Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 25
Filtrer
1.
Cell ; 187(11): 2690-2702.e17, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38723627

RÉSUMÉ

The quality and quantity of tumor-infiltrating lymphocytes, particularly CD8+ T cells, are important parameters for the control of tumor growth and response to immunotherapy. Here, we show in murine and human cancers that these parameters exhibit circadian oscillations, driven by both the endogenous circadian clock of leukocytes and rhythmic leukocyte infiltration, which depends on the circadian clock of endothelial cells in the tumor microenvironment. To harness these rhythms therapeutically, we demonstrate that efficacy of chimeric antigen receptor T cell therapy and immune checkpoint blockade can be improved by adjusting the time of treatment during the day. Furthermore, time-of-day-dependent T cell signatures in murine tumor models predict overall survival in patients with melanoma and correlate with response to anti-PD-1 therapy. Our data demonstrate the functional significance of circadian dynamics in the tumor microenvironment and suggest the importance of leveraging these features for improving future clinical trial design and patient care.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie , Lymphocytes TIL , Souris de lignée C57BL , Microenvironnement tumoral , Animaux , Humains , Souris , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Horloges circadiennes , Rythme circadien , Cellules endothéliales/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Lymphocytes TIL/immunologie , Mélanome/immunologie , Mélanome/thérapie , Mélanome/anatomopathologie , Microenvironnement tumoral/immunologie
2.
Head Neck Pathol ; 17(4): 969-975, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37930471

RÉSUMÉ

Immune checkpoint inhibitors have improved the outcome of patients diagnosed with inoperable recurrent or metastatic head and neck squamous cell carcinoma. However, as only a subset of head and neck cancer patients benefit from this treatment, biomarkers predicting treatment response help guide physicians in their clinical decision-making. PD-L1 expression assessed by immunohistochemistry is the single most clinically relevant biomarker predicting response to PD-1-blocking antibodies. Here, we discuss in which clinical context assessment of PD-L1 expression is instrumental for the choice of therapy, how pathologists score it, and how it affects the approval of anti-PD-1 antibodies. Furthermore, we discuss the heterogeneity of PD-L1 expression and review technical aspects of determining this prominent biomarker-knowledge that might influence clinical decision-making.


Sujet(s)
Antigène CD274 , Tumeurs de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou , Antigène CD274/métabolisme , Immunohistochimie , Marqueurs biologiques
3.
Science ; 381(6657): 515-524, 2023 08 04.
Article de Anglais | MEDLINE | ID: mdl-37535729

RÉSUMÉ

Tumor microenvironments (TMEs) influence cancer progression but are complex and often differ between patients. Considering that microenvironment variations may reveal rules governing intratumoral cellular programs and disease outcome, we focused on tumor-to-tumor variation to examine 52 head and neck squamous cell carcinomas. We found that macrophage polarity-defined by CXCL9 and SPP1 (CS) expression but not by conventional M1 and M2 markers-had a noticeably strong prognostic association. CS macrophage polarity also identified a highly coordinated network of either pro- or antitumor variables, which involved each tumor-associated cell type and was spatially organized. We extended these findings to other cancer indications. Overall, these results suggest that, despite their complexity, TMEs coordinate coherent responses that control human cancers and for which CS macrophage polarity is a relevant yet simple variable.


Sujet(s)
Polarité de la cellule , Chimiokine CXCL9 , Tumeurs de la tête et du cou , Macrophages , Ostéopontine , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Chimiokine CXCL9/analyse , Chimiokine CXCL9/métabolisme , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/anatomopathologie , Macrophages/immunologie , Ostéopontine/analyse , Ostéopontine/métabolisme , Pronostic , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Polarité de la cellule/immunologie
4.
J Immunother Cancer ; 11(5)2023 05.
Article de Anglais | MEDLINE | ID: mdl-37208130

RÉSUMÉ

BACKGROUND: Although immune checkpoint inhibitors have been a breakthrough in clinical oncology, these therapies fail to produce durable responses in a significant fraction of patients. This lack of long-term efficacy may be due to a poor pre-existing network linking innate and adaptive immunity. Here, we present an antisense oligonucleotide (ASO)-based strategy that dually targets toll-like receptor 9 (TLR9) and programmed cell death ligand 1 (PD-L1), aiming to overcome resistance to anti-PD-L1 monoclonal therapy. METHODS: We designed a high-affinity immunomodulatory IM-TLR9:PD-L1-ASO antisense oligonucleotide (hereafter, IM-T9P1-ASO) targeting mouse PD-L1 messenger RNA and activating TLR9. Then, we performed in vitro and in vivo studies to validate the IM-T9P1-ASO activity, efficacy, and biological effects in tumors and draining lymph nodes. We also performed intravital imaging to study IM-T9P1-ASO pharmacokinetics in the tumor. RESULTS: IM-T9P1-ASO therapy, unlike PD-L1 antibody therapy, results in durable antitumor responses in multiple mouse cancer models. Mechanistically, IM-T9P1-ASO activates a state of tumor-associated dendritic cells (DCs), referred to here as DC3s, which have potent antitumor potential but express the PD-L1 checkpoint. IM-T9P1-ASO has two roles: it triggers the expansion of DC3s by engaging with TLR9 and downregulates PD-L1, thereby unleashing the antitumor functions of DC3s. This dual action leads to tumor rejection by T cells. The antitumor efficacy of IM-T9P1-ASO depends on the antitumor cytokine interleukin-12 (IL-12), produced by DC3s, and Batf3, a transcription factor required for DC development. CONCLUSIONS: By simultaneously targeting TLR9 and PD-L1, IM-T9P1-ASO amplifies antitumor responses via DC activation, leading to sustained therapeutic efficacy in mice. By highlighting differences and similarities between mouse and human DCs, this study could serve to develop similar therapeutic strategies for patients with cancer.


Sujet(s)
Tumeurs , Récepteur-9 de type Toll-like , Humains , Souris , Animaux , Récepteur-9 de type Toll-like/métabolisme , Immunothérapie/méthodes , Tumeurs/traitement médicamenteux , Oligonucléotides antisens , Cellules dendritiques
5.
Cell ; 186(7): 1448-1464.e20, 2023 03 30.
Article de Anglais | MEDLINE | ID: mdl-37001504

RÉSUMÉ

Neutrophils accumulate in solid tumors, and their abundance correlates with poor prognosis. Neutrophils are not homogeneous, however, and could play different roles in cancer therapy. Here, we investigate the role of neutrophils in immunotherapy, leading to tumor control. We show that successful therapies acutely expanded tumor neutrophil numbers. This expansion could be attributed to a Sellhi state rather than to other neutrophils that accelerate tumor progression. Therapy-elicited neutrophils acquired an interferon gene signature, also seen in human patients, and appeared essential for successful therapy, as loss of the interferon-responsive transcription factor IRF1 in neutrophils led to failure of immunotherapy. The neutrophil response depended on key components of anti-tumor immunity, including BATF3-dependent DCs, IL-12, and IFNγ. In addition, we found that a therapy-elicited systemic neutrophil response positively correlated with disease outcome in lung cancer patients. Thus, we establish a crucial role of a neutrophil state in mediating effective cancer therapy.


Sujet(s)
Tumeurs du poumon , Granulocytes neutrophiles , Humains , Tumeurs du poumon/génétique , Transduction du signal/génétique , Immunothérapie , Interférons
6.
Nature ; 614(7946): 136-143, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36470303

RÉSUMÉ

The process of cancer immunosurveillance is a mechanism of tumour suppression that can protect the host from cancer development throughout its lifetime1,2. However, it is unknown whether the effectiveness of cancer immunosurveillance fluctuates over a single day. Here we demonstrate that the initial time of day of tumour engraftment dictates the ensuing tumour size across mouse cancer models. Using immunodeficient mice as well as mice lacking lineage-specific circadian functions, we show that dendritic cells (DCs) and CD8+ T cells exert circadian anti-tumour functions that control melanoma volume. Specifically, we find that rhythmic trafficking of DCs to the tumour draining lymph node governs a circadian response of tumour-antigen-specific CD8+ T cells that is dependent on the circadian expression of the co-stimulatory molecule CD80. As a consequence, cancer immunotherapy is more effective when synchronized with DC functions, shows circadian outcomes in mice and suggests similar effects in humans. These data demonstrate that the circadian rhythms of anti-tumour immune components are not only critical for controlling tumour size but can also be of therapeutic relevance.


Sujet(s)
Lymphocytes T CD8+ , Rythme circadien , Cellules dendritiques , Mélanome , Animaux , Humains , Souris , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Immunothérapie/méthodes , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/thérapie , Souris de lignée C57BL , Antigène CD80 , Antigènes néoplasiques/immunologie , Noeuds lymphatiques , Rythme circadien/immunologie
7.
Cancer Immunol Res ; 10(1): 40-55, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34795032

RÉSUMÉ

Macrophages often abound within tumors, express colony-stimulating factor 1 receptor (CSF1R), and are linked to adverse patient survival. Drugs blocking CSF1R signaling have been used to suppress tumor-promoting macrophage responses; however, their mechanisms of action remain incompletely understood. Here, we assessed the lung tumor immune microenvironment in mice treated with BLZ945, a prototypical small-molecule CSF1R inhibitor, using single-cell RNA sequencing and mechanistic validation approaches. We showed that tumor control was not caused by CSF1R+ cell depletion; instead, CSF1R targeting reshaped the CSF1R+ cell landscape, which unlocked cross-talk between antitumoral CSF1R- cells. These cells included IFNγ-producing natural killer and T cells, and an IL12-producing dendritic cell subset, denoted as DC3, which were all necessary for CSF1R inhibitor-mediated lung tumor control. These data indicate that CSF1R targeting can activate a cardinal cross-talk between cells that are not macrophages and that are essential to mediate the effects of T cell-targeted immunotherapies and promote antitumor immunity.See related Spotlight by Burrello and de Visser, p. 4.


Sujet(s)
Cellules dendritiques/immunologie , Immunothérapie/méthodes , Interféron gamma/métabolisme , Interleukine-12/métabolisme , Tumeurs du poumon/thérapie , Animaux , Benzothiazoles/pharmacologie , Lignée cellulaire tumorale , Femelle , Tumeurs du poumon/immunologie , Souris , Souris de lignée C57BL , Acides picoliniques/pharmacologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Cancer Rep (Hoboken) ; 5(3): e1491, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34231337

RÉSUMÉ

BACKGROUND: Secretory carcinoma is a more recently described subtype of salivary gland carcinoma that may pose diagnostic challenges and frequently harbors NTRK fusions that may successfully be targeted by TRK inhibitors in advanced disease. CASE: We present the case of a female patient with secretory carcinoma arising in the base of tongue with persistent disease after debulking surgery and definitive chemoradiation. As an alternative to salvage surgery, which would have resulted in significant impairment of swallowing and speech function, a targeted therapy with the TRK-inhibitor larotrectinib against an identified ETV6-NTRK3 fusion product was initiated. Larotrectinib treatment has been well tolerated, resulted in durable complete response and the patient maintains good swallowing and speech function. CONCLUSION: The presented case underscores the importance of the accurate diagnosis of secretory carcinoma. It further highlights the impact of molecular testing as targeted therapies may play an important role in the management of advanced salivary gland cancers.


Sujet(s)
Tumeurs des glandes salivaires , Glandes salivaires mineures , Tumeurs du sein , Carcinomes , Femelle , Humains , Immunohistochimie , Protéines de fusion oncogènes , Tumeurs des glandes salivaires/diagnostic , Tumeurs des glandes salivaires/génétique , Tumeurs des glandes salivaires/thérapie , Glandes salivaires mineures/anatomopathologie
9.
Dev Cell ; 56(23): 3203-3221.e11, 2021 12 06.
Article de Anglais | MEDLINE | ID: mdl-34847378

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) is a transient, reversible process of cell de-differentiation where cancer cells transit between various stages of an EMT continuum, including epithelial, partial EMT, and mesenchymal cell states. We have employed Tamoxifen-inducible dual recombinase lineage tracing systems combined with live imaging and 5-cell RNA sequencing to track cancer cells undergoing partial or full EMT in the MMTV-PyMT mouse model of metastatic breast cancer. In primary tumors, cancer cells infrequently undergo EMT and mostly transition between epithelial and partial EMT states but rarely reach full EMT. Cells undergoing partial EMT contribute to lung metastasis and chemoresistance, whereas full EMT cells mostly retain a mesenchymal phenotype and fail to colonize the lungs. However, full EMT cancer cells are enriched in recurrent tumors upon chemotherapy. Hence, cancer cells in various stages of the EMT continuum differentially contribute to hallmarks of breast cancer malignancy, such as tumor invasion, metastasis, and chemoresistance.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/anatomopathologie , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/secondaire , Animaux , Antinéoplasiques/pharmacologie , Apoptose , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Mouvement cellulaire , Prolifération cellulaire , Femelle , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Souris , Souris de lignée NOD , Souris SCID , Invasion tumorale , Analyse de séquence d'ARN , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Sci Immunol ; 6(61)2021 07 02.
Article de Anglais | MEDLINE | ID: mdl-34215680

RÉSUMÉ

Immunotherapy is revolutionizing cancer treatment but is often restricted by toxicities. What distinguishes adverse events from concomitant antitumor reactions is poorly understood. Here, using anti-CD40 treatment in mice as a model of TH1-promoting immunotherapy, we showed that liver macrophages promoted local immune-related adverse events. Mechanistically, tissue-resident Kupffer cells mediated liver toxicity by sensing lymphocyte-derived IFN-γ and subsequently producing IL-12. Conversely, dendritic cells were dispensable for toxicity but drove tumor control. IL-12 and IFN-γ were not toxic themselves but prompted a neutrophil response that determined the severity of tissue damage. We observed activation of similar inflammatory pathways after anti-PD-1 and anti-CTLA-4 immunotherapies in mice and humans. These findings implicated macrophages and neutrophils as mediators and effectors of aberrant inflammation in TH1-promoting immunotherapy, suggesting distinct mechanisms of toxicity and antitumor immunity.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires/effets indésirables , Immunothérapie/effets indésirables , Cellules de Küpffer/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Tumeurs/thérapie , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Animaux , Antigènes CD40/antagonistes et inhibiteurs , Antigènes CD40/immunologie , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Cytokines/immunologie , Humains , Cellules de Küpffer/immunologie , Foie/immunologie , Souris transgéniques , Tumeurs/immunologie , Granulocytes neutrophiles/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
11.
J Exp Med ; 218(1)2021 01 04.
Article de Anglais | MEDLINE | ID: mdl-33601412

RÉSUMÉ

Dendritic cells (DCs) contribute a small fraction of the tumor microenvironment but are emerging as an essential antitumor component based on their ability to foster T cell immunity and immunotherapy responses. Here, we discuss our expanding view of DC heterogeneity in human tumors, as revealed with meta-analysis of single-cell transcriptome profiling studies. We further examine tumor-infiltrating DC states that are conserved across patients, cancer types, and species and consider the fundamental and clinical relevance of these findings. Finally, we provide an outlook on research opportunities to further explore mechanisms governing tumor-infiltrating DC behavior and functions.


Sujet(s)
Cellules dendritiques/immunologie , Tumeurs/immunologie , Microenvironnement tumoral/immunologie , Cellules dendritiques/anatomopathologie , Analyse de profil d'expression de gènes , Humains , Tumeurs/classification , Tumeurs/anatomopathologie
12.
Proc Natl Acad Sci U S A ; 117(1): 541-551, 2020 01 07.
Article de Anglais | MEDLINE | ID: mdl-31889004

RÉSUMÉ

Cancer immunotherapies are increasingly combined with targeted therapies to improve therapeutic outcomes. We show that combination of agonistic anti-CD40 with antiangiogenic antibodies targeting 2 proangiogenic factors, vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (Ang2/ANGPT2), induces pleiotropic immune mechanisms that facilitate tumor rejection in several tumor models. On the one hand, VEGFA/Ang2 blockade induced regression of the tumor microvasculature while decreasing the proportion of nonperfused vessels and reducing leakiness of the remaining vessels. On the other hand, both anti-VEGFA/Ang2 and anti-CD40 independently promoted proinflammatory macrophage skewing and increased dendritic cell activation in the tumor microenvironment, which were further amplified upon combination of the 2 treatments. Finally, combined therapy provoked brisk infiltration and intratumoral redistribution of cytotoxic CD8+ T cells in the tumors, which was mainly driven by Ang2 blockade. Overall, these nonredundant synergistic mechanisms endowed T cells with improved effector functions that were conducive to more efficient tumor control, underscoring the therapeutic potential of antiangiogenic immunotherapy in cancer.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Antigènes CD40/agonistes , Tumeurs/traitement médicamenteux , Néovascularisation pathologique/traitement médicamenteux , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Angiopoïétine-2/antagonistes et inhibiteurs , Angiopoïétine-2/métabolisme , Animaux , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigènes CD40/immunologie , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Humains , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Souris , Tumeurs/vascularisation , Tumeurs/immunologie , Tumeurs/anatomopathologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/immunologie , Microenvironnement tumoral/immunologie , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/métabolisme
13.
Ther Umsch ; 76(4): 187-194, 2019 Sep.
Article de Allemand | MEDLINE | ID: mdl-31498037

RÉSUMÉ

Immunotherapies - Overview, mode of action and clinical implications Abstract. The introduction of immunotherapies has led to major advances in the treatment of cancer patients. The mainstays of immunotherapies in clinical routine are immune checkpoint inhibitors. Immune checkpoints like CTLA-4 or the PD-1 / PD-L1 axis are important contributors to the immune homeostasis by preventing overshooting immune responses against pathogens and thus preventing collateral damage to normal tissue, or by preventing autoimmunity. However, immune checkpoints can impede the development of an efficient anti-tumor immune response. Thus, therapeutic monoclonal antibodies against CTLA-4 and PD-1 or PD-L1 displayed remarkable clinical activity such as complete sustained clinical remission even in patients bearing multiple metastases. Malignant melanoma, non-small cell lung cancer or Hodgkin's lymphoma are examples of cancer entities with especially well clinical responses to immune checkpoint inhibitors. This fast-developing field is rapidly expanding the indications for immune checkpoint inhibitors and combinations with other therapeutic strategies like vessel-modulating agents or classical chemotherapy are in preclinical and clinical testing. In this article, the mechanistic principles of immune checkpoint inhibition and their clinical applications are illustrated.


Sujet(s)
Immunothérapie , Tumeurs/thérapie , Anticorps monoclonaux/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Antigène CTLA-4/antagonistes et inhibiteurs , Humains , Immunothérapie/méthodes
14.
J Mammary Gland Biol Neoplasia ; 24(1): 39-45, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30209717

RÉSUMÉ

Genetically engineered mouse models have become an indispensable tool for breast cancer research. Combination of multiple site-specific recombination systems such as Cre/loxP and Flippase (Flp)/Frt allows for engineering of sophisticated, multi-layered conditional mouse models. Here, we report the generation and characterization of a novel transgenic mouse line expressing a mouse codon-optimized Flp under the control of the mouse mammary tumor virus (MMTV) promoter. These mice show robust Flp-mediated recombination in luminal mammary gland and breast cancer cells but no Flp activity in non-mammary tissues, with the exception of limited activity in salivary glands. These mice provide a unique tool for studying mammary gland biology and carcinogenesis in mice.


Sujet(s)
Carcinogenèse/génétique , DNA nucleotidyltransferases/génétique , Glandes mammaires animales/anatomopathologie , Tumeurs expérimentales de la mamelle/génétique , Virus de la tumeur mammaire de la souris/génétique , Animaux , Carcinogenèse/anatomopathologie , Évolution de la maladie , Cellules épithéliales/anatomopathologie , Femelle , Gènes rapporteurs/génétique , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique , Protéines luminescentes/génétique , Glandes mammaires animales/cytologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris transgéniques , Microinjections , Régions promotrices (génétique)/génétique , Recombinaison génétique , Glandes salivaires/anatomopathologie , Microenvironnement tumoral/génétique ,
15.
Oncogenesis ; 7(9): 73, 2018 09 20.
Article de Anglais | MEDLINE | ID: mdl-30237500

RÉSUMÉ

Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that regulates a plethora of downstream signaling pathways essential for cell migration, proliferation and death, processes that are exploited by cancer cells during malignant progression. These well-established tumorigenic activities, together with its high expression and activity in different cancer types, highlight FAK as an attractive target for cancer therapy. We have assessed and characterized the therapeutic potential and the biological effects of BI 853520, a novel small chemical inhibitor of FAK, in several preclinical mouse models of breast cancer. Treatment with BI 853520 elicits a significant reduction in primary tumor growth caused by an anti-proliferative activity by BI 853520. In contrast, BI 853520 exerts effects with varying degrees of robustness on the different stages of the metastatic cascade. Together, the data demonstrate that the repression of FAK activity by the specific FAK inhibitor BI 853520 offers a promising anti-proliferative approach for cancer therapy.

17.
Mol Cancer Ther ; 16(11): 2502-2515, 2017 11.
Article de Anglais | MEDLINE | ID: mdl-28729403

RÉSUMÉ

Increasing the efficacy of approved systemic treatments in metastasized pancreatic neuroendocrine tumors (PanNET) is an unmet medical need. The antiangiogenic tyrosine kinase inhibitor sunitinib is approved for PanNET treatment. In addition, sunitinib is a lysosomotropic drug and such drugs can induce lysosomal membrane permeabilization as well as autophagy. We investigated sunitinib-induced autophagy as a possible mechanism of PanNET therapy resistance. Sunitinib accumulated in lysosomes and induced autophagy in PanNET cell lines. Adding the autophagy inhibitor chloroquine reduced cell viability in cell lines and in primary cells isolated from PanNET patients. The same treatment combination reduced tumor burden in the Rip1Tag2 transgenic PanNET mouse model. The combination of sunitinib and chloroquine reduced recovery and induced apoptosis in vitro, whereas single treatments did not. Knockdown of key autophagy proteins in combination with sunitinib showed similar effect as chloroquine. Sunitinib also induced lysosomal membrane permeabilization, which further increased in the presence of chloroquine or knockdown of lysosome-associated membrane protein (LAMP2). Both combinations led to cell death. Our data indicate that chloroquine increases sunitinib efficacy in PanNET treatment via autophagy inhibition and lysosomal membrane permeabilization. We suggest that adding chloroquine to sunitinib treatment will increase efficacy of PanNET treatment and that such patients should be included in respective ongoing clinical trials. Mol Cancer Ther; 16(11); 2502-15. ©2017 AACR.


Sujet(s)
Indoles/administration et posologie , Protéine de membrane-2 associée au lysosome/génétique , Néovascularisation pathologique/traitement médicamenteux , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs du pancréas/traitement médicamenteux , Pyrroles/administration et posologie , Inhibiteurs de l'angiogenèse/administration et posologie , Inhibiteurs de l'angiogenèse/composition chimique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Perméabilité des membranes cellulaires/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Synergie des médicaments , Techniques de knock-down de gènes , Humains , Indoles/composition chimique , Lysosomes/composition chimique , Souris , Néovascularisation pathologique/génétique , Néovascularisation pathologique/anatomopathologie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/composition chimique , Pyrroles/composition chimique , Sunitinib
18.
Methods Mol Biol ; 1464: 151-161, 2016.
Article de Anglais | MEDLINE | ID: mdl-27858364

RÉSUMÉ

The Rip1Tag2 transgenic mouse model of ß-cell carcinogenesis has been instrumental in studying various aspects of tumor angiogenesis and in investigating the response to anti-angiogenic therapeutics. Thereby, the in-depth assessment of blood and lymphatic vessel phenotypes and functionality represents key experimental analyses. In this chapter, we describe basic protocols to assess tumor blood vessel morphology (pericyte coverage), functionality (perfusion, leakiness, and hypoxia), lymphatic tumor coverage, and tumor cell proliferation and apoptosis based on immunofluorescence microscopy analysis.


Sujet(s)
Carcinome neuroendocrine/génétique , Insulinome/génétique , Néovascularisation pathologique/anatomopathologie , Tumeurs du pancréas/génétique , Animaux , Antigènes des virus oncogènes/génétique , Apoptose , Carcinome neuroendocrine/vascularisation , Carcinome neuroendocrine/anatomopathologie , Prolifération cellulaire , Insuline/génétique , Insulinome/vascularisation , Insulinome/anatomopathologie , Souris , Souris transgéniques , Tumeurs expérimentales/vascularisation , Tumeurs expérimentales/génétique , Néovascularisation pathologique/génétique , Tumeurs du pancréas/vascularisation , Tumeurs du pancréas/anatomopathologie , Régions promotrices (génétique)
19.
Cell Rep ; 15(6): 1161-74, 2016 05 10.
Article de Anglais | MEDLINE | ID: mdl-27134168

RÉSUMÉ

Despite the approval of several anti-angiogenic therapies, clinical results remain unsatisfactory, and transient benefits are followed by rapid tumor recurrence. Here, we demonstrate potent anti-angiogenic efficacy of the multi-kinase inhibitors nintedanib and sunitinib in a mouse model of breast cancer. However, after an initial regression, tumors resume growth in the absence of active tumor angiogenesis. Gene expression profiling of tumor cells reveals metabolic reprogramming toward anaerobic glycolysis. Indeed, combinatorial treatment with a glycolysis inhibitor (3PO) efficiently inhibits tumor growth. Moreover, tumors establish metabolic symbiosis, illustrated by the differential expression of MCT1 and MCT4, monocarboxylate transporters active in lactate exchange in glycolytic tumors. Accordingly, genetic ablation of MCT4 expression overcomes adaptive resistance against anti-angiogenic therapy. Hence, targeting metabolic symbiosis may be an attractive avenue to avoid resistance development to anti-angiogenic therapy in patients.


Sujet(s)
Inhibiteurs de l'angiogenèse/pharmacologie , Résistance aux médicaments antinéoplasiques , Tumeurs mammaires de l'animal/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Inhibiteurs de l'angiogenèse/usage thérapeutique , Animaux , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Glycolyse/effets des médicaments et des substances chimiques , Humains , Indoles/pharmacologie , Indoles/usage thérapeutique , Tumeurs mammaires de l'animal/vascularisation , Tumeurs mammaires de l'animal/traitement médicamenteux , Tumeurs mammaires de l'animal/anatomopathologie , Souris , Modèles biologiques , Transporteurs d'acides monocarboxyliques/métabolisme , Protéines du muscle/métabolisme , Néovascularisation pathologique/anatomopathologie
20.
Oncotarget ; 7(18): 25983-6002, 2016 May 03.
Article de Anglais | MEDLINE | ID: mdl-27036020

RÉSUMÉ

An epithelial to mesenchymal transition (EMT) enables epithelial tumor cells to break out of the primary tumor mass and to metastasize. Understanding the molecular mechanisms driving EMT in more detail will provide important tools to interfere with the metastatic process. To identify pharmacological modulators and druggable targets of EMT, we have established a novel multi-parameter, high-content, microscopy-based assay and screened chemical compounds with activities against known targets. Out of 3423 compounds, we have identified 19 drugs that block transforming growth factor beta (TGFß)-induced EMT in normal murine mammary gland epithelial cells (NMuMG). The active compounds include inhibitors against TGFß receptors (TGFBR), Rho-associated protein kinases (ROCK), myosin II, SRC kinase and uridine analogues. Among the EMT-repressing compounds, we identified a group of inhibitors targeting multiple receptor tyrosine kinases, and biochemical profiling of these multi-kinase inhibitors reveals TGFBR as a thus far unknown target of their inhibitory spectrum. These findings demonstrate the feasibility of a multi-parameter, high-content microscopy screen to identify modulators and druggable targets of EMT. Moreover, the newly discovered "off-target" effects of several receptor tyrosine kinase inhibitors have important consequences for in vitro and in vivo studies and might beneficially contribute to the therapeutic effects observed in vivo.


Sujet(s)
Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Tests de criblage à haut débit/méthodes , Tumeurs mammaires de l'animal/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Récepteurs TGF-bêta/antagonistes et inhibiteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Tumeurs mammaires de l'animal/métabolisme , Tumeurs mammaires de l'animal/anatomopathologie , Souris , Récepteurs à activité tyrosine kinase , Récepteur de type II du facteur de croissance transformant bêta , Facteur de croissance transformant bêta/métabolisme , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE