Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
J Extracell Biol ; 3(7): e166, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39022723

RÉSUMÉ

Natural killer cell-derived extracellular vesicles (NK-EVs) are candidate biotherapeutics against various cancers. However, standardised potency assays are necessary for a reliable assessment of NK-EVs' cytotoxicity. This study aims to thoroughly evaluate a highly sensitive resazurin phenoxazine-based cell viability potency assay (measurement of the cellular redox metabolism) for quantifying the cytotoxicity of NK-EVs against leukaemia K562 cells (suspension model) and breast cancer MDA-MB-231 cells (adherent model) in vitro. The assay was evaluated based on common analytical parameters setforth by regulatory guidelines, including specificity, selectivity,accuracy, precision, linearity, range and stability. Our results revealed that this resazurin-based cell viability potency assay reliably and reproducibly measured a dose-response of NK-EVs' cytotoxic activity against both cancer models. The assay showed precision with 5% and 20% variation for intra-run and inter-run variability. The assay signal showed specificity and selectivity of NK-EVs against cancer target cells, as evidenced by the diminished viability of cancer cells following a 5-hour treatment with NK-EVs, without any detectable interference or background. The linearity analysis of target cancer cells revealed strong linearity for densities of 5000 K562 and 1000 MDA-MB-231 cells per test with a consistent range. Importantly, NK-EVs' dose-response for cytotoxicity showed a strong correlation (|ρ| ∼ 0.8) with the levels of known cytotoxic factors associated with the NK-EVs' corona (FasL, GNLY, GzmB, PFN and IFN-γ), thereby validating the accuracy of the assay. The assay also distinguished cytotoxicity changes in degraded NK-EVs, indicating the ability of the assay to detect the potential loss of sample integrity. Compared to other commonly reported bioassays (i.e., flow cytometry, cell counting, lactate dehydrogenase release assay, DNA-binding reporter assay and confluence assay), our results support this highly sensitive resazurin-based viability potency assay as a high-throughput and quantitative method for assessing NK-EVs' cytotoxicity against both suspension and adherent cancer models for evaluating NK-EVs' biotherapeutics.

2.
J Exp Med ; 221(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38869480

RÉSUMÉ

While conventional wisdom initially postulated that PD-L1 serves as the inert ligand for PD-1, an emerging body of literature suggests that PD-L1 has cell-intrinsic functions in immune and cancer cells. In line with these studies, here we show that engagement of PD-L1 via cellular ligands or agonistic antibodies, including those used in the clinic, potently inhibits the type I interferon pathway in cancer cells. Hampered type I interferon responses in PD-L1-expressing cancer cells resulted in enhanced efficacy of oncolytic viruses in vitro and in vivo. Consistently, PD-L1 expression marked tumor explants from cancer patients that were best infected by oncolytic viruses. Mechanistically, PD-L1 promoted a metabolic shift characterized by enhanced glycolysis rate that resulted in increased lactate production. In turn, lactate inhibited type I IFN responses. In addition to adding mechanistic insight into PD-L1 intrinsic function, our results will also help guide the numerous ongoing efforts to combine PD-L1 antibodies with oncolytic virotherapy in clinical trials.


Sujet(s)
Antigène CD274 , Interféron de type I , Thérapie virale de cancers , Virus oncolytiques , Animaux , Femelle , Humains , Souris , Antigène CD274/métabolisme , Antigène CD274/immunologie , Antigène CD274/génétique , Lignée cellulaire tumorale , Glycolyse , Interféron de type I/métabolisme , Interféron de type I/immunologie , Acide lactique/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/métabolisme , Thérapie virale de cancers/méthodes , Virus oncolytiques/physiologie , Transduction du signal , Mâle
3.
Front Immunol ; 15: 1366197, 2024.
Article de Anglais | MEDLINE | ID: mdl-38601156

RÉSUMÉ

Introduction: Chemotherapy remains the mainstay treatment for triple-negative breast cancer (TNBC) due to the lack of specific targets. Given a modest response of immune checkpoint inhibitors in TNBC patients, improving immunotherapy is an urgent and crucial task in this field. CD73 has emerged as a novel immunotherapeutic target, given its elevated expression on tumor, stromal, and specific immune cells, and its established role in inhibiting anti-cancer immunity. CD73-generated adenosine suppresses immunity by attenuating tumor-infiltrating T- and NK-cell activation, while amplifying regulatory T cell activation. Chemotherapy often leads to increased CD73 expression and activity, further suppressing anti-tumor immunity. While debulking the tumor mass, chemotherapy also enriches heterogenous cancer stem cells (CSC), potentially leading to tumor relapse. Therefore, drugs targeting both CD73, and CSCs hold promise for enhancing chemotherapy efficacy, overcoming treatment resistance, and improving clinical outcomes. However, safe and effective inhibitors of CD73 have not been developed as of now. Methods: We used in silico docking to screen compounds that may be repurposed for inhibiting CD73. The efficacy of these compounds was investigated through flow cytometry, RT-qPCR, CD73 activity, cell viability, tumorsphere formation, and other in vitro functional assays. For assessment of clinical translatability, TNBC patient-derived xenograft organotypic cultures were utilized. We also employed the ovalbumin-expressing AT3 TNBC mouse model to evaluate tumor-specific lymphocyte responses. Results: We identified quercetin and luteolin, currently used as over-the-counter supplements, to have high in silico complementarity with CD73. When quercetin and luteolin were combined with the chemotherapeutic paclitaxel in a triple-drug regimen, we found an effective downregulation in paclitaxel-enhanced CD73 and CSC-promoting pathways YAP and Wnt. We found that CD73 expression was required for the maintenance of CD44highCD24low CSCs, and co-targeting CD73, YAP, and Wnt effectively suppressed the growth of human TNBC cell lines and patient-derived xenograft organotypic cultures. Furthermore, triple-drug combination inhibited paclitaxel-enriched CSCs and simultaneously improved lymphocyte infiltration in syngeneic TNBC mouse tumors. Discussion: Conclusively, our findings elucidate the significance of CSCs in impairing anti-tumor immunity. The high efficacy of our triple-drug regimen in clinically relevant platforms not only underscores the importance for further mechanistic investigations but also paves the way for potential development of new, safe, and cost-effective therapeutic strategies for TNBC.


Sujet(s)
Antigènes CD47 , Tumeurs du sein triple-négatives , Animaux , Humains , Souris , Lignée cellulaire tumorale , Flavonoïdes/pharmacologie , Lutéoline/métabolisme , Cellules souches tumorales/métabolisme , Paclitaxel/usage thérapeutique , Quercétine/pharmacologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Antigènes CD47/antagonistes et inhibiteurs
4.
Nat Cancer ; 5(3): 463-480, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38351181

RÉSUMÉ

Cancer stem cells (CSCs), functionally characterized by self-renewal and tumor-initiating activity, contribute to decreased tumor immunogenicity, while fostering tumor growth and metastasis. Targeting G9a histone methyltransferase (HMTase) effectively blocks CSC functions in colorectal tumors by altering pluripotent-like molecular networks; however, existing molecules directly targeting G9a HMTase activity failed to reach clinical stages due to safety concerns. Using a stem cell-based phenotypic drug-screening pipeline, we identified the dopamine transporter (DAT) antagonist vanoxerine, a compound with previously demonstrated clinical safety, as a cancer-specific downregulator of G9a expression. Here we show that gene silencing and chemical antagonism of DAT impede colorectal CSC functions by repressing G9a expression. Antagonizing DAT also enhanced tumor lymphocytic infiltration by activating endogenous transposable elements and type-I interferon response. Our study unveils the direct implication of the DAT-G9a axis in the maintenance of CSC populations and an approach to improve antitumor immune response in colon tumors.


Sujet(s)
Tumeurs du côlon , Histone-lysine N-methyltransferase , Pipérazines , Humains , Histone-lysine N-methyltransferase/génétique , Transporteurs de la dopamine/métabolisme , Transporteurs de la dopamine/pharmacologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie
5.
Sci Rep ; 14(1): 787, 2024 01 08.
Article de Anglais | MEDLINE | ID: mdl-38191799

RÉSUMÉ

The tumour microenvironment is infiltrated by immunosuppressive cells, such as regulatory T cells (Tregs), which contribute to tumour escape and impede immunotherapy outcomes. Soluble fibrinogen-like protein 2 (sFGL2), a Treg effector protein, inhibits immune cell populations, via receptors FcγRIIB and FcγRIII, leading to downregulation of CD86 in antigen presenting cells and limiting T cell activation. Increased FGL2 expression is associated with tumour progression and poor survival in several different cancers, such as glioblastoma multiforme, lung, renal, liver, colorectal, and prostate cancer. Querying scRNA-seq human cancer data shows FGL2 is produced by cells in the tumour microenvironment (TME), particularly monocytes and macrophages as well as T cells and dendritic cells (DCs), while cancer cells have minimal expression of FGL2. We studied the role of FGL2 exclusively produced by cells in the TME, by leveraging Fgl2 knockout mice. We tested two murine models of cancer in which the role of FGL2 has not been previously studied: epithelial ovarian cancer and melanoma. We show that absence of FGL2 leads to a more activated TME, including activated DCs (CD86+, CD40+) and T cells (CD25+, TIGIT+), as well as demonstrating for the first time that the absence of FGL2 leads to more activated natural killer cells (DNAM-1+, NKG2D+) in the TME. Furthermore, the absence of FGL2 leads to prolonged survival in the B16F10 melanoma model, while the absence of FGL2 synergizes with oncolytic virus to prolong survival in the ID8-p53-/-Brca2-/- ovarian cancer model. In conclusion, targeting FGL2 is a promising cancer treatment strategy alone and in combination immunotherapies.


Sujet(s)
Fibrinogène , Mélanome , Tumeurs de l'ovaire , Animaux , Femelle , Humains , Souris , Cellules présentatrices d'antigène , Carcinome épithélial de l'ovaire , Mélanome/génétique , Mélanome/anatomopathologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Microenvironnement tumoral
6.
Front Immunol ; 14: 1166038, 2023.
Article de Anglais | MEDLINE | ID: mdl-37205115

RÉSUMÉ

Advancements in chimeric antigen receptor engineered T-cell (CAR-T) therapy have revolutionized treatment for several cancer types over the past decade. Despite this success, obstacles including the high price tag, manufacturing complexity, and treatment-associated toxicities have limited the broad application of this therapy. Chimeric antigen receptor engineered natural killer cell (CAR-NK) therapy offers a potential opportunity for a simpler and more affordable "off-the-shelf" treatment, likely with fewer toxicities. Unlike CAR-T, CAR-NK therapies are still in early development, with few clinical trials yet reported. Given the challenges experienced through the development of CAR-T therapies, this review explores what lessons we can apply to build better CAR-NK therapies. In particular, we explore the importance of optimizing the immunochemical properties of the CAR construct, understanding factors leading to cell product persistence, enhancing trafficking of transferred cells to the tumor, ensuring the metabolic fitness of the transferred product, and strategies to avoid tumor escape through antigen loss. We also review trogocytosis, an important emerging challenge that likely equally applies to CAR-T and CAR-NK cells. Finally, we discuss how these limitations are already being addressed in CAR-NK therapies, and what future directions may be possible.


Sujet(s)
Tumeurs , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/effets indésirables , Lymphocytes T , Cellules tueuses naturelles
7.
Nat Commun ; 14(1): 3035, 2023 05 26.
Article de Anglais | MEDLINE | ID: mdl-37236967

RÉSUMÉ

The large coding potential of vaccinia virus (VV) vectors is a defining feature. However, limited regulatory switches are available to control viral replication as well as timing and dosing of transgene expression in order to facilitate safe and efficacious payload delivery. Herein, we adapt drug-controlled gene switches to enable control of virally encoded transgene expression, including systems controlled by the FDA-approved rapamycin and doxycycline. Using ribosome profiling to characterize viral promoter strength, we rationally design fusions of the operator element of different drug-inducible systems with VV promoters to produce synthetic promoters yielding robust inducible expression with undetectable baseline levels. We also generate chimeric synthetic promoters facilitating additional regulatory layers for VV-encoded synthetic transgene networks. The switches are applied to enable inducible expression of fusogenic proteins, dose-controlled delivery of toxic cytokines, and chemical regulation of VV replication. This toolbox enables the precise modulation of transgene circuitry in VV-vectored oncolytic virus design.


Sujet(s)
Thérapie virale de cancers , Virus oncolytiques , Vecteurs génétiques/génétique , Virus de la vaccine/génétique , Virus oncolytiques/génétique , Régions promotrices (génétique)/génétique
8.
Cells ; 13(1)2023 12 29.
Article de Anglais | MEDLINE | ID: mdl-38201278

RÉSUMÉ

Targeted therapy resistance frequently develops in melanoma due to intratumor heterogeneity and epigenetic reprogramming. This also typically induces cross-resistance to immunotherapies. Whether this includes additional modes of therapy has not been fully assessed. We show that co-treatments of MAPKi with VSV-based oncolytics do not function in a synergistic fashion; rather, the MAPKis block infection. Melanoma resistance to vemurafenib further perturbs the cells' ability to be infected by oncolytic viruses. Resistance to vemurafenib can be induced by the loss of SOX10, a common proliferative marker in melanoma. The loss of SOX10 promotes a cross-resistant state by further inhibiting viral infection and replication. Analysis of RNA-seq datasets revealed an upregulation of interferon-stimulated genes (ISGs) in SOX10 knockout populations and targeted therapy-resistant cells. Interestingly, the induction of ISGs appears to be independent of type I IFN production. Overall, our data suggest that the pathway mediating oncolytic resistance is due to the loss of SOX10 during acquired drug resistance in melanoma.


Sujet(s)
Mélanome , Virus oncolytiques , Virus à ARN , Humains , Virus oncolytiques/génétique , Mélanome/thérapie , Vémurafénib , Épigénomique , Interférons , ARN
9.
iScience ; 25(12): 105524, 2022 Dec 22.
Article de Anglais | MEDLINE | ID: mdl-36437876

RÉSUMÉ

SOX10 is a key regulator of melanoma progression and promotes a melanocytic/differentiated state. Here we identified melanoma cell lines lacking SOX10 expression which retain their in vivo growth capabilities. More importantly, we find that SOX10 can regulate T-cell infiltration in melanoma while also decreasing common cancer stem cell (CSC) properties. We show that SOX10 regulates CEACAM1, a surface protein with immunomodulatory properties. SOX10 directly binds to a distal CEACAM1 promoter region approximately 3-4kbps from the CEACAM1 transcriptional start site. Furthermore, we show that a SOX10-CEACAM1 axis can suppress CD8+ T-cell infiltration as well as reduce CSC pool within tumors, leading to reduced tumor growth. Overall, these results identify SOX10 as a direct regulator of CEACAM1, and uncover both a pro- and anti-tumorigenic roles for SOX10 in melanoma.

10.
Cancers (Basel) ; 14(12)2022 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-35740500

RÉSUMÉ

Due to their crucial role in tumor immunity, NK cells have quickly became a prime target for immunotherapies, with the adoptive transfer of NK cells and the use of NK cell engagers quickly moving to the clinical stage. On the other hand, only a few studies have focused on small molecule drugs capable of unleashing NK cells against cancer. In this context, repurposing small molecules is an attractive strategy to identify new immunotherapies from already approved drugs. Here, we developed a new platform to screen small molecule compounds based on a high-throughput luciferase-release cytotoxicity assay. We tested 1200 FDA approved drugs from the Prestwick Chemical Library, to identify compounds that increase NK cells' cytotoxic potential. We found that the antibiotic colistin sulfate increased the cytotoxicity of human NK cells towards cancer cells. The effect of colistin was short lived and was not observed when NK cells were pretreated with the drug, showing how NK cell activity was potentiated only when the compound was present at the time of recognition of cancer cells. Further studies are needed to uncover the mechanism of action and the pre-clinical efficacy of colistin sulfate in mouse cancer models.

11.
Sci Adv ; 8(15): eabj3286, 2022 04 15.
Article de Anglais | MEDLINE | ID: mdl-35417234

RÉSUMÉ

Trogocytosis modulates immune responses, with still unclear underlying molecular mechanisms. Using leukemia mouse models, we found that lymphocytes perform trogocytosis at high rates with tumor cells. While performing trogocytosis, both Natural Killer (NK) and CD8+ T cells acquire the checkpoint receptor PD-1 from leukemia cells. In vitro and in vivo investigation revealed that PD-1 on the surface of NK cells, rather than being endogenously expressed, was derived entirely from leukemia cells in a SLAM receptor-dependent fashion. PD-1 acquired via trogocytosis actively suppressed NK cell antitumor immunity. PD-1 trogocytosis was corroborated in patients with clonal plasma cell disorders, where NK cells that stained for PD-1 also stained for tumor cell markers. Our results, in addition to shedding light on a previously unappreciated mechanism underlying the presence of PD-1 on NK and cytotoxic T cells, reveal the immunoregulatory effect of membrane transfer occurring when immune cells contact tumor cells.


Sujet(s)
Leucémies , Tumeurs , Animaux , Lymphocytes T CD8+ , Humains , Cellules tueuses naturelles , Leucémies/métabolisme , Souris , Tumeurs/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme
12.
Eur J Immunol ; 51(11): 2568-2575, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34347289

RÉSUMÉ

Type 1 innate lymphoid cells (ILC1) are tissue-resident lymphocytes that provide early protection against bacterial and viral infections. Discrete transcriptional states of ILC1 have been identified in homeostatic and pathological contexts. However, whether these states delineate ILC1 with different functional properties is not completely understood. Here, we show that liver ILC1 are heterogeneous for the expression of distinct effector molecules and surface receptors, including granzyme A (GzmA) and CD160, in mice. ILC1 expressing high levels of GzmA are enriched in the liver of adult mice, and represent the main hepatic ILC1 population at birth. However, the heterogeneity of GzmA and CD160 expression in hepatic ILC1 begins perinatally and increases with age. GzmA+ ILC1 differ from NK cells for the limited homeostatic requirements of JAK/STAT signals and the transcription factor Nfil3. Moreover, by employing Rorc(γt)-fate map (fm) reporter mice, we established that ILC3-ILC1 plasticity contributes to delineate the heterogeneity of liver ILC1, with RORγt-fm+ cells skewed toward a GzmA- CD160+ phenotype. Finally, we showed that ILC1 defined by the expression of GzmA and CD160 are characterized by graded cytotoxic potential and ability to produce IFN-γ. In conclusion, our findings help deconvoluting ILC1 heterogeneity and provide evidence for functional diversification of liver ILC1.


Sujet(s)
Foie/cytologie , Foie/immunologie , Sous-populations de lymphocytes/cytologie , Lymphocytes/cytologie , Animaux , Antigènes CD/métabolisme , Protéines liées au GPI/métabolisme , Granzymes/métabolisme , Immunité innée/immunologie , Sous-populations de lymphocytes/immunologie , Sous-populations de lymphocytes/métabolisme , Lymphocytes/immunologie , Lymphocytes/métabolisme , Souris , Récepteurs immunologiques/métabolisme
13.
J Immunother Cancer ; 9(2)2021 02.
Article de Anglais | MEDLINE | ID: mdl-33526607

RÉSUMÉ

Sarcomas are a rare malignancy of mesenchymal tissues, comprizing a plethora of unique subtypes, with more than 60 types. The sheer heterogeneity of disease phenotype makes this a particularly difficult cancer to treat. Radiotherapy, chemotherapy and surgery have been employed for over three decades and, although effective in early disease (stages I-II), in later stages, where metastatic tumors are present, these treatments are less effective. Given the spectacular results obtained by cancer immunotherapy in a variety of solid cancers and leukemias, there is now a great interest in appliying this new realm of therapy for sarcomas. The widespread use of immunotherapy for sarcoma relies on immuno-profiling of subtypes, immunomonitoring for prognosis, preclinical studies and insight into the safety profile of these novel therapies. Herein, we discuss preclinical and clinical data highlighting how immunotherapy is being used in soft tissue sarcoma and bone sarcomas.


Sujet(s)
Immunothérapie/méthodes , Ostéosarcome/traitement médicamenteux , Sarcomes/traitement médicamenteux , Essais cliniques comme sujet , Humains , Stadification tumorale , Ostéosarcome/immunologie , Pronostic , Sarcomes/immunologie , Sarcomes/anatomopathologie , Résultat thérapeutique
14.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118917, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33259860

RÉSUMÉ

Duchenne's muscular dystrophy (DMD) is a severe muscle wasting disorder characterized by the loss of dystrophin expression, muscle necrosis, inflammation and fibrosis. Ongoing muscle regeneration is impaired by persistent cytokine stress, further decreasing muscle function. Patients with DMD rarely survive beyond their early 20s, with cardiac and respiratory dysfunction being the primary cause of death. Despite an increase in our understanding of disease progression as well as promising preclinical animal models for therapeutic intervention, treatment options for muscular dystrophy remain limited and novel therapeutic targets are required. Many reports suggest that the TGFß signalling pathway is activated in dystrophic muscle and contributes to the pathology of DMD in part by impairing the differentiation of myoblasts into mature myofibers. Here, we show that in vitro knockdown of the Ste20-like kinase, SLK, can partially restore myoblast differentiation downstream of TGFß in a Smad2/3 independent manner. In an mdx model, we demonstrate that SLK is expressed at high levels in regenerating myofibers. Muscle-specific deletion of SLK reduced leukocyte infiltration, increased myogenin and utrophin expression and enhanced differentiation. This was accompanied by resistance to eccentric contraction-induced injury in slow fiber type-enriched soleus muscles. Finally, we found that these effects were partially dependent on the upregulation of p38 signalling. Collectively, these results demonstrate that SLK downregulation can restore some aspects of disease progression in DMD.


Sujet(s)
Techniques de knock-out de gènes , Système de signalisation des MAP kinases/génétique , Développement musculaire/génétique , Muscles squelettiques/métabolisme , Myopathie de Duchenne/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Animaux , Cellules cultivées , Modèles animaux de maladie humaine , Chiens , Souris , Souris de lignée mdx , Souris knockout , Myopathie de Duchenne/anatomopathologie , Myoblastes/métabolisme , Myogénine/métabolisme , Protein-Serine-Threonine Kinases/génétique , Facteur de croissance transformant bêta/métabolisme
15.
Front Immunol ; 11: 1512, 2020.
Article de Anglais | MEDLINE | ID: mdl-32655581

RÉSUMÉ

Natural Killer (NK) cells are innate immune responders critical for viral clearance and immunomodulation. Despite their vital role in viral infection, the contribution of NK cells in fighting SARS-CoV-2 has not yet been directly investigated. Insights into pathophysiology and therapeutic opportunities can therefore be inferred from studies assessing NK cell phenotype and function during SARS, MERS, and COVID-19. These studies suggest a reduction in circulating NK cell numbers and/or an exhausted phenotype following infection and hint toward the dampening of NK cell responses by coronaviruses. Reduced circulating NK cell levels and exhaustion may be directly responsible for the progression and severity of COVID-19. Conversely, in light of data linking inflammation with coronavirus disease severity, it is necessary to examine NK cell potential in mediating immunopathology. A common feature of coronavirus infections is that significant morbidity and mortality is associated with lung injury and acute respiratory distress syndrome resulting from an exaggerated immune response, of which NK cells are an important component. In this review, we summarize the current understanding of how NK cells respond in both early and late coronavirus infections, and the implication for ongoing COVID-19 clinical trials. Using this immunological lens, we outline recommendations for therapeutic strategies against COVID-19 in clearing the virus while preventing the harm of immunopathological responses.


Sujet(s)
Transfert adoptif/méthodes , Betacoronavirus/immunologie , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/immunologie , Cellules tueuses naturelles/immunologie , Pneumopathie virale/traitement médicamenteux , Pneumopathie virale/immunologie , Hormones corticosurrénaliennes/usage thérapeutique , Animaux , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Acide ascorbique/usage thérapeutique , Betacoronavirus/effets des médicaments et des substances chimiques , COVID-19 , Infections à coronavirus/mortalité , Infections à coronavirus/virologie , Cytokines/antagonistes et inhibiteurs , Cytokines/métabolisme , Prédisposition aux maladies/immunologie , Humains , Immunité innée/effets des médicaments et des substances chimiques , Immunité innée/immunologie , Mémoire immunologique , Interféron de type I/métabolisme , Interféron de type I/usage thérapeutique , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Souris , Pandémies , Pneumopathie virale/mortalité , Pneumopathie virale/virologie , SARS-CoV-2
17.
Oncogene ; 39(23): 4592-4602, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32393835

RÉSUMÉ

HER2 is overexpressed in 20-30% of all breast cancers and is associated with an invasive disease and poor clinical outcome. The Ste20-like kinase (SLK) is activated downstream of HER2/Neu and is required for efficient epithelial-to-mesenchymal transition, cell cycle progression, and migration in the mammary epithelium. Here we show that loss of SLK in a murine model of HER2/Neu-positive breast cancers significantly accelerates tumor onset and decreases overall survival. Transcriptional profiling of SLK knockout HER2/Neu-derived tumor cells revealed a strong induction in the triple-negative breast cancer marker, Sox10, accompanied by an increase in mammary stem/progenitor activity. Similarly, we demonstrate that SLK and Sox10 expression are inversely correlated in patient samples, with the loss of SLK and acquisition of Sox10 marking the triple-negative subtype. Furthermore, pharmacological inhibition of AKT reduces SLK-null tumor growth in vivo and is rescued by ectopic Sox10 expression, suggesting that Sox10 is a critical regulator of tumor growth downstream of SLK/AKT. These findings highlight a role for SLK in negatively regulating HER2-induced mammary tumorigenesis and provide mechanistic insight into the regulation of Sox10 expression in breast cancer.


Sujet(s)
Transformation cellulaire néoplasique/anatomopathologie , Protein-Serine-Threonine Kinases/génétique , Récepteur ErbB-2/métabolisme , Facteurs de transcription SOX-E/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Animaux , Transition épithélio-mésenchymateuse/génétique , Femelle , Souris , Souris SCID , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Sphéroïdes de cellules , Tumeurs du sein triple-négatives/génétique , Cellules cancéreuses en culture
18.
J Clin Invest ; 129(9): 3499-3510, 2019 09 03.
Article de Anglais | MEDLINE | ID: mdl-31478911

RÉSUMÉ

Natural killer (NK) cells are innate cytotoxic lymphocytes involved in the surveillance and elimination of cancer. As we have learned more and more about the mechanisms NK cells employ to recognize and eliminate tumor cells, and how, in turn, cancer evades NK cell responses, we have gained a clear appreciation that NK cells can be harnessed in cancer immunotherapy. Here, we review the evidence for NK cells' critical role in combating transformed and malignant cells, and how cancer immunotherapies potentiate NK cell responses for therapeutic purposes. We highlight cutting-edge immunotherapeutic strategies in preclinical and clinical development such as adoptive NK cell transfer, chimeric antigen receptor-expressing NK cells (CAR-NKs), bispecific and trispecific killer cell engagers (BiKEs and TriKEs), checkpoint blockade, and oncolytic virotherapy. Further, we describe the challenges that NK cells face (e.g., postsurgical dysfunction) that must be overcome by these therapeutic modalities to achieve cancer clearance.


Sujet(s)
Immunothérapie adoptive , Cellules tueuses naturelles , Tumeurs , Animaux , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/anatomopathologie , Cellules tueuses naturelles/transplantation , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tumeurs/thérapie
19.
J Clin Invest ; 128(10): 4654-4668, 2018 10 01.
Article de Anglais | MEDLINE | ID: mdl-30198904

RÉSUMÉ

Checkpoint blockade immunotherapy targeting the PD-1/PD-L1 inhibitory axis has produced remarkable results in the treatment of several types of cancer. Whereas cytotoxic T cells are known to provide important antitumor effects during checkpoint blockade, certain cancers with low MHC expression are responsive to therapy, suggesting that other immune cell types may also play a role. Here, we employed several mouse models of cancer to investigate the effect of PD-1/PD-L1 blockade on NK cells, a population of cytotoxic innate lymphocytes that also mediate antitumor immunity. We discovered that PD-1 and PD-L1 blockade elicited a strong NK cell response that was indispensable for the full therapeutic effect of immunotherapy. PD-1 was expressed on NK cells within transplantable, spontaneous, and genetically induced mouse tumor models, and PD-L1 expression in cancer cells resulted in reduced NK cell responses and generation of more aggressive tumors in vivo. PD-1 expression was more abundant on NK cells with an activated and more responsive phenotype and did not mark NK cells with an exhausted phenotype. These results demonstrate the importance of the PD-1/PD-L1 axis in inhibiting NK cell responses in vivo and reveal that NK cells, in addition to T cells, mediate the effect of PD-1/PD-L1 blockade immunotherapy.


Sujet(s)
Antigène CD274/immunologie , Immunothérapie , Cellules tueuses naturelles/immunologie , Tumeurs expérimentales/thérapie , Récepteur-1 de mort cellulaire programmée/immunologie , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/génétique , Humains , Cellules K562 , Cellules tueuses naturelles/anatomopathologie , Souris , Souris knockout , Tumeurs expérimentales/génétique , Tumeurs expérimentales/anatomopathologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/génétique
20.
J Immunol ; 197(10): 4127-4136, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-27798146

RÉSUMÉ

Many NK cells express inhibitory receptors that bind self-MHC class I (MHC I) molecules and prevent killing of self-cells, while enabling killing of MHC I-deficient cells. But tolerance also occurs for NK cells that lack inhibitory receptors for self-MHC I, and for all NK cells in MHC I-deficient animals. In both cases, NK cells are unresponsive to MHC I-deficient cells and hyporesponsive when stimulated through activating receptors, suggesting that hyporesponsiveness is responsible for self-tolerance. We generated irradiation chimeras, or carried out adoptive transfers, with wild-type (WT) and/or MHC I-deficient hematopoietic cells in WT or MHC I-deficient C57BL/6 host mice. Unexpectedly, in WT hosts, donor MHC I-deficient hematopoietic cells failed to induce hyporesponsiveness to activating receptor stimulation, but did induce tolerance to MHC I-deficient grafts. Therefore, these two properties of NK cells are separable. Both tolerance and hyporesponsiveness occurred when the host was MHC I deficient. Interestingly, infections of mice or exposure to inflammatory cytokines reversed the tolerance of NK cells that was induced by MHC I-deficient hematopoietic cells, but not the tolerance induced by MHC I-deficient nonhematopoietic cells. These data have implications for successful bone marrow transplantation, and suggest that tolerance induced by hematopoietic cells versus nonhematopoietic cells may be imposed by distinct mechanisms.


Sujet(s)
Tolérance immunitaire , Cellules tueuses naturelles/immunologie , Autotolérance , Transfert adoptif , Animaux , Transplantation de moelle osseuse , Cytokines , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/métabolisme , Cellules tueuses naturelles/classification , Cellules tueuses naturelles/physiologie , Activation des lymphocytes , Souris , Souris de lignée C57BL , Chimère post-radique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE