were evaluated in vitro for toxicity and immunostimulatory activity by Alamar Blue, ELISA and flow cytometry using primary human and murine M-CSF-differentiated macrophages. Cytotoxic activity of TLR-treated macrophages toward cancer cells was evaluated with an in vitro functional assay by flow cytometry. For in vivo experiments, the CMT167 lung cancer model and the MN/MCA1 fibrosarcoma model metastasizing to lungs were used; tumor-infiltrating leukocytes were evaluated by flow cytometry, RT-qPCR, multispectral immunophenotyping, quantitative proteomic experiments, and protein-protein interaction analysis. RESULTS: Results demonstrated the higher efficacy of poly(I:C) combined with R848 versus single treatments or combined with R837 to polarize macrophages toward M1-like antitumor effectors in vitro. In vivo, the intratumoral synergistic combination of poly(I:C)+R848 significantly prevented tumor growth and metastasis in lung cancer and fibrosarcoma immunocompetent murine models. Regressing tumors showed increased infiltration of macrophages with a higher M1:M2 ratio, recruitment of CD4+ and CD8+ T cells, accompanied by a reduction of immunosuppressive CD206+ TAMs and FOXP3+/CD4+ T cells. The depletion of both CD4+ and CD8+ T cells resulted in complete loss of treatment efficacy. Treated mice acquired systemic antitumoral response and resistance to tumor rechallenge mediated by boosted macrophage cytotoxic activity and T-cell proliferation. Proteomic experiments validate the superior activation of innate immunity by poly(I:C)+R848 combination versus single treatments or poly(I:C)+R837, and protein-protein-interaction network analysis reveal the key activation of the STAT1 pathway. DISCUSSION: These findings demonstrate the antitumor immune responses mediated by macrophage activation on local administration of poly(I:C)+R848 combination and support the intratumoral application of this therapy to patients with solid tumors in the clinic.


Sujet(s)
Antiviraux/usage thérapeutique , Association thérapeutique/méthodes , Imidazoles/usage thérapeutique , Immunothérapie/méthodes , Tumeurs/traitement médicamenteux , Poly I-C/usage thérapeutique , Macrophages associés aux tumeurs/métabolisme , Animaux , Antiviraux/pharmacologie , Lignée cellulaire tumorale , Synergie des médicaments , Humains , Imidazoles/pharmacologie , Souris , Poly I-C/pharmacologie
10.
J Exp Clin Cancer Res ; 40(1): 213, 2021 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-34172082

RÉSUMÉ

Immunotherapy is currently under intensive investigation as a potential breakthrough treatment option for glioblastoma. Given the anatomical and immunological complexities surrounding glioblastoma, lymphocytes that infiltrate the brain to develop durable immunity with memory will be key. Polyinosinic:polycytidylic acid, or poly(I:C), and its derivative poly-ICLC could serve as a priming or boosting therapy to unleash lymphocytes and other factors in the (immuno)therapeutic armory against glioblastoma. Here, we present a systematic review on the effects and efficacy of poly(I:C)/poly-ICLC for glioblastoma treatment, ranging from preclinical work on cellular and murine glioblastoma models to reported and ongoing clinical studies. MEDLINE was searched until 15 May 2021 to identify preclinical (glioblastoma cells, murine models) and clinical studies that investigated poly(I:C) or poly-ICLC in glioblastoma. A systematic review approach was conducted according to PRISMA guidelines. ClinicalTrials.gov was queried for ongoing clinical studies. Direct pro-tumorigenic effects of poly(I:C) on glioblastoma cells have not been described. On the contrary, poly(I:C) changes the immunological profile of glioblastoma cells and can also kill them directly. In murine glioblastoma models, poly(I:C) has shown therapeutic relevance as an adjuvant therapy to several treatment modalities, including vaccination and immune checkpoint blockade. Clinically, mostly as an adjuvant to dendritic cell or peptide vaccines, poly-ICLC has been demonstrated to be safe and capable of eliciting immunological activity to boost therapeutic responses. Poly-ICLC could be a valuable tool to enhance immunotherapeutic approaches for glioblastoma. We conclude by proposing several promising combination strategies that might advance glioblastoma immunotherapy and discuss key pre-clinical aspects to improve clinical translation.


Sujet(s)
Tumeurs du cerveau/traitement médicamenteux , Carboxyméthylcellulose de sodium/analogues et dérivés , Glioblastome/traitement médicamenteux , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Animaux , Tumeurs du cerveau/immunologie , Vaccins anticancéreux/usage thérapeutique , Carboxyméthylcellulose de sodium/usage thérapeutique , Essais cliniques comme sujet , Glioblastome/immunologie , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie , Souris , Polylysine/usage thérapeutique
11.
Nat Commun ; 12(1): 3958, 2021 06 25.
Article de Anglais | MEDLINE | ID: mdl-34172753

RÉSUMÉ

Astrocytes play important roles in neurological disorders such as stroke, injury, and neurodegeneration. Most knowledge on astrocyte biology is based on studies of mouse models and the similarities and differences between human and mouse astrocytes are insufficiently characterized, presenting a barrier in translational research. Based on analyses of acutely purified astrocytes, serum-free cultures of primary astrocytes, and xenografted chimeric mice, we find extensive conservation in astrocytic gene expression between human and mouse samples. However, the genes involved in defense response and metabolism show species-specific differences. Human astrocytes exhibit greater susceptibility to oxidative stress than mouse astrocytes, due to differences in mitochondrial physiology and detoxification pathways. In addition, we find that mouse but not human astrocytes activate a molecular program for neural repair under hypoxia, whereas human but not mouse astrocytes activate the antigen presentation pathway under inflammatory conditions. Here, we show species-dependent properties of astrocytes, which can be informative for improving translation from mouse models to humans.


Sujet(s)
Astrocytes/physiologie , Animaux , Présentation d'antigène , Astrocytes/effets des médicaments et des substances chimiques , Cellules cultivées , Expression des gènes/effets des médicaments et des substances chimiques , Humains , Inactivation métabolique , Inflammation , Souris , Mitochondries/métabolisme , Maladies du système nerveux/traitement médicamenteux , Maladies du système nerveux/anatomopathologie , Stress oxydatif , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Spécificité d'espèce , Transcriptome/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/usage thérapeutique
12.
Int J Mol Sci ; 22(8)2021 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-33920699

RÉSUMÉ

Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a growing number of malignancies. However, overcoming primary or secondary resistances is difficult due to pharmacokinetics issues and side effects associated with high systemic exposure. Local or regional expression of monoclonal antibodies (mAbs) using gene therapy vectors can alleviate this problem. In this work, we describe a high-capacity adenoviral vector (HCA-EFZP-aPDL1) equipped with a mifepristone-inducible system for the controlled expression of an anti-programmed death ligand 1 (PD-L1) blocking antibody. The vector was tested in an immune-competent mouse model of colorectal cancer based on implantation of MC38 cells. A single local administration of HCA-EFZP-aPDL1 in subcutaneous lesions led to a significant reduction in tumor growth with minimal release of the antibody in the circulation. When the vector was tested in a more stringent setting (rapidly progressing peritoneal carcinomatosis), the antitumor effect was marginal even in combination with other immune-stimulatory agents such as polyinosinic-polycytidylic acid (pI:C), blocking mAbs for T cell immunoglobulin, mucin-domain containing-3 (TIM-3) or agonistic mAbs for 4-1BB (CD137). In contrast, macrophage depletion by clodronate liposomes enhanced the efficacy of HCA-EFZP-aPDL1. These results highlight the importance of addressing macrophage-associated immunoregulatory mechanisms to overcome resistance to ICIs in the context of colorectal cancer.


Sujet(s)
Anticorps bloquants/génétique , Antigène CD274/métabolisme , Carcinomes/thérapie , Thérapie génétique/méthodes , Immunothérapie/méthodes , Macrophages/immunologie , Tumeurs du péritoine/thérapie , Adenoviridae/génétique , Animaux , Anticorps bloquants/immunologie , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Lignée cellulaire , Femelle , Vecteurs génétiques/génétique , Inhibiteurs de points de contrôle immunitaires/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Facteurs immunologiques/usage thérapeutique , Souris , Souris de lignée C57BL , Poly I-C/usage thérapeutique
13.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-33751208

RÉSUMÉ

The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (µg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.


Sujet(s)
Antigènes néoplasiques/usage thérapeutique , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Carboxyméthylcellulose de sodium/analogues et dérivés , Tumeurs de l'oesophage/traitement médicamenteux , Glucanes/usage thérapeutique , Protéines membranaires/usage thérapeutique , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Adjuvants immunologiques/usage thérapeutique , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Antigènes néoplasiques/immunologie , Carboxyméthylcellulose de sodium/usage thérapeutique , Tumeurs de l'oesophage/immunologie , Femelle , Glucanes/immunologie , Humains , Inducteurs de l'interféron/immunologie , Inducteurs de l'interféron/usage thérapeutique , Mâle , Protéines membranaires/immunologie , Souris , Adulte d'âge moyen , Nanoparticules , Poly I-C/immunologie , Polylysine/immunologie , Polylysine/usage thérapeutique
14.
Minerva Med ; 112(1): 144-152, 2021 Feb.
Article de Anglais | MEDLINE | ID: mdl-33104300

RÉSUMÉ

The world is now entering its 9th month of combat against a pandemic of deadly pneumonia. Started out from China in December 2019, the disease has been declared as caused by infection with a so far unknown RNA Coronavirus of the respiratory family, then named severe acute respiratory syndrome coronavirus SARS-CoV-2. In the absence of a vaccine, and with scientists still struggling for an effective therapy, COVID-19 (the SARS-dependent syndrome) carries up to now, a death toll of more than 590,000 (July 18,2020) undermining jobs and finance of contemporary society in all continents. Social distancing, the only measure hitherto shown to restrain virus spread, has been progressively loosened from May 2020 in some countries, leaving us in the fear of repeat attacks from the unchecked virus. We discuss the problem and propose to tentatively boost the antivirus cell machinery by using lab-made viral mimics to engage cell receptors.


Sujet(s)
COVID-19/thérapie , SARS-CoV-2 , COVID-19/complications , COVID-19/épidémiologie , Carboxyméthylcellulose de sodium/analogues et dérivés , Carboxyméthylcellulose de sodium/usage thérapeutique , Syndrome de libération de cytokines/étiologie , Humains , Immunisation passive , Inducteurs de l'interféron/usage thérapeutique , Maladie de Kawasaki/étiologie , Distanciation physique , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Polylysine/usage thérapeutique , Guides de bonnes pratiques cliniques comme sujet , ARN double brin/effets des médicaments et des substances chimiques , ARN viral/effets des médicaments et des substances chimiques , Récidive , SARS-CoV-2/classification , SARS-CoV-2/génétique , SARS-CoV-2/pathogénicité , Prévention secondaire , Traitements médicamenteux de la COVID-19 , Sérothérapie COVID-19
15.
Cancer Discov ; 11(2): 408-423, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33046534

RÉSUMÉ

For anthracycline-based chemotherapy to be immunogenic, dying cancer cells must release annexin A1 (ANXA1) that subsequently interacts with the pattern recognition receptor, formyl peptide receptor 1 (FPR1), on the surface of dendritic cells (DC). Approximately 30% of individuals bear loss-of-function alleles of FPR1, calling for strategies to ameliorate their anticancer immune response. Here, we show that immunotherapy with a ligand of Toll-like receptor-3, polyinosinic:polycytidylic acid (pIC), restores the deficient response to chemotherapy of tumors lacking ANXA1 developing in immunocompetent mice or those of normal cancers growing in FPR1-deficient mice. This effect was accompanied by improved DC- and T-lymphocyte-mediated anticancer immunity. Of note, carcinogen-induced breast cancers precociously developed in FPR1-deficient mice as compared with wild-type controls. A similar tendency for earlier cancer development was found in patients carrying the loss-of-function allele of FPR1. These findings have potential implications for the clinical management of FPR1-deficient patients. SIGNIFICANCE: The loss-of-function variant rs867228 in FPR1, harbored by approximately 30% of the world population, is associated with the precocious manifestation of breast, colorectal, esophageal, and head and neck carcinomas. pIC restores deficient chemotherapeutic responses in mice lacking Fpr1, suggesting a personalized strategy for compensating for the FPR1 defect.This article is highlighted in the In This Issue feature, p. 211.


Sujet(s)
Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , Ligands , Poly I-C/usage thérapeutique , Récepteur de type Toll-3 , Animaux , Tumeurs colorectales/génétique , Modèles animaux de maladie humaine , Humains , Souris , Souris transgéniques , Poly I-C/pharmacologie , Récepteurs aux peptides formylés/génétique
16.
Mol Oncol ; 15(5): 1289-1307, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33342034

RÉSUMÉ

Development of innovative therapeutic modalities would address an unmet clinical need in the treatment of triple negative breast cancer (TNBC). Activation of retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) such as melanoma differentiation-associated gene 5 (MDA5) and RIG-I in cancer cells is suggested to suppress tumor progression by inducing cell death. Transfection of polyI:C, a conventionally used synthetic double-stranded RNA (dsRNA) analogue that activates RLRs, has been evaluated in clinical trials. However, detailed mechanisms of tumor suppression by RLRs, especially interactions with other signaling pathways, remain elusive. Here, we showed that transfection of polyI:C suppressed transforming growth factor-ß (TGF-ß) signaling in a MDA5- and RIG-I-dependent manner. We found that suppression of TGF-ß signaling by polyI:C promoted cancer cell death, which was attenuated by forced expression of constitutively active Smad3. More detailed analysis suggested that cell death by polyI:C transfection exhibited characteristics of pyroptosis, which is distinct from apoptosis. Therapeutic efficacy of polyI:C transfection was also demonstrated using a mouse model. These results indicated that intratumor administration of polyI:C and related dsRNA analogues may be promising treatments for TNBC through inhibition of the anti-pyroptotic function of TGF-ß.


Sujet(s)
Pyroptose , ARN double brin/pharmacologie , Facteur de croissance transformant bêta/pharmacologie , Tumeurs du sein triple-négatives/anatomopathologie , Animaux , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Femelle , Humains , Souris , Souris de lignée BALB C , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Pyroptose/effets des médicaments et des substances chimiques , Pyroptose/génétique , ARN double brin/synthèse chimique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Cellules THP-1 , Facteur de croissance transformant bêta/physiologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/thérapie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
17.
PLoS One ; 15(10): e0240403, 2020.
Article de Anglais | MEDLINE | ID: mdl-33119613

RÉSUMÉ

BACKGROUND: Rintatolimod is a selective TLR3 agonist, which has demonstrated clinical activity for ME/CFS in Phase II and Phase III double-blind, placebo-controlled, randomized, multi-site clinical trials. METHODS AND FINDINGS: A hypothesis-based post-hoc analysis of the Intent to Treat (ITT) population diagnosed with ME/CFS from 12 independent clinical sites of a Phase III trial was performed to evaluate the effect of rintatolimod therapy based on disease duration. The clinical activity of rintatolimod was evaluated by exercise treadmill tolerance (ETT) using a modified Bruce protocol. The ITT population (n = 208) was divided into two subsets of symptom duration. Patients with symptom duration of 2-8 years were identified as the Target Subset (n = 75); the remainder (<2 year plus >8 year) were identified as the Non-Target Subset (n = 133). Placebo-adjusted percentage improvements in exercise duration and the vertical rise for the Target Subset (n = 75) were more than twice that of the ITT population. The Non-Target Subset (n = 133) failed to show any clinically significant ETT response to rintatolimod when compared to placebo. Within the Target Subset, 51.2% of rintatolimod-treated patients improved their exercise duration by ≥25% (p = 0.003) despite reduced statistical power from division of the original ITT population into two subsets. CONCLUSION/SIGNIFICANCE: Analysis of ETT from a Phase III trial has identified within the ITT population, a subset of ME/CFS patients with ≥2 fold increased exercise response to rintatolimod. Substantial improvement in physical performance was seen for the majority (51.2%) of these severely debilitated patients who improved exercise duration by ≥25%. This magnitude of exercise improvement was associated with clinically significant enhancements in quality of life. The data indicate that ME/CFS patients have a relatively short disease duration window (<8 years) to expect a significant response to rintatolimod under the dosing conditions utilized in this Phase III clinical trial. These results may have direct relevance to the cognitive impairment and fatigue being experienced by patients clinically recovered from COVID-19 and free of detectable SARS-CoV-2. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00215800.


Sujet(s)
Tolérance à l'effort , Syndrome de fatigue chronique/traitement médicamenteux , Poly I-C/usage thérapeutique , Poly U/usage thérapeutique , Adulte , Méthode en double aveugle , Femelle , Humains , Mâle , Adulte d'âge moyen , Études prospectives , Facteurs temps
18.
Semin Immunol ; 49: 101414, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-33011064

RÉSUMÉ

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Sujet(s)
Antinéoplasiques/pharmacologie , Carboxyméthylcellulose de sodium/analogues et dérivés , Facteurs immunologiques/pharmacologie , Immunomodulation , Poly I-C/immunologie , Poly I-C/pharmacologie , Polylysine/analogues et dérivés , Animaux , Antinéoplasiques/usage thérapeutique , Carboxyméthylcellulose de sodium/pharmacologie , Carboxyméthylcellulose de sodium/usage thérapeutique , Cytokines/métabolisme , Humains , Immunité innée/effets des médicaments et des substances chimiques , Facteurs immunologiques/usage thérapeutique , Immunomodulation/effets des médicaments et des substances chimiques , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/étiologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Poly I-C/usage thérapeutique , Polylysine/immunologie , Polylysine/pharmacologie , Polylysine/usage thérapeutique , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Récepteur de type Toll-3/métabolisme
19.
DNA Cell Biol ; 39(9): 1730-1740, 2020 Sep.
Article de Anglais | MEDLINE | ID: mdl-32580635

RÉSUMÉ

Polyinosinic-polycytidylic acid (PIC) is a potent double-stranded RNA (dsRNA) adjuvant useful in intranasal influenza vaccination. In mice, the intensity and duration of immune responses to PIC correlated with the double-stranded chain length. A rational method to avoid PIC chain extension in PIC production is to use multiple short poly(I) molecules and one long poly(C) molecule for PIC assembly. In this study, we elucidate that a newly developed uPIC100-400 molecule comprising multiple 0.1 kb poly(I) molecules and one 0.4 kb poly(C) molecule effectively enhanced the immune responses in mice, by preventing the challenged viral propagation and inducing hemagglutinin-specific IgA, after intranasal A(H1N1)pdm09 influenza vaccination. Reduced intraperitoneal toxicity of PIC prepared with multiple short poly(I) molecules in mice indicates the widened effective range of uPIC100-400 as an adjuvant. In contrast to uPIC100-400, the PIC molecule comprising multiple 0.05 kb poly(I) molecules failed to elicit mouse mucosal immunity. These results were consistent with TLR3 response but not retinoic acid inducible gene I (RIG-I)-like receptor response in the cell assays, which suggests that the adjuvant effect of PIC in mouse intranasal immunization depends on TLR3 signaling. In conclusion, the double-stranded PIC with reduced toxicity developed in this study would contribute to the development of PIC-adjuvanted vaccines.


Sujet(s)
Adjuvants immunologiques/usage thérapeutique , Inducteurs de l'interféron/usage thérapeutique , Infections à Orthomyxoviridae/immunologie , Poly I-C/usage thérapeutique , Récepteur de type Toll-3/métabolisme , Vaccination/méthodes , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/effets indésirables , Animaux , Cellules cultivées , Femelle , Glycoprotéine hémagglutinine du virus influenza/immunologie , Immunoglobuline A/immunologie , Vaccins antigrippaux/immunologie , Inducteurs de l'interféron/administration et posologie , Inducteurs de l'interféron/effets indésirables , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Infections à Orthomyxoviridae/prévention et contrôle , Poly I-C/administration et posologie , Poly I-C/effets indésirables , Transduction du signal
20.
Expert Opin Biol Ther ; 20(8): 937-946, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32223572

RÉSUMÉ

INTRODUCTION: Activation of innate immune system is a key step to develop anti-tumor immunity. Antigen-presenting dendritic cells (DCs) cross-present tumor-associated antigens to cytotoxic CD8+ T cells (CTLs). Signaling from pattern-recognition receptors (PRRs) in DCs is required to induce tumor-specific CTLs. AREAS COVERED: This review summarizes the properties of PRRs expressed by antigen-presenting DCs, especially TLR3, and provides the recent knowledge of their function in anti-tumor immunity. We also summarize the characteristics of newly-developed TLR3-specific agonist, ARNAX, which efficiently primes DCs to induce anti-tumor immunity without systemic inflammation in mice. EXPERT OPINION: In cancer immunotherapy, the induction of tumor-specific CTLs is significant for tumor regression and to augment the efficacy of PD-1/PD-L1 blockade. Non-inflammatory TLR3 adjuvant ARNAX that can induce tumor-specific CTLs without inducing inflammation benefits cancer immunotherapy. Development of appropriate protocols for ARNAX vaccine therapy would be useful to overcome the PD-1/PD-L1 blockade resistance.


Sujet(s)
Immunothérapie , Tumeurs/thérapie , Récepteur de type Toll-3/agonistes , Animaux , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/usage thérapeutique , Antigène CD274/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Humains , Poly I-C/pharmacologie , Poly I-C/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur de type Toll-3/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE