Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 35
Filtrer
1.
Med ; 5(7): 759-779.e7, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38593812

RÉSUMÉ

BACKGROUND: The treatment of melanoma, the deadliest form of skin cancer, has greatly benefited from immunotherapy. However, many patients do not show a durable response, which is only partially explained by known resistance mechanisms. METHODS: We performed single-cell RNA sequencing of tumor immune infiltrates and matched peripheral blood mononuclear cells of 22 checkpoint inhibitor (CPI)-naive stage III-IV metastatic melanoma patients. After sample collection, the same patients received CPI treatment, and their response was assessed. FINDINGS: CPI responders showed high levels of classical monocytes in peripheral blood, which preferentially transitioned toward CXCL9-expressing macrophages in tumors. Trajectories of tumor-infiltrating CD8+ T cells diverged at the level of effector memory/stem-like T cells, with non-responder cells progressing into a state characterized by cellular stress and apoptosis-related gene expression. Consistently, predicted non-responder-enriched myeloid-T/natural killer cell interactions were primarily immunosuppressive, while responder-enriched interactions were supportive of T cell priming and effector function. CONCLUSIONS: Our study illustrates that the tumor immune microenvironment prior to CPI treatment can be indicative of response. In perspective, modulating the myeloid and/or effector cell compartment by altering the described cell interactions and transitions could improve immunotherapy response. FUNDING: This research was funded by Roche Pharma Research and Early Development.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Mélanome , Tumeurs cutanées , Microenvironnement tumoral , Humains , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Tumeurs cutanées/immunologie , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Mâle , Femelle , Cellules myéloïdes/immunologie , Cellules myéloïdes/effets des médicaments et des substances chimiques , Cellules myéloïdes/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/métabolisme , Adulte d'âge moyen , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques
2.
Cancer Res ; 82(20): 3785-3801, 2022 10 17.
Article de Anglais | MEDLINE | ID: mdl-35979635

RÉSUMÉ

Agonistic αCD40 therapy has been shown to inhibit cancer progression in only a fraction of patients. Understanding the cancer cell-intrinsic and microenvironmental determinants of αCD40 therapy response is therefore crucial to identify responsive patient populations and to design efficient combinatorial treatments. Here, we show that the therapeutic efficacy of αCD40 in subcutaneous melanoma relies on preexisting, type 1 classical dendritic cell (cDC1)-primed CD8+ T cells. However, after administration of αCD40, cDC1s were dispensable for antitumor efficacy. Instead, the abundance of activated cDCs, potentially derived from cDC2 cells, increased and further activated antitumor CD8+ T cells. Hence, distinct cDC subsets contributed to the induction of αCD40 responses. In contrast, lung carcinomas, characterized by a high abundance of macrophages, were resistant to αCD40 therapy. Combining αCD40 therapy with macrophage depletion led to tumor growth inhibition only in the presence of strong neoantigens. Accordingly, treatment with immunogenic cell death-inducing chemotherapy sensitized lung tumors to αCD40 therapy in subcutaneous and orthotopic settings. These insights into the microenvironmental regulators of response to αCD40 suggest that different tumor types would benefit from different combinations of therapies to optimize the clinical application of CD40 agonists. SIGNIFICANCE: This work highlights the temporal roles of different dendritic cell subsets in promoting CD8+ T-cell-driven responses to CD40 agonist therapy in cancer.


Sujet(s)
Antigènes CD40 , Cellules dendritiques , Macrophages , Tumeurs , Animaux , Antigènes CD40/agonistes , Lymphocytes T CD8+ , Cellules dendritiques/métabolisme , Humains , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Tumeurs/métabolisme
3.
Sci Transl Med ; 13(598)2021 06 16.
Article de Anglais | MEDLINE | ID: mdl-34135110

RÉSUMÉ

Colony-stimulating factor 1 receptor (CSF1R) blockade abates tumor-associated macrophage (TAM) infiltrates and provides marked clinical benefits in diffuse-type tenosynovial giant cell tumors. However, facial edema is a common adverse event associated with TAM elimination in patients. In this study, we examined molecular and cellular events associated with edema formation in mice and human patients with cancer treated with a CSF1R blocking antibody. Extended antibody treatment of mice caused marked body weight gain, an indicator of enhanced body fluid retention. This was associated with an increase of extracellular matrix-remodeling metalloproteinases (MMPs), namely MMP2 and MMP3, and enhanced deposition of hyaluronan (HA) and proteoglycans, leading to skin thickening. Discontinuation of anti-CSF1R treatment or blockade of MMP activity restored unaltered body weight and normal skin morphology in the mice. In patients, edema developed at doses well below the established optimal biological dose for emactuzumab, a CSF1R dimerization inhibitor. Patients who developed edema in response to emactuzumab had elevated HA in peripheral blood. Our findings indicate that an early increase of peripheral HA can serve as a pharmacodynamic marker for edema development and suggest potential interventions based on MMP inhibition for relieving periorbital edema in patients treated with CSF1R inhibitors.


Sujet(s)
Oedème , Macrophages , Tumeurs , Peptide hydrolases , Protéoglycanes , Animaux , Anticorps monoclonaux humanisés/usage thérapeutique , Humains , Souris , Récepteur de facteur de croissance granulocyte-macrophage/antagonistes et inhibiteurs
4.
Front Immunol ; 11: 2082, 2020.
Article de Anglais | MEDLINE | ID: mdl-33013879

RÉSUMÉ

Particular interest to harness the innate immune system for cancer immunotherapy is fueled by limitations of immune checkpoint blockade. Plasmacytoid dendritic cells (pDC) are detected in a variety of solid tumors and correlate with poor clinical outcome. Release of type I interferons in response to toll-like-receptor (TLR)7 and TLR9 activation is the pDC hallmark. Mouse and human pDC differ substantially in their biology concerning surface marker expression and cytokine production. Here, we employed humanized mouse models (HIS) to study pDC function. We performed a comprehensive characterization of transgenic, myeloid-enhanced mouse strains (NOG-EXL and NSG-SGM3) expressing human interleukin-3 (hIL-3) and granulocyte-macrophage colony stimulating factor (GM-CSF) using identical humanization protocols. Only in HIS-NOG-EXL mice sufficient pDC infiltration was detectable. Therefore, we selected this strain for subsequent tumor studies. We analyzed pDC frequency in peripheral blood and tumors by comparing HIS-NOG-EXL with HIS-NOG mice bearing three different ovarian and breast tumors. Despite the substantially increased pDC numbers in peripheral blood of HIS-NOG-EXL mice, we detected TLR7/8 agonist responsive and thus functional pDCs only in certain tumor models independent of the mouse strain employed. However, HIS-NOG-EXL mice showed in general a superior humanization phenotype characterized by reconstitution of different myeloid subsets, NK cells and B cells producing physiologic IgG levels. Hence, we provide first evidence that the tumor milieu but not genetically introduced cytokines defines intratumoral (i.t.) frequencies of the rare pDC subset. This study provides model systems to investigate in vivo pro- and anti-tumoral human pDC functions.


Sujet(s)
Lymphocytes B/immunologie , Carcinome épithélial de l'ovaire/immunologie , Cellules dendritiques/immunologie , Cellules tueuses naturelles/immunologie , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Humains , Interleukine-3/génétique , Souris , Souris SCID , Souris transgéniques , Microenvironnement tumoral
5.
J Immunother Cancer ; 8(2)2020 10.
Article de Anglais | MEDLINE | ID: mdl-33097612

RÉSUMÉ

BACKGROUND: This phase Ib study evaluated the safety, clinical activity, pharmacokinetics, and pharmacodynamics (PD) of emactuzumab (anti-colony stimulating factor 1 receptor monoclonal antibody (mAb)) in combination with selicrelumab (agonistic cluster of differentiation 40 mAb) in patients with advanced solid tumors. METHODS: Both emactuzumab and selicrelumab were administered intravenously every 3 weeks and doses were concomitantly escalated (emactuzumab: 500 to 1000 mg flat; selicrelumab: 2 to 16 mg flat). Dose escalation was conducted using the product of independent beta probabilities dose-escalation design. PD analyzes were performed on peripheral blood samples and tumor/skin biopsies at baseline and on treatment. Clinical activity was evaluated using investigator-based and Response Evaluation Criteria In Solid Tumors V.1.1-based tumor assessments. RESULTS: Three dose-limiting toxicities (all infusion-related reactions (IRRs)) were observed at 8, 12 and 16 mg of selicrelumab together with 1000 mg of emactuzumab. The maximum tolerated dose was not reached at the predefined top doses of emactuzumab (1000 mg) and selicrelumab (16 mg). The most common adverse events were IRRs (75.7%), fatigue (54.1%), facial edema (37.8%), and increase in aspartate aminotransferase and creatinine phosphokinase (35.1% both). PD analyzes demonstrated an increase of Ki67+-activated CD8+ T cells accompanied by a decrease of B cells and the reduction of CD14Dim CD16bright monocytes in peripheral blood. The best objective clinical response was stable disease in 40.5% of patients. CONCLUSION: Emactuzumab in combination with selicrelumab demonstrated a manageable safety profile and evidence of PD activity but did not translate into objective clinical responses. TRIALREGISTRATION NUMBER: NCT02760797.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigènes CD40/métabolisme , Tumeurs/traitement médicamenteux , Récepteur du facteur de stimulation des colonies de macrophages/antagonistes et inhibiteurs , Anticorps monoclonaux humanisés/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Femelle , Humains , Mâle , Tumeurs/immunologie , Récepteur du facteur de stimulation des colonies de macrophages/métabolisme
6.
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
7.
PLoS One ; 14(7): e0219517, 2019.
Article de Anglais | MEDLINE | ID: mdl-31291357

RÉSUMÉ

Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and has a high mortality rate due to limited treatment options. Hence, the response of HCC to different cancer immunotherapies is being intensively investigated in clinical trials. Immune checkpoint blockers (ICB) show promising results, albeit for a minority of HCC patients. Mouse models are commonly used to evaluate new therapeutic agents or regimens. However, to make clinical translation more successful, better characterized preclinical models are required. We therefore extensively investigated two immune-competent orthotopic HCC mouse models, namely transplanted Hep-55.1c and transgenic iAST, with respect to morphological, immunological and genetic traits and evaluated both models' responsiveness to immunotherapies. Hep-55.1c tumors were characterized by rich fibrous stroma, high mutational load and pronounced immune cell infiltrates, all of which are features of immune-responsive tumors. These characteristics were less distinct in iAST tumors, though these were highly vascularized. Cell depletion revealed that CD8+ T cells from iAST mice do not affect tumor growth and are tumor tolerant. This corresponds to the failure of single and combined ICB targeting PD-1 and CTLA-4. In contrast, combining anti-PD-1 and anti-CTLA-4 showed significant antitumor efficacy in the Hep-55.1c mouse model. Collectively, our data comprehensively characterize two immune-competent HCC mouse models representing ICB responsive and refractory characteristics. Our characterization confirms these models to be suitable for preclinical investigation of novel cancer immunotherapy approaches that aim to either deepen preexisting immune responses or generate de novo immunity against the tumor.


Sujet(s)
Antinéoplasiques immunologiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Modèles animaux de maladie humaine , Tumeurs du foie/traitement médicamenteux , Animaux , Antigènes transformants de polyomavirus/génétique , Antinéoplasiques immunologiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphocytes T CD8+/immunologie , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/immunologie , Lignée cellulaire tumorale/transplantation , Résistance aux médicaments antinéoplasiques , Tests de criblage d'agents antitumoraux/méthodes , Femelle , Humains , Tumeurs du foie/génétique , Tumeurs du foie/immunologie , Souris , Souris de lignée C57BL , Souris transgéniques , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Résultat thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
8.
Hepatology ; 70(4): 1280-1297, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31002440

RÉSUMÉ

Antiangiogenic and cytotoxic effects are considered the principal mechanisms of action of sorafenib, a multitarget kinase inhibitor approved for the treatment of hepatocellular carcinoma (HCC). We report that sorafenib also acts through direct immune modulation, indispensable for its antitumor activity. In vivo cell depletion experiments in two orthotopic HCC mouse models as well as in vitro analysis identified macrophages (MΦ) as the key mediators of the antitumoral effect and demonstrate a strong interdependency of MΦ and natural killer (NK) cells for efficient tumor cell killing. Caspase 1 analysis in sorafenib-treated MΦ revealed an induction of pyroptosis. As a result, cytotoxic NK cells become activated when cocultured with sorafenib-treated MΦ, leading to tumor cell death. In addition, sorafenib was found to down-regulate major histocompatibility complex class I expression of tumor cells, which may reduce the tumor responsiveness to immune checkpoint therapies and favor NK-cell response. In vivo cytokine blocking revealed that sorafenib efficacy is abrogated after inhibition of interleukins 1B and 18. Conclusion: We report an immunomodulatory mechanism of sorafenib involving MΦ pyroptosis and unleashing of an NK-cell response that sets it apart from other spectrum kinase inhibitors as a promising immunotherapy combination partner for the treatment of HCC.


Sujet(s)
Carcinome hépatocellulaire/traitement médicamenteux , Cellules tueuses naturelles/métabolisme , Tumeurs du foie/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Pyroptose/effets des médicaments et des substances chimiques , Sorafénib/pharmacologie , Analyse de variance , Animaux , Carcinome hépatocellulaire/anatomopathologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Femelle , Cytométrie en flux , Humains , Injections veineuses , Tumeurs du foie/anatomopathologie , Macrophages , Souris , Souris transgéniques , Répartition aléatoire , Charge tumorale/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Microtomographie aux rayons X/méthodes , Tests d'activité antitumorale sur modèle de xénogreffe
9.
J Immunol ; 201(8): 2273-2286, 2018 10 15.
Article de Anglais | MEDLINE | ID: mdl-30209192

RÉSUMÉ

Immunotherapy with checkpoint inhibitors has proved to be highly effective, with durable responses in a subset of patients. Given their encouraging clinical activity, checkpoint inhibitors are increasingly being tested in clinical trials in combination with chemotherapy. In many instances, there is little understanding of how chemotherapy might influence the quality of the immune response generated by checkpoint inhibitors. In this study, we evaluated the impact of chemotherapy alone or in combination with anti-PD-L1 in a responsive syngeneic tumor model. Although multiple classes of chemotherapy treatment reduced immune cell numbers and activity in peripheral tissues, chemotherapy did not antagonize but in many cases augmented the antitumor activity mediated by anti-PD-L1. This dichotomy between the detrimental effects in peripheral tissues and enhanced antitumor activity was largely explained by the reduced dependence on incoming cells for antitumor efficacy in already established tumors. The effects of the various chemotherapies were also agent specific, and synergy with anti-PD-L1 was achieved by different mechanisms that ultimately helped establish a new threshold for response. These results rationalize the combination of chemotherapy with immunotherapy and suggest that, despite the negative systemic effects of chemotherapy, effective combinations can be obtained through distinct mechanisms acting within the tumor.


Sujet(s)
Adénocarcinome/thérapie , Anticorps monoclonaux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Tumeurs du côlon/thérapie , Immunothérapie/méthodes , Adénocarcinome/immunologie , Animaux , Antigènes néoplasiques/immunologie , Antigène CD274/immunologie , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Tumeurs du côlon/immunologie , Femelle , Cytométrie en flux , Humains , Immunité cellulaire , Souris , Souris de lignée C57BL , Tumeurs expérimentales , Charge tumorale/effets des médicaments et des substances chimiques
10.
Sci Transl Med ; 10(436)2018 04 11.
Article de Anglais | MEDLINE | ID: mdl-29643229

RÉSUMÉ

Colony-stimulating factor 1 (CSF1) is a key regulator of monocyte/macrophage differentiation that sustains the protumorigenic functions of tumor-associated macrophages (TAMs). We show that CSF1 is expressed in human melanoma, and patients with metastatic melanoma have increased CSF1 in blood compared to healthy subjects. In tumors, CSF1 expression correlated with the abundance of CD8+ T cells and CD163+ TAMs. Human melanoma cell lines consistently produced CSF1 after exposure to melanoma-specific CD8+ T cells or T cell-derived cytokines in vitro, reflecting a broadly conserved mechanism of CSF1 induction by activated CD8+ T cells. Mining of publicly available transcriptomic data sets suggested co-enrichment of CD8+ T cells with CSF1 or various TAM-specific markers in human melanoma, which was associated with nonresponsiveness to programmed cell death protein 1 (PD1) checkpoint blockade in a smaller patient cohort. Combination of anti-PD1 and anti-CSF1 receptor (CSF1R) antibodies induced the regression of BRAFV600E -driven, transplant mouse melanomas, a result that was dependent on the effective elimination of TAMs. Collectively, these data implicate CSF1 induction as a CD8+ T cell-dependent adaptive resistance mechanism and show that simultaneous CSF1R targeting may be beneficial in melanomas refractory to immune checkpoint blockade and, possibly, other T cell-based therapies.


Sujet(s)
Facteur de stimulation des colonies de macrophages/sang , Mélanome/sang , Mélanome/anatomopathologie , Animaux , Antigènes CD/métabolisme , Antigènes de différenciation des myélomonocytes/métabolisme , Lymphocytes T CD8+/métabolisme , Différenciation cellulaire/physiologie , Lignée cellulaire tumorale , Humains , Macrophages/métabolisme , Souris , Protéines proto-oncogènes B-raf/métabolisme , Récepteurs de surface cellulaire/métabolisme , Transduction du signal
11.
J Exp Med ; 215(3): 859-876, 2018 03 05.
Article de Anglais | MEDLINE | ID: mdl-29436396

RÉSUMÉ

Depletion of immunosuppressive tumor-associated macrophages (TAMs) or reprogramming toward a proinflammatory activation state represent different strategies to therapeutically target this abundant myeloid population. In this study, we report that inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling sensitizes TAMs to profound and rapid reprogramming in the presence of a CD40 agonist before their depletion. Despite the short-lived nature of macrophage hyperactivation, combined CSF-1R+CD40 stimulation of macrophages is sufficient to create a proinflammatory tumor milieu that reinvigorates an effective T cell response in transplanted tumors that are either responsive or insensitive to immune checkpoint blockade. The central role of macrophages in regulating preexisting immunity is substantiated by depletion experiments, transcriptome analysis of ex vivo sorted TAMs, and gene expression profiling of whole tumor lysates at an early treatment time point. This approach enabled the identification of specific combination-induced changes among the pleiotropic activation spectrum of the CD40 agonist. In patients, CD40 expression on human TAMs was detected in mesothelioma and colorectal adenocarcinoma.


Sujet(s)
Immunité , Macrophages/immunologie , Tumeurs/immunologie , Tumeurs/anatomopathologie , Animaux , Antigènes CD40/agonistes , Antigènes CD40/métabolisme , Lymphocytes T CD8+/immunologie , Femelle , Humains , Inflammation/anatomopathologie , Souris de lignée BALB C , Souris de lignée C57BL , Modèles biologiques , Phénotype , Récepteur du facteur de stimulation des colonies de macrophages/antagonistes et inhibiteurs , Récepteur du facteur de stimulation des colonies de macrophages/métabolisme
12.
FEBS J ; 285(4): 763-776, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-28941174

RÉSUMÉ

Despite decades of research, cancer remains a devastating disease and new treatment options are needed. Today cancer is acknowledged as a multifactorial disease not only comprising of aberrant tumor cells but also the associated stroma including tumor vasculature, fibrotic plaques, and immune cells that interact in a complex heterotypic interplay. Myeloid cells represent one of the most abundant immune cell population within the tumor stroma and are equipped with a broad functional repertoire that promotes tumor growth by suppressing cytotoxic T cell activity, stimulating neoangiogenesis and tissue remodeling. Therefore, myeloid cells have become an attractive target for pharmacological intervention. In this review, we summarize the pharmacological approaches to therapeutically target tumor-associated myeloid cells with a focus on advanced programs that are clinically evaluated. In addition, for each therapeutic strategy, the preclinical rationale as well as advantages and challenges from a drug development perspective are discussed.


Sujet(s)
Antinéoplasiques/pharmacologie , Développement de médicament , Cellules myéloïdes/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Humains , Tumeurs/métabolisme , Tumeurs/anatomopathologie
14.
Cancer Cell ; 30(3): 377-390, 2016 09 12.
Article de Anglais | MEDLINE | ID: mdl-27622331

RÉSUMÉ

Effective cancer immunotherapy requires overcoming immunosuppressive tumor microenvironments. We found that local nitric oxide (NO) production by tumor-infiltrating myeloid cells is important for adoptively transferred CD8(+) cytotoxic T cells to destroy tumors. These myeloid cells are phenotypically similar to inducible nitric oxide synthase (NOS2)- and tumor necrosis factor (TNF)-producing dendritic cells (DC), or Tip-DCs. Depletion of immunosuppressive, colony stimulating factor 1 receptor (CSF-1R)-dependent arginase 1(+) myeloid cells enhanced NO-dependent tumor killing. Tumor elimination via NOS2 required the CD40-CD40L pathway. We also uncovered a strong correlation between survival of colorectal cancer patients and NOS2, CD40, and TNF expression in their tumors. Our results identify a network of pro-tumor factors that can be targeted to boost cancer immunotherapies.


Sujet(s)
Cellules dendritiques/immunologie , Immunothérapie adoptive/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Nitric oxide synthase type II/immunologie , Lymphocytes T cytotoxiques/immunologie , Facteur de nécrose tumorale alpha/immunologie , Animaux , Arginase/biosynthèse , Arginase/immunologie , Antigènes CD40/immunologie , Ligand de CD40/immunologie , Lignée cellulaire tumorale , Humains , Souris , Souris de lignée C57BL , Souris knockout , Nitric oxide synthase type II/biosynthèse , Microenvironnement tumoral , Facteur de nécrose tumorale alpha/biosynthèse
15.
Nat Cell Biol ; 18(7): 790-802, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27295554

RÉSUMÉ

Tumour-associated macrophages (TAMs) largely express an alternatively activated (or M2) phenotype, which entails immunosuppressive and tumour-promoting capabilities. Reprogramming TAMs towards a classically activated (M1) phenotype may thwart tumour-associated immunosuppression and unleash anti-tumour immunity. Here we show that conditional deletion of the microRNA (miRNA)-processing enzyme DICER in macrophages prompts M1-like TAM programming, characterized by hyperactive IFN-γ/STAT1 signalling. This rewiring abated the immunosuppressive capacity of TAMs and fostered the recruitment of activated cytotoxic T lymphocytes (CTLs) to the tumours. CTL-derived IFN-γ exacerbated M1 polarization of Dicer1-deficient TAMs and inhibited tumour growth. Remarkably, DICER deficiency in TAMs negated the anti-tumoral effects of macrophage depletion by anti-CSF1R antibodies, and enabled complete tumour eradication by PD1 checkpoint blockade or CD40 agonistic antibodies. Finally, genetic rescue of Let-7 miRNA activity in Dicer1-deficient TAMs partly restored their M2-like phenotype and decreased tumour-infiltrating CTLs. These findings suggest that DICER/Let-7 activity opposes IFN-γ-induced, immunostimulatory M1-like TAM activation, with potential therapeutic implications.


Sujet(s)
DEAD-box RNA helicases/métabolisme , Interféron gamma/métabolisme , Activation des macrophages/immunologie , Macrophages/immunologie , microARN/génétique , Ribonuclease III/métabolisme , Microenvironnement tumoral/génétique , Animaux , Cellules cultivées , DEAD-box RNA helicases/déficit , Humains , Souris , Tumeurs/génétique , Tumeurs/immunologie , Ribonuclease III/déficit
16.
Curr Opin Pharmacol ; 23: 45-51, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26051995

RÉSUMÉ

Macrophage infiltration has been identified as an independent poor prognostic factor for several cancer entities. In mouse tumor models macrophages orchestrate various tumor-promoting processes. This observation sparked an interest to therapeutically target these plastic innate immune cells. To date, blockade of colony stimulating factor-1 or its receptor represents the only truly selective approach to manipulate macrophages in cancer patients. Here, we discuss the currently available information on efficacy and safety of various CSF-1/CSF-1R inhibitors in cancer patients and highlight potential combination partners emerging from preclinical studies while considering the differences between mouse and human macrophage biology.


Sujet(s)
Antinéoplasiques/immunologie , Systèmes de délivrance de médicaments/tendances , Facteur de stimulation des colonies de macrophages/immunologie , Macrophages/immunologie , Tumeurs/immunologie , Récepteur du facteur de stimulation des colonies de macrophages/immunologie , Animaux , Antinéoplasiques/administration et posologie , Découverte de médicament/tendances , Humains , Macrophages/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie
17.
Cancer Cell ; 25(6): 846-59, 2014 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-24898549

RÉSUMÉ

Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80(+) tumor-associated macrophages accompanied by an increase of the CD8(+)/CD4(+) T cell ratio. Administration of RG7155 to patients led to striking reductions of CSF-1R(+)CD163(+) macrophages in tumor tissues, which translated into clinical objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.


Sujet(s)
Anticorps monoclonaux/immunologie , Anticorps monoclonaux/pharmacologie , Tumeurs du côlon/thérapie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Récepteur du facteur de stimulation des colonies de macrophages/antagonistes et inhibiteurs , Récepteur du facteur de stimulation des colonies de macrophages/immunologie , Animaux , Anticorps monoclonaux/pharmacocinétique , Anticorps monoclonaux humanisés , Différenciation cellulaire/physiologie , Lignée cellulaire tumorale , Essais cliniques de phase I comme sujet , Études de cohortes , Tumeurs du côlon/immunologie , Tumeurs du côlon/métabolisme , Femelle , Humains , Macaca fascicularis , Macrophages/cytologie , Macrophages/métabolisme , Mâle , Souris de lignée C57BL , Modèles moléculaires , Récepteur du facteur de stimulation des colonies de macrophages/métabolisme
18.
Oncoimmunology ; 3(9): e955356, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25941615

RÉSUMÉ

Macrophage infiltration has been identified as an independent, poor prognostic factor for patients afflicted with various cancer entities. However, the characterization of tumor-associated macrophages (TAMs) prior to and following cancer patient treatment has been limited. Our study analyzed tumor biopsies before and after anti-CSF-1R antibody treatment unraveling the nature of TAMs and providing novel insights into their phenotypic and functional characteristics in cancer.

19.
Cancer Res ; 73(6): 1709-20, 2013 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-23338611

RÉSUMÉ

Deregulated TGF-ß signaling in pancreatic cancer promotes tumor growth, invasion, metastasis, and a potent immunosuppressive network. A strategy for disrupting this tumor-promoting pathway is silencing TGF-ß by siRNA. By introducing a triphosphate group at the 5' end of siRNA (ppp-siRNA), gene silencing can be combined with immune activation via the cytosolic helicase retinoic acid-inducible gene I (RIG-I), a ubiquitously expressed receptor recognizing viral RNA. We validated RIG-I as a therapeutic target by showing that activation of RIG-I in pancreatic carcinoma cells induced IRF-3 phosphorylation, production of type I IFN, the chemokine CXCL10, as well as caspase-9-mediated tumor cell apoptosis. Next, we generated a bifunctional ppp-siRNA that combines RIG-I activation with gene silencing of TGF-ß1 (ppp-TGF-ß) and studied its therapeutic efficacy in the orthotopic Panc02 mouse model of pancreatic cancer. Intravenous injection of ppp-TGF-ß reduced systemic and tumor-associated TGF-ß levels. In addition, it induced high levels of type I IFN and CXCL10 in serum and tumor tissue, systemic immune cell activation, and profound tumor cell apoptosis in vivo. Treatment of mice with established tumors with ppp-TGF-ß significantly prolonged survival as compared with ppp-RNA or TGF-ß siRNA alone. Furthermore, we observed the recruitment of activated CD8(+) T cells to the tumor and a reduced frequency of CD11b(+) Gr-1(+) myeloid cells. Therapeutic efficacy was dependent on CD8(+) T cells, whereas natural killer cells were dispensable. In conclusion, combing TGF-ß gene silencing with RIG-I signaling confers potent antitumor efficacy against pancreatic cancer by breaking tumor-induced CD8(+) T cell suppression.


Sujet(s)
DEAD-box RNA helicases/métabolisme , Extinction de l'expression des gènes , Tumeurs du pancréas/génétique , Petit ARN interférent/génétique , Facteur de croissance transformant bêta-1/génétique , Animaux , Apoptose , Lymphocytes T CD8+/immunologie , Chimiokine CXCL10/sang , Protéine-58 à domaine DEAD , Test ELISA , Femelle , Cytométrie en flux , Interféron de type I/sang , Souris , Souris de lignée C57BL , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Petit ARN interférent/usage thérapeutique , Réaction de polymérisation en chaine en temps réel , Transduction du signal
20.
Oncoimmunology ; 1(2): 219-221, 2012 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-22720248

RÉSUMÉ

The cytotoxic properties of granzymes are well established, though recent publications suggest additional roles for granzymes in immunity. We demonstrated that granzymes can act as regulators of cross-presentation by dendritic cells by inducing critical "eat-me" signals on the dying tumor cell, resulting in efficient phagocytosis of cell-associated tumor antigen.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE