Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
1.
Front Immunol ; 15: 1355769, 2024.
Article de Anglais | MEDLINE | ID: mdl-38343540

RÉSUMÉ

Tumors educate their environment to prime the occurrence of suppressive cell subsets, which enable tumor evasion and favors tumor progression. Among these, there are the myeloid-derived suppressor cells (MDSCs), their presence being associated with the poor clinical outcome of cancer patients. Tumor-derived prostaglandin E2 (PGE2) is known to mediate MDSC differentiation and the acquisition of pro-tumor features. In myeloid cells, PGE2 signaling is mediated via E-prostanoid receptor type 2 (EP2) and EP4. Although the suppressive role of PGE2 is well established in MDSCs, the role of EP2/4 on human MDSCs or whether EP2/4 modulation can prevent MDSCs suppressive features upon exposure to tumor-derived PGE2 is poorly defined. In this study, using an in vitro model of human monocytic-MDSCs (M-MDSCs) we demonstrate that EP2 and EP4 signaling contribute to the induction of a pro-tumor phenotype and function on M-MDSCs. PGE2 signaling via EP2 and EP4 boosted M-MDSC ability to suppress T and NK cell responses. Combined EP2/4 blockade on M-MDSCs during PGE2 exposure prevented the occurrence of these suppressive features. Additionally, EP2/4 blockade attenuated the suppressive phenotype of M-MDSCs in a 3D coculture with colorectal cancer patient-derived organoids. Together, these results identify the role of tumor-derived PGE2 signaling via EP2 and EP4 in this human M-MDSC model, supporting the therapeutic value of targeting PGE2-EP2/4 axis in M-MDSCs to alleviate immunosuppression and facilitate the development of anti-tumor immunity.


Sujet(s)
Cellules myéloïdes suppressives , Humains , Cellules myéloïdes suppressives/métabolisme , Dinoprostone/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Monocytes
2.
Cell Rep Med ; 5(2): 101386, 2024 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-38242119

RÉSUMÉ

The human dendritic cell (DC) family has recently been expanded by CD1c+CD14+CD163+ DCs, introduced as DC3s. DC3s are found in tumors and peripheral blood of cancer patients. Here, we report elevated frequencies of CD14+ cDC2s, which restore to normal frequencies after tumor resection, in non-small cell lung cancer patients. These CD14+ cDC2s phenotypically resemble DC3s and exhibit increased PD-L1, MERTK, IL-10, and IDO expression, consistent with inferior T cell activation ability compared with CD14- cDC2s. In melanoma patients undergoing CD1c+ DC vaccinations, increased CD1c+CD14+ DC frequencies correlate with reduced survival. We demonstrate conversion of CD5+/-CD1c+CD14- cDC2s to CD14+ cDC2s by tumor-associated factors, whereas monocytes failed to express CD1c under similar conditions. Targeted proteomics identified IL-6 and M-CSF as dominant drivers, and we show that IL-6R and CSF1R inhibition prevents tumor-induced CD14+ cDC2s. Together, this indicates cDC2s as direct pre-cursors of DC3-like CD1c+CD14+ DCs and provides insights into the importance and modulation of CD14+ DC3s in anti-tumor immune responses.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Cellules dendritiques , Tumeurs du poumon/métabolisme , Transduction du signal , Monocytes , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs aux facteurs de croissance hématopoïétique/métabolisme
3.
Eur J Immunol ; 54(3): e2350770, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38088451

RÉSUMÉ

Dendritic cells (DCs) shape adaptive immunity in response to environmental cues such as cytokines or lipid mediators, including prostaglandin E2 (PGE2). In cancer, tumors are known to establish an enriched PGE2 microenvironment. Tumor-derived PGE2 primes regulatory features across immune cells, including DCs, facilitating tumor progression. PGE2 shapes DC function by providing signaling via its two so-called E-prostanoid receptors (EPs) EP2 and EP4. Although studies with monocyte-derived DCs have shown the importance of PGE2 signaling, the role of PGE2-EP2/EP4 on conventional DCs type 2 (cDC2s), is still poorly defined. In this study, we investigated the function of EP2 and EP4 using specific EP antagonists on human cDC2s. Our results show that EP2 and EP4 exhibit different functions in cDC2s, with EP4 modulating the upregulation of activation markers (CD80, CD86, CD83, MHC class II) and the production of IL-10 and IL-23. Furthermore, PGE2-EP4 boosts CCR type 7-based migration as well as a higher T-cell expansion capacity, characterized by the enrichment of suppressive rather than pro-inflammatory T-cell populations. Our findings are relevant to further understanding the role of EP receptors in cDC2s, underscoring the benefit of targeting the PGE2-EP2/4 axis for therapeutic purposes in diseases such as cancer.


Sujet(s)
Dinoprostone , Tumeurs , Humains , Lymphocytes T , Sous-type EP2 des récepteurs des prostaglandines E , Sous-type EP4 des récepteurs des prostaglandines E , Microenvironnement tumoral
4.
Eur J Immunol ; 54(1): e2350616, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37840200

RÉSUMÉ

Dendritic cells (DCs) are essential in antitumor immunity. In humans, three main DC subsets are defined: two types of conventional DCs (cDC1s and cDC2s) and plasmacytoid DCs (pDCs). To study DC subsets in the tumor microenvironment (TME), it is important to correctly identify them in tumor tissues. Tumor-derived DCs are often analyzed in cell suspensions in which spatial information about DCs which can be important to determine their function within the TME is lost. Therefore, we developed the first standardized and optimized multiplex immunohistochemistry panel, simultaneously detecting cDC1s, cDC2s, and pDCs within their tissue context. We report on this panel's development, validation, and quantitative analysis. A multiplex immunohistochemistry panel consisting of CD1c, CD303, X-C motif chemokine receptor 1, CD14, CD19, a tumor marker, and DAPI was established. The ImmuNet machine learning pipeline was trained for the detection of DC subsets. The performance of ImmuNet was compared with conventional cell phenotyping software. Ultimately, frequencies of DC subsets within several tumors were defined. In conclusion, this panel provides a method to study cDC1s, cDC2s, and pDCs in the spatial context of the TME, which supports unraveling their specific roles in antitumor immunity.


Sujet(s)
Tumeurs , Microenvironnement tumoral , Humains , Immunohistochimie , Marqueurs biologiques tumoraux , Tumeurs/métabolisme , Cellules dendritiques
5.
Bio Protoc ; 13(20): e4851, 2023 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-37900109

RÉSUMÉ

Dendritic cells have been investigated for cell-based immunotherapy for various applications. The low abundance of dendritic cells in blood hampers their clinical application, resulting in the use of monocyte-derived dendritic cells as an alternative cell type. Limited knowledge is available regarding blood-circulating human dendritic cells, which can be divided into three subsets: type 2 conventional dendritic cells, type 1 conventional dendritic cells, and plasmacytoid dendritic cells. These subsets exhibit unique and desirable features for dendritic cell-based therapies. To enable efficient and reliable human research on dendritic cell subsets, we developed an efficient isolation protocol for the three human dendritic cell subsets, resulting in pure populations. The sequential steps include peripheral blood mononuclear cell isolation, magnetic-microbead lineage depletion (CD14, CD56, CD3, and CD19), and individual magnetic-microbead isolation of the three human dendritic cell subsets.

6.
J Biol Chem ; 299(11): 105276, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37739035

RÉSUMÉ

Imbalanced immune responses are a prominent hallmark of cancer and autoimmunity. Myeloid cells can be overly suppressive, inhibiting protective immune responses or inactive not controlling autoreactive immune cells. Understanding the mechanisms that induce suppressive myeloid cells, such as myeloid-derived suppressor cells (MDSCs) and tolerogenic dendritic cells (TolDCs), can facilitate the development of immune-restoring therapeutic approaches. MDSCs are a major barrier for effective cancer immunotherapy by suppressing antitumor immune responses in cancer patients. TolDCs are administered to patients to promote immune tolerance with the intent to control autoimmune disease. Here, we investigated the development and suppressive/tolerogenic activity of human MDSCs and TolDCs to gain insight into signaling pathways that drive immunosuppression in these different myeloid subsets. Moreover, monocyte-derived MDSCs (M-MDSCs) generated in vitro were compared to M-MDSCs isolated from head-and-neck squamous cell carcinoma patients. PI3K-AKT signaling was identified as being crucial for the induction of human M-MDSCs. PI3K inhibition prevented the downregulation of HLA-DR and the upregulation of reactive oxygen species and MerTK. In addition, we show that the suppressive activity of dexamethasone-induced TolDCs is induced by ß-catenin-dependent Wnt signaling. The identification of PI3K-AKT and Wnt signal transduction pathways as respective inducers of the immunomodulatory capacity of M-MDSCs and TolDCs provides opportunities to overcome suppressive myeloid cells in cancer patients and optimize therapeutic application of TolDCs. Lastly, the observed similarities between generated- and patient-derived M-MDSCs support the use of in vitro-generated M-MDSCs as powerful model to investigate the functionality of human MDSCs.


Sujet(s)
Cellules dendritiques , Cellules myéloïdes suppressives , Phosphatidylinositol 3-kinases , Transduction du signal , Voie de signalisation Wnt , Humains , Cellules dendritiques/immunologie , Immunomodulation/immunologie , Immunothérapie , Cellules myéloïdes suppressives/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Phosphatidylinositol 3-kinases/immunologie , Protéines proto-oncogènes c-akt/immunologie , Transduction du signal/immunologie , Voie de signalisation Wnt/immunologie , Cellules cancéreuses en culture
7.
Int J Mol Sci ; 24(2)2023 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-36674907

RÉSUMÉ

Prostaglandin E2 (PGE2) is an important maturation mediator for dendritic cells (DCs). However, increased PGE2 levels in the tumor exert immunosuppressive effects on DCs by signaling through two E-Prostanoid (EP) receptors: EP2 and EP4. Blocking EP-receptor signaling of PGE2 with antagonists is currently being investigated for clinical applications to enhance anti-tumor immunity. In this study, we investigated a new delivery approach by encapsulating EP2/EP4 antagonists in polymeric nanoparticles. The nanoparticles were characterized for size, antagonist loading, and release. The efficacy of the encapsulated antagonists to block PGE2 signaling was analyzed using monocyte-derived DCs (moDCs). The obtained nanoparticles were sized between 210 and 260 nm. The encapsulation efficacy of the EP2/EP4 antagonists was 20% and 17%, respectively, and was further increased with the co-encapsulation of both antagonists. The treatment of moDCs with co-encapsulation EP2/EP4 antagonists prevented PGE2-induced co-stimulatory marker expression. Even though both antagonists showed a burst release within 15 min at 37 °C, the nanoparticles executed the immunomodulatory effects on moDCs. In summary, we demonstrate the functionality of EP2/EP4 antagonist-loaded nanoparticles to overcome PGE2 modulation of moDCs.


Sujet(s)
Dinoprostone , Sous-type EP2 des récepteurs des prostaglandines E , Dinoprostone/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Monocytes/métabolisme , Immunomodulation
8.
Eur J Immunol ; 53(11): e2249816, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-36303448

RÉSUMÉ

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. This article provides protocols with top ticks and pitfalls for preparation and successful generation of mouse and human DC from different cellular sources, such as murine BM and HoxB8 cells, as well as human CD34+ cells from cord blood, BM, and peripheral blood or peripheral blood monocytes. We describe murine cDC1, cDC2, and pDC generation with Flt3L and the generation of BM-derived DC with GM-CSF. Protocols for human DC generation focus on CD34+ cell culture on OP9 cell layers for cDC1, cDC2, cDC3, and pDC subset generation and DC generation from peripheral blood monocytes (MoDC). Additional protocols include enrichment of murine DC subsets, CRISPR/Cas9 editing, and clinical grade human DC generation. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.


Sujet(s)
Cellules dendritiques , Monocytes , Animaux , Souris , Humains , Antigènes CD34 , Phénotype , Différenciation cellulaire
9.
J Immunother Cancer ; 10(4)2022 04.
Article de Anglais | MEDLINE | ID: mdl-35428705

RÉSUMÉ

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) are characterized by their ability to induce potent CD8+ T cell responses. In efforts to generate novel vaccination strategies, notably against cancer, human cDC1s emerge as an ideal target to deliver antigens. cDC1s uniquely express XCR1, a seven transmembrane G protein-coupled receptor. Due to its restricted expression and endocytic nature, XCR1 represents an attractive receptor to mediate antigen-delivery to human cDC1s. METHODS: To explore tumor antigen delivery to human cDC1s, we used an engineered version of XCR1-binding lymphotactin (XCL1), XCL1(CC3). Site-specific sortase-mediated transpeptidation was performed to conjugate XCL1(CC3) to an analog of the HLA-A*02:01 epitope of the cancer testis antigen New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1). While poor epitope solubility prevented isolation of stable XCL1-antigen conjugates, incorporation of a single polyethylene glycol (PEG) chain upstream of the epitope-containing peptide enabled generation of soluble XCL1(CC3)-antigen fusion constructs. Binding and chemotactic characteristics of the XCL1-antigen conjugate, as well as its ability to induce antigen-specific CD8+ T cell activation by cDC1s, was assessed. RESULTS: PEGylated XCL1(CC3)-antigen conjugates retained binding to XCR1, and induced cDC1 chemoattraction in vitro. The model epitope was efficiently cross-presented by human cDC1s to activate NY-ESO-1-specific CD8+ T cells. Importantly, vaccine activity was increased by targeting XCR1 at the surface of cDC1s. CONCLUSION: Our results present a novel strategy for the generation of targeted vaccines fused to insoluble antigens. Moreover, our data emphasize the potential of targeting XCR1 at the surface of primary human cDC1s to induce potent CD8+ T cell responses.


Sujet(s)
Antigènes néoplasiques , Vaccins anticancéreux , Cellules dendritiques , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Lymphokines , Protéines membranaires , Sialoglycoprotéines , Antigènes néoplasiques/administration et posologie , Antigènes néoplasiques/immunologie , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Cross-priming , Cellules dendritiques/immunologie , Épitopes/immunologie , Tumeurs de l'oesophage/immunologie , Tumeurs de l'oesophage/thérapie , Carcinome épidermoïde de l'oesophage/immunologie , Carcinome épidermoïde de l'oesophage/thérapie , Humains , Lymphokines/administration et posologie , Lymphokines/immunologie , Mâle , Protéines membranaires/administration et posologie , Protéines membranaires/immunologie , Sialoglycoprotéines/administration et posologie , Sialoglycoprotéines/immunologie
10.
Front Immunol ; 12: 704776, 2021.
Article de Anglais | MEDLINE | ID: mdl-34262573

RÉSUMÉ

Current treatment for patients with non-small-cell lung cancer (NSCLC) is suboptimal since therapy is only effective in a minority of patients and does not always induce a long-lasting response. This highlights the importance of exploring new treatment options. The clinical success of immunotherapy relies on the ability of the immune system to mount an adequate anti-tumor response. The activation of cytotoxic T cells, the effector immune cells responsible for tumor cell killing, is of paramount importance for the immunotherapy success. These cytotoxic T cells are primarily instructed by dendritic cells (DCs). DCs are the most potent antigen-presenting cells (APCs) and are capable of orchestrating a strong anti-cancer immune response. DC function is often suppressed in NSCLC. Therefore, resurrection of DC function is an interesting approach to enhance anti-cancer immune response. Recent data from DC-based treatment studies has given rise to the impression that DC-based treatment cannot induce clinical benefit in NSCLC by itself. However, these are all early-phase studies that were mainly designed to study safety and were not powered to study clinical benefit. The fact that these studies do show that DC-based therapies were well-tolerated and could induce the desired immune responses, indicates that DC-based therapy is still a promising option. Especially combination with other treatment modalities might enhance immunological response and clinical outcome. In this review, we will identify the possibilities from current DC-based treatment trials that could open up new venues to improve future treatment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Cellules dendritiques , Immunothérapie , Tumeurs du poumon , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Cellules dendritiques/immunologie , Cellules dendritiques/transplantation , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie
11.
Oncoimmunology ; 10(1): 1935557, 2021.
Article de Anglais | MEDLINE | ID: mdl-34239773

RÉSUMÉ

Cancer immunotherapies have induced long-lasting responses in cancer patients including those with melanoma and head and neck squamous cell carcinoma (HNSCC). However, the majority of treated patients does not achieve clinical benefit from immunotherapy because of systemic tumor-induced immunosuppression. Monocytic myeloid-derived suppressor cells (M-MDSCs) are implicated as key players in inhibiting anti-tumor immune responses and their frequencies are closely associated with tumor progression. Tumor-derived signals, including signaling via STAT3-COX-2, induce the transformation of monocytic precursors into suppressive M-MDSCs. In a retrospective assessment, we observed that survival of melanoma patients undergoing dendritic cell vaccination was negatively associated with blood M-MDSC levels. Previously, it was shown that platinum-based chemotherapeutics inhibit STAT signaling. Here, we show that cisplatin and oxaliplatin treatment interfere with the development of M-MDSCs, potentially synergizing with cancer immunotherapy. In vitro, subclinical doses of platinum-based drugs prevented the generation of COX-2+ M-MDSCs induced by tumor cells from melanoma patients. This was confirmed in HNSCC patients where intravenous cisplatin treatment drastically lowered M-MDSC frequency while monocyte levels remained stable. In treated patients, expression of COX-2 and arginase-1 in M-MDSCs was significantly decreased after two rounds of cisplatin, indicating inhibition of STAT3 signaling. In line, the capacity of M-MDSCs to inhibit activated T cell responses ex vivo was significantly decreased after patients received cisplatin. These results show that platinum-based chemotherapeutics inhibit the expansion and suppressive activity of M-MDSCs in vitro and in cancer patients. Therefore, platinum-based drugs have the potential to enhance response rates of immunotherapy by overcoming M-MDSC-mediated immunosuppression.


Sujet(s)
Mélanome , Cellules myéloïdes suppressives , Cisplatine/pharmacologie , Humains , Mélanome/traitement médicamenteux , Monocytes , Études rétrospectives
12.
J Autoimmun ; 120: 102645, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33901801

RÉSUMÉ

Current treatment for patients with autoimmune disorders including rheumatoid arthritis, multiple sclerosis and type 1 diabetes, often consists of long-term drug regimens that broadly dampen immune responses. These non-specific treatments are frequently associated with severe side effects creating an urgent need for safer and more effective therapy to promote peripheral tolerance in autoimmune diseases. Cell-based immunotherapy may offer an encouraging alternative, where tolerogenic CD14+ myeloid cells are infused to inhibit autoreactive effector cells. In this review, we compared in depth three promising tolerogenic CD14+ candidates for the treatment of autoimmune disease: 1) tolerogenic dendritic cells, 2) monocytic myeloid-derived suppressor cells and 3) CD14+ type 2 conventional dendritic cells. TolDC-based therapy has entered clinical testing whereas evidence from the latter two cell types m-MDSCs and CD14+ cDC2s is predominantly coming from cancer immunology research. These three cell types have distinct cellular properties and immunosuppressive mechanisms offering unique opportunities to be explored. However, these cells differ in stage of development towards immunotherapy each facing additional hurdles. Therefore, we speculate on the potential benefits and risks of these cell types as novel cell-based immunotherapies to control autoimmune disease in patients.


Sujet(s)
Maladies auto-immunes/étiologie , Maladies auto-immunes/métabolisme , Auto-immunité , Tolérance immunitaire , Antigènes CD14/métabolisme , Cellules myéloïdes/immunologie , Cellules myéloïdes/métabolisme , Animaux , Maladies auto-immunes/anatomopathologie , Marqueurs biologiques , Études cliniques comme sujet , Association thérapeutique , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Prise en charge de la maladie , Modèles animaux de maladie humaine , Prédisposition aux maladies , Humains , Monocytes/immunologie , Monocytes/métabolisme , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Résultat thérapeutique
13.
Eur J Immunol ; 51(6): 1494-1504, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33675038

RÉSUMÉ

Dendritic cells (DCs) are key regulators of the immune system that shape T cell responses. Regulation of T cell induction by DCs may occur via the intracellular enzyme indoleamine 2,3-dioxygenase 1 (IDO), which catalyzes conversion of the essential amino acid tryptophan into kynurenine. Here, we examined the role of IDO in human peripheral blood plasmacytoid DCs (pDCs), and type 1 and type 2 conventional DCs (cDC1s and cDC2s). Our data demonstrate that under homeostatic conditions, IDO is selectively expressed by cDC1s. IFN-γ or TLR ligation further increases IDO expression in cDC1s and induces modest expression of the enzyme in cDC2s, but not pDCs. IDO expressed by conventional DCs is functionally active as measured by kynurenine production. Furthermore, IDO activity in TLR-stimulated cDC1s and cDC2s inhibits T cell proliferation in settings were DC-T cell cell-cell contact does not play a role. Selective inhibition of IDO1 with epacadostat, an inhibitor currently tested in clinical trials, rescued T cell proliferation without affecting DC maturation status or their ability to cross-present soluble antigen. Our findings provide new insights into the functional specialization of human blood DC subsets and suggest a possible synergistic enhancement of therapeutic efficacy by combining DC-based cancer vaccines with IDO inhibition.


Sujet(s)
Cellules dendritiques/immunologie , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Lymphocytes T/immunologie , Vaccins anticancéreux , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Techniques de coculture , Cross-priming , Régulation de l'expression des gènes , Homéostasie , Humains , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Indoleamine-pyrrole 2,3,-dioxygenase/génétique , Activation des lymphocytes , Thérapie moléculaire ciblée , Spécificité d'organe , Oximes/pharmacologie , Phénotype , Sulfonamides/pharmacologie
14.
Cell Rep ; 30(4): 1027-1038.e4, 2020 01 28.
Article de Anglais | MEDLINE | ID: mdl-31995747

RÉSUMÉ

Plasmacytoid dendritic cells (pDCs) and type 2 conventional dendritic cells (cDC2s) are currently under evaluation for use in cancer vaccines. Although both DC subsets can activate adaptive and innate lymphocytes, their capacity to recruit such cells is rarely considered. Here, we show that pDCs and cDC2s display a striking difference in chemokine secretion, which correlates with the recruitment of distinct types of immune effector cells. Activated pDCs express high levels of CXCR3 ligands and attract more CD8+ T cells, CD56+ T cells, and γδ T cells in vitro, compared to cDC2s. Skin from melanoma patients shows an influx of immune effector cells following intradermal vaccination with pDCs or cDC2s, with pDCs inducing the strongest influx of lymphocytes known to possess cytolytic activity. These findings suggest that combining both DC subsets could unite the preferred chemoattractive properties of pDCs with the superior T cell priming properties of cDC2s to ultimately enhance vaccine efficacy.


Sujet(s)
Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/immunologie , Chimiokines/métabolisme , Cellules dendritiques/immunologie , Mélanome/immunologie , Récepteurs CXCR3/métabolisme , Lymphocytes T/immunologie , Différenciation cellulaire/immunologie , Mouvement cellulaire/immunologie , Cellules cultivées , Chimiokines/immunologie , Cellules dendritiques/cytologie , Cellules dendritiques/métabolisme , Régulation de l'expression des gènes/immunologie , Humains , Immunité innée , Activation des lymphocytes , Récepteurs CXCR3/immunologie , Tumeurs cutanées/immunologie
15.
Front Immunol ; 11: 631713, 2020.
Article de Anglais | MEDLINE | ID: mdl-33679726

RÉSUMÉ

Immunotherapeutic approaches have revolutionized the treatment of several diseases such as cancer. The main goal of immunotherapy for cancer is to modulate the anti-tumor immune responses by favoring the recognition and destruction of tumor cells. Recently, a better understanding of the suppressive effect of the tumor microenvironment (TME) on immune cells, indicates that restoring the suppressive effect of the TME is crucial for an efficient immunotherapy. Natural killer (NK) cells and dendritic cells (DCs) are cell types that are currently administered to cancer patients. NK cells are used because of their ability to kill tumor cells directly via cytotoxic granzymes. DCs are employed to enhance anti-tumor T cell responses based on their ability to present antigens and induce tumor-antigen specific CD8+ T cell responses. In preclinical models, a particular DC subset, conventional type 1 DCs (cDC1s) is shown to be specialized in cross-presenting extracellular antigens to CD8+ T cells. This feature makes them a promising DC subset for cancer treatment. Within the TME, cDC1s show a bidirectional cross-talk with NK cells, resulting in a higher cDC1 recruitment, differentiation, and maturation as well as activation and stimulation of NK cells. Consequently, the presence of cDC1s and NK cells within the TME might be of utmost importance for the success of immunotherapy. In this review, we discuss the function of cDC1s and NK cells, their bidirectional cross-talk and potential strategies that could improve cancer immunotherapy.


Sujet(s)
Communication cellulaire/immunologie , Cellules dendritiques/immunologie , Cellules tueuses naturelles/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Microenvironnement tumoral/immunologie , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Cellules dendritiques/anatomopathologie , Humains , Cellules tueuses naturelles/anatomopathologie , Tumeurs/anatomopathologie
16.
J Immunol Res ; 2019: 7458238, 2019.
Article de Anglais | MEDLINE | ID: mdl-31309123

RÉSUMÉ

The STAT signaling pathway is important in dendritic cell (DC) development and function. Tumor cells can induce STAT signaling, thereby inhibiting DC maturation and immunostimulatory functions, leading to hampered efficacy of DC-based immunotherapies. Platinum-based chemotherapeutics can inhibit STAT signaling, thereby making them an interesting tool to improve DC development and function. In this study, we provide a comprehensive overview of STAT expression and phosphorylation during DC differentiation and maturation and investigate the effects of platinum drugs on STAT signaling during these processes. Monocytes were differentiated into monocyte-derived DCs (moDCs) with IL-4 and GM-CSF and matured with cytokines or TLR ligands. STAT expression and phosphorylation were analyzed by western blotting, and moDC viability and phenotype were analyzed by flow cytometry. Platinum drugs were added at day 3 of differentiation or at the start of maturation to investigate regulation of the STAT signaling pathway. All STAT proteins were expressed during moDC differentiation and STAT1, STAT5, and STAT6 were phosphorylated. No significant changes occurred in the expression and phosphorylation state of the STAT proteins during differentiation. After maturation with TLR ligands, the expression of STAT1 increased, but other STAT proteins were not affected. Phosphorylation of STAT1 and STAT3 increased during maturation, where TLR ligands induced significantly higher levels of phosphorylation than cytokines. Platinum drugs cisplatin and oxaliplatin significantly inhibited phosphorylation of STAT6 during differentiation and maturation. Treatment did not affect the phenotype or viability of the cells. As STAT6 is an important regulator of DC function, these findings suggest a role for platinum-based chemotherapeutics to enhance DC function via inhibition of STAT signaling, thereby potentially enhancing efficacy of DC-based immunotherapies.


Sujet(s)
Antinéoplasiques/pharmacologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme , Platine/pharmacologie , Facteurs de transcription STAT/métabolisme , Marqueurs biologiques , Différenciation cellulaire , Cellules dendritiques/cytologie , Cellules dendritiques/immunologie , Expression des gènes , Humains , Immunophénotypage , Phosphorylation , Facteurs de transcription STAT/génétique , Transduction du signal
17.
Proc Natl Acad Sci U S A ; 116(17): 8463-8470, 2019 04 23.
Article de Anglais | MEDLINE | ID: mdl-30962374

RÉSUMÉ

There are adaptive T-cell and antibody autoimmune responses to myelin-derived peptides in multiple sclerosis (MS) and to aquaporin-4 (AQP4) in neuromyelitis optica spectrum disorders (NMOSDs). Strategies aimed at antigen-specific tolerance to these autoantigens are thus indicated for these diseases. One approach involves induction of tolerance with engineered dendritic cells (tolDCs) loaded with specific antigens. We conducted an in-human phase 1b clinical trial testing increasing concentrations of autologous tolDCs loaded with peptides from various myelin proteins and from AQP4. We tested this approach in 12 patients, 8 with MS and 4 with NMOSD. The primary end point was the safety and tolerability, while secondary end points were clinical outcomes (relapses and disability), imaging (MRI and optical coherence tomography), and immunological responses. Therapy with tolDCs was well tolerated, without serious adverse events and with no therapy-related reactions. Patients remained stable clinically in terms of relapse, disability, and in various measurements using imaging. We observed a significant increase in the production of IL-10 levels in PBMCs stimulated with the peptides as well as an increase in the frequency of a regulatory T cell, known as Tr1, by week 12 of follow-up. In this phase 1b trial, we concluded that the i.v. administration of peptide-loaded dendritic cells is safe and feasible. Elicitation of specific IL-10 production by peptide-specific T cells in MS and NMOSD patients indicates that a key element in antigen specific tolerance is activated with this approach. The results warrant further clinical testing in larger trials.


Sujet(s)
Thérapie cellulaire et tissulaire/méthodes , Cellules dendritiques , Tolérance immunitaire , Sclérose en plaques/thérapie , Neuromyélite optique/thérapie , Adulte , Aquaporine-4/génétique , Thérapie cellulaire et tissulaire/effets indésirables , Cellules cultivées , Cellules dendritiques/métabolisme , Cellules dendritiques/transplantation , Femelle , Humains , Tolérance immunitaire/génétique , Tolérance immunitaire/immunologie , Tolérance immunitaire/physiologie , Immunothérapie , Interleukine-10/métabolisme , Mâle , Adulte d'âge moyen , Sclérose en plaques/immunologie , Protéines de la myéline/génétique , Neuromyélite optique/immunologie , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/métabolisme , Lymphocytes T régulateurs/métabolisme
18.
Cancer Immunol Immunother ; 67(9): 1425-1436, 2018 Sep.
Article de Anglais | MEDLINE | ID: mdl-30019146

RÉSUMÉ

There has recently been a paradigm shift in the field of dendritic cell (DC)-based immunotherapy, where several clinical studies have confirmed the feasibility and advantageousness of using directly isolated human blood-derived DCs over in vitro differentiated subsets. There are two major DC subsets found in blood; plasmacytoid DCs (pDCs) and myeloid DCs (mDCs), and both have been tested clinically. CD1c+ mDCs are highly efficient antigen-presenting cells that have the ability to secrete IL-12p70, while pDCs are professional IFN-α-secreting cells that are shown to induce innate immune responses in melanoma patients. Hence, combining mDCs and pDCs poses as an attractive, multi-functional vaccine approach. However, type I IFNs have been reported to inhibit IL-12p70 production and mDC-induced T-cell activation. In this study, we investigate the effect of IFN-α on mDC maturation and function. We demonstrate that both recombinant IFN-α and activated pDCs strongly enhance mDC maturation and increase IL-12p70 production. Co-cultured mDCs and pDCs additionally have beneficial effect on NK and NKT-cell activation and also enhances IFN-γ production by allogeneic T cells. In contrast, the presence of type I IFNs reduces the proliferative T-cell response. The mere presence of a small fraction of activated pDCs is sufficient for these effects and the required ratio between the subsets is non-stringent. Taken together, these results support the usage of mDCs and pDCs combined into one immunotherapeutic vaccine with broad immunostimulatory features.


Sujet(s)
Cellules dendritiques/immunologie , Interféron de type I/pharmacologie , Interleukine-12/biosynthèse , Cellules myéloïdes/immunologie , Antigènes CD1/immunologie , Antigènes CD1/pharmacologie , Techniques de coculture , Cellules dendritiques/cytologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Glycoprotéines/immunologie , Glycoprotéines/pharmacologie , Humains , Immunité innée , Interféron de type I/immunologie , Interféron alpha-2 , Interféron alpha/immunologie , Interféron alpha/pharmacologie , Interféron gamma/biosynthèse , Interféron gamma/immunologie , Interleukine-12/immunologie , Interleukine-12/pharmacologie , Activation des lymphocytes , Cellules myéloïdes/cytologie , Cellules myéloïdes/effets des médicaments et des substances chimiques , Quinoléines/pharmacologie , Protéines recombinantes/immunologie , Protéines recombinantes/pharmacologie , Lymphocytes T/cytologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie
19.
Front Immunol ; 9: 1169, 2018.
Article de Anglais | MEDLINE | ID: mdl-29904379

RÉSUMÉ

The identification of activated T-lymphocytes restricted to myelin-derived immunogenic peptides in multiple sclerosis (MS) and aquaporin-4 water channel in neuromyelitis optica (NMO) in the blood of patients opened the possibility for developing highly selective and disease-specific therapeutic approaches. Antigen presenting cells and in particular dendritic cells (DCs) represent a strategy to inhibit pro-inflammatory T helper cells. DCs are located in peripheral and lymphoid tissues and are essential for homeostasis of T cell-dependent immune responses. The expression of a particular set of receptors involved in pathogen recognition confers to DCs the property to initiate immune responses. However, in the absence of danger signals different DC subsets have been revealed to induce active tolerance by inducing regulatory T cells, inhibiting pro-inflammatory T helper cells responses or both. Interestingly, several protocols to generate clinical-grade tolerogenic DC (Tol-DC) in vitro have been described, offering the possibility to restore the homeostasis to central nervous system-related antigens. In this review, we discuss about different DC subsets and their role in tolerance induction, the different protocols to generate Tol-DCs and preclinical studies in animal models as well as describe recent characterization of Tol-DCs for clinical application in autoimmune diseases and in particular in MS and NMO patients. In addition, we discuss the clinical trials ongoing based on Tol-DCs to treat different autoimmune diseases.


Sujet(s)
Cellules dendritiques/immunologie , Immunothérapie/méthodes , Sclérose en plaques/thérapie , Neuromyélite optique/thérapie , Lymphocytes T auxiliaires/immunologie , Alarmines/métabolisme , Animaux , Présentation d'antigène , Aquaporine-4/immunologie , Autoantigènes/immunologie , Cellules dendritiques/transplantation , Humains , Tolérance immunitaire , Immunothérapie/tendances , Activation des lymphocytes , Gaine de myéline/immunologie , Spécificité antigénique des récepteurs des lymphocytes T
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE