Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Lancet Oncol ; 20(8): 1160-1170, 2019 08.
Article in English | MEDLINE | ID: mdl-31253572

ABSTRACT

BACKGROUND: IPH4102 is a first-in-class monoclonal antibody targeting KIR3DL2, a cell surface protein that is expressed in cutaneous T-cell lymphoma, and predominantly in its leukaemic form, Sézary syndrome. We aimed to assess the safety and activity of IPH4102 in cutaneous T-cell lymphoma. METHODS: We did an international, first-in-human, open-label, phase 1 clinical trial with dose-escalation and cohort-expansion parts in five academic hospitals in the USA, France, the UK, and the Netherlands. Eligible patients had histologically confirmed relapsed or refractory primary cutaneous T-cell lymphoma, an Eastern Cooperative Oncology group performance score of 2 or less, were aged 18 years or older, and had received at least two previous systemic therapies. Ten dose levels of IPH4102, administered as an intravenous infusion, ranging from 0·0001 mg/kg to 10 mg/kg, were assessed using an accelerated 3 + 3 design. The primary endpoint was the occurrence of dose-limiting toxicities during the first 2 weeks of treatment, defined as toxicity grade 3 or worse lasting for 8 or more days, except for lymphopenia. Global overall response by cutaneous T-cell lymphoma subtype was a secondary endpoint. Safety and activity analyses were done in the per-protocol population. The study is ongoing and recruitment is complete. This trial is registered with ClinicalTrials.gov, number NCT02593045. FINDINGS: Between Nov 4, 2015, and Nov 20, 2017, 44 patients were enrolled. 35 (80%) patients had Sézary syndrome, eight (18%) had mycosis fungoides, and one (2%) had primary cutaneous T-cell lymphoma, not otherwise specified. In the dose-escalation part, no dose limiting toxicity was reported and the trial's safety committee recommended a flat dose of 750 mg for the cohort-expansion, corresponding to the maximum administered dose. The most common adverse events were peripheral oedema (12 [27%] of 44 patients) and fatigue (nine [20%]), all of which were grade 1-2. Lymphopenia was the most common grade 3 or worse adverse event (three [7%]). One patient developed possibly treatment-related fulminant hepatitis 6 weeks after IPH4102 discontinuation and subsequently died. However, the patient had evidence of human herpes virus-6B infection. Median follow-up was 14·1 months (IQR 11·3-20·5). A confirmed global overall response was achieved in 16 (36·4% [95% CI 23·8-51·1]) of 44 patients, and of those, 15 responses were observed in 35 patients with Sézary syndrome (43% [28·0-59·1]). INTERPRETATION: IPH4102 is safe and shows encouraging clinical activity in patients with relapsed or refractory cutaneous T-cell lymphoma, particularly those with Sézary syndrome. If confirmed in future trials, IPH4102 could become a novel treatment option for these patients. A multi-cohort, phase 2 trial (TELLOMAK) is underway to confirm the activity in patients with Sézary syndrome and explore the role of IPH4102 in other subtypes of T-cell lymphomas that express KIR3DL2. FUNDING: Innate Pharma.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Lymphoma, T-Cell, Cutaneous/drug therapy , Receptors, KIR3DL2/antagonists & inhibitors , Skin Neoplasms/drug therapy , Aged , Antineoplastic Agents, Immunological/adverse effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/drug therapy
2.
Immunity ; 29(3): 464-75, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18789731

ABSTRACT

Oral tolerance prevents oral sensitization to dietary antigens (Ags), including proteins and haptens, and development of delayed-type hypersensitivity (DTH) responses. We showed here that plasmacytoid dendritic cells (pDCs) prevented oral T cell priming and were responsible for systemic tolerance to CD4(+) and CD8(+) T cell-mediated DTH responses induced by Ag feeding. Systemic depletion of pDCs prevented induction of tolerance by antigen feeding. Transfer of oral Ag-loaded liver pDCs to naive recipient mice induced Ag-specific suppression of CD4(+) and CD8(+) T cell responses to protein and hapten, respectively. Liver is a site of oral Ag presentation, and pDCs appeared to induce anergy or deletion of Ag-specific T cells in the liver relatively rapidly via a CD4(+) T cell-independent mechanism. These data demonstrate that oral tolerance relies on Ag presentation by pDC to T cells and suggest that pDC could represent a key therapeutic target for intestinal and systemic inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Hypersensitivity, Delayed/immunology , Immune Tolerance , Mouth Mucosa/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Dinitrofluorobenzene/immunology , Female , Hypersensitivity, Delayed/metabolism , Liver/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Spleen/immunology
3.
J Immunol ; 193(4): 1622-35, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25009205

ABSTRACT

Human monocyte-derived dendritic cell (MoDC) have been used in the clinic with moderately encouraging results. Mouse XCR1(+) DC excel at cross-presentation, can be targeted in vivo to induce protective immunity, and share characteristics with XCR1(+) human DC. Assessment of the immunoactivation potential of XCR1(+) human DC is hindered by their paucity in vivo and by their lack of a well-defined in vitro counterpart. We report in this study a protocol generating both XCR1(+) and XCR1(-) human DC in CD34(+) progenitor cultures (CD34-DC). Gene expression profiling, phenotypic characterization, and functional studies demonstrated that XCR1(-) CD34-DC are similar to canonical MoDC, whereas XCR1(+) CD34-DC resemble XCR1(+) blood DC (bDC). XCR1(+) DC were strongly activated by polyinosinic-polycytidylic acid but not LPS, and conversely for MoDC. XCR1(+) DC and MoDC expressed strikingly different patterns of molecules involved in inflammation and in cross-talk with NK or T cells. XCR1(+) CD34-DC but not MoDC efficiently cross-presented a cell-associated Ag upon stimulation by polyinosinic-polycytidylic acid or R848, likewise to what was reported for XCR1(+) bDC. Hence, it is feasible to generate high numbers of bona fide XCR1(+) human DC in vitro as a model to decipher the functions of XCR1(+) bDC and as a potential source of XCR1(+) DC for clinical use.


Subject(s)
Antigens, CD34/immunology , Blood Cells/immunology , Dendritic Cells/immunology , Monocytes/immunology , Receptors, G-Protein-Coupled/immunology , Adjuvants, Immunologic/pharmacology , Antigen Presentation/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Cell Line , Cross-Priming/immunology , Gene Expression Profiling , Green Fluorescent Proteins , Humans , Imidazoles/immunology , Killer Cells, Natural/immunology , Lipopolysaccharides/immunology , Phenotype , Poly I-C/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 3 , Toll-Like Receptor 4
4.
J Clin Pharmacol ; 63(7): 817-829, 2023 07.
Article in English | MEDLINE | ID: mdl-36852723

ABSTRACT

Monalizumab is a novel, first-in-class humanized immunoglobulin G4 monoclonal antibody immune checkpoint inhibitor that targets the inhibitory CD94/NKG2A receptors. The objectives of this analysis were to develop a population pharmacokinetic (PK) model of monalizumab, evaluate the impact of clinically relevant covariates on monalizumab PK, and provide dose justification for clinical trials. We developed a monalizumab population PK model to characterize the PK properties of monalizumab in patients with advanced solid tumors or head and neck squamous cell carcinoma. Data from clinical studies D419NC00001 (NCT02671435) and IPH2201-203 (NCT02643550) were pooled for the analysis, resulting in a data set of 3066 PK samples derived from 507 subjects. The PK of monalizumab were reasonably described by a 2-compartment model with first-order elimination. Monalizumab generally exhibited linear PK over a dose range of 22.5-750 mg or 10 mg/kg every 2 weeks. The estimate of clearance was ≈0.255 L/day, and apparent volume of distribution was 6.36 L for a typical individual, consistent with previous findings for endogenous immunoglobulin Gs and other therapeutic monoclonal antibodies. Baseline albumin and body weight were identified as significant covariates of clearance; body weight, sex, and smoking status had a significant impact on volume of distribution; and none of these covariates had impact on peripheral volume of distribution. Although these covariates were identified as statistically significant, they are considered to be not clinically meaningful, as changes in monalizumab exposure were <30%. Therefore, no dose adjustments of monalizumab based on patient or disease characteristics are recommended.


Subject(s)
Antibodies, Monoclonal, Humanized , Neoplasms , Humans , Antibodies, Monoclonal, Humanized/pharmacokinetics , Neoplasms/drug therapy , Antibodies, Monoclonal/pharmacokinetics , Body Weight , Models, Biological
5.
Blood ; 115(10): 1949-57, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20065291

ABSTRACT

There is a high demand for the development of adjuvants that induce cytotoxic T lymphocytes, which are crucial for the elimination of intracellular pathogens and tumor cells. Toll-like receptor (TLR) agonists are prime candidates to fulfill this role because they induce innate immune activation and promote adaptive immune responses. The successful application of the TLR7 agonist R837 for treatment of basal cell carcinoma shows the potential for exploiting this pathway in tumor immunotherapy. Imidazoquinolines like R837 and stimulatory ssRNA oligonucleotides both trigger TLR7-mediated immune activation, but little is known about their comparative ability to promote immunity induction. We investigated differences in innate immune activation and adjuvant activity between the imidazoquinoline R848 and the ssRNA TLR7 agonist polyUs21. In contrast to R848, polyUs21 induced detectable levels of intracellular interferon-alpha (IFN-alpha) in plasmacytoid dendritic cells (PDCs). In immunization studies, only polyUs21 led to robust priming of type 1 T helper cells and cytotoxic T lymphocytes, and it was more efficient in inducing antitumor immunity than R848. Notably, exogenous IFN-alpha augmented the adjuvant activity of R848, whereas depletion of PDC abrogated the adjuvanticity of polyUs21. This study, therefore, identifies sufficient IFN-alpha production by PDC as an important determinant of vaccine efficacy.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/metabolism , Interferon Type I/physiology , Membrane Glycoproteins/agonists , Toll-Like Receptor 7/agonists , Aminoquinolines/pharmacology , Aminoquinolines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Imidazoles/therapeutic use , Imiquimod , Interferon Type I/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolines/pharmacology , RNA/pharmacology , Receptor, Interferon alpha-beta/genetics , Tumor Cells, Cultured
6.
J Immunol ; 185(4): 2080-8, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20639488

ABSTRACT

Cross-talk between NK cells and dendritic cells (DCs) is critical for the potent therapeutic response to dsRNA, but the receptors involved remained controversial. We show in this paper that two dsRNAs, polyadenylic-polyuridylic acid and polyinosinic-polycytidylic acid [poly(I:C)], similarly engaged human TLR3, whereas only poly(I:C) triggered human RIG-I and MDA5. Both dsRNA enhanced NK cell activation within PBMCs but only poly(I:C) induced IFN-gamma. Although myeloid DCs (mDCs) were required for NK cell activation, induction of cytolytic potential and IFN-gamma production did not require contact with mDCs but was dependent on type I IFN and IL-12, respectively. Poly(I:C) but not polyadenylic-polyuridylic acid synergized with mDC-derived IL-12 for IFN-gamma production by acting directly on NK cells. Finally, the requirement of both TLR3 and Rig-like receptor (RLR) on mDCs and RLRs but not TLR3 on NK cells for IFN-gamma production was demonstrated using TLR3- and Cardif-deficient mice and human RIG-I-specific activator. Thus, we report the requirement of cotriggering TLR3 and RLR on mDCs and RLRs on NK cells for a pathogen product to induce potent innate cell activation.


Subject(s)
DEAD-box RNA Helicases/metabolism , Dendritic Cells/drug effects , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Humans , Interferon-Induced Helicase, IFIH1 , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Poly A-U/pharmacology , Poly I-C/pharmacology , RNA, Double-Stranded/pharmacology , Receptors, Immunologic , Toll-Like Receptor 3/genetics , Transfection
7.
J Exp Med ; 202(4): 461-5, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103406

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are specialized producers of type I interferons (IFNs) that respond to most viruses. Because of their antiviral activity and regulatory functions in innate and adaptive immunity, type I IFNs are important not only for antiviral resistance but also in other types of infections and in immune pathology. Here we discuss recent data that begin to reveal the unique molecular mechanisms underlying the remarkably rapid and efficient type I IFN production by pDCs.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Interferon Type I/biosynthesis , Lymphocyte Activation/immunology , Plasma Cells/immunology , Viruses/immunology , Animals , Dendritic Cells/ultrastructure , Humans , Plasma Cells/ultrastructure
8.
J Exp Med ; 201(7): 1157-67, 2005 Apr 04.
Article in English | MEDLINE | ID: mdl-15795237

ABSTRACT

Differential expression of Toll-like receptor (TLR) by conventional dendritic cells (cDCs) and plasmacytoid DC (pDCs) has been suggested to influence the type of immune response induced by microbial pathogens. In this study we show that, in vivo, cDCs and pDCs are equally activated by TLR4, -7, and -9 ligands. Type I interferon (IFN) was important for pDC activation in vivo in response to all three TLR ligands, whereas cDCs required type I IFN signaling only for TLR9- and partially for TLR7-mediated activation. Although TLR ligands induced in situ migration of spleen cDC into the T cell area, spleen pDCs formed clusters in the marginal zone and in the outer T cell area 6 h after injection of TLR9 and TLR7 ligands, respectively. In vivo treatment with TLR9 ligands decreased pDC ability to migrate ex vivo in response to IFN-induced CXCR3 ligands and increased their response to CCR7 ligands. Unlike cDCs, the migration pattern of pDCs required type I IFN for induction of CXCR3 ligands and responsiveness to CCR7 ligands. These data demonstrate that mouse pDCs differ from cDCs in the in vivo response to TLR ligands, in terms of pattern and type I IFN requirement for activation and migration.


Subject(s)
Cell Movement/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Interferon Type I/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Chemotaxis/immunology , Female , Flow Cytometry , Imidazoles , Immunohistochemistry , Ligands , Mice , Mice, Knockout , Oligodeoxyribonucleotides , Specific Pathogen-Free Organisms , Spleen/immunology , Toll-Like Receptor 4 , Toll-Like Receptor 7 , Toll-Like Receptors
9.
Cancers (Basel) ; 13(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884993

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAF) are heterogeneous with multiple functions in breast cancer. Recently, we identified a specific CAF subpopulation (referred to as CAF-S1), which promotes immunosuppression and immunotherapy resistance. METHODS AND RESULTS: Here, by studying a large collection of human samples, we highlight the key function of CD73/NT5E in CAF-S1-mediated immunosuppression in breast cancer. We first reveal that CD73 protein level specifically accumulates in CAF-S1 in breast cancer patients. Interestingly, infiltration of regulatory T lymphocytes (Tregs) is significantly correlated with CD73 expression in stroma but not in epithelium, indicating that CD73 contributes to immunosuppression when expressed in CAF-S1 and not in tumor cells. By performing functional assays based on relevant systems using primary CAF-S1 isolated from patients, we demonstrate that CAF-S1 increase the content in both PD-1+ and CTLA-4+ Tregs. Importantly, the use of a blocking anti-CD73 antibody on CAF-S1 reduces CAF-S1-mediated immunosuppression by preventing expression of these immune checkpoints on Tregs. CONCLUSIONS: Our data support the potential clinical benefit of using both anti-CD73 and immune-checkpoint inhibitors in breast cancer patients for inhibiting CAF-S1-mediated immunosuppression and enhancing anti-tumor immune response.

10.
Open Res Eur ; 1: 107, 2021.
Article in English | MEDLINE | ID: mdl-35967081

ABSTRACT

Background: MICA and MICB are tightly regulated stress-induced proteins that trigger the immune system by binding to the activating receptor NKG2D on cytotoxic lymphocytes. MICA and MICB are highly polymorphic molecules with prevalent expression on several types of solid tumors and limited expression in normal/healthy tissues, making them attractive targets for therapeutic intervention. Methods: We have generated a series of anti-MICA and MICB cross-reactive antibodies with the unique feature of binding to the most prevalent isoforms of both these molecules. Results: The anti-MICA and MICB antibody MICAB1, a human IgG1 Fc-engineered monoclonal antibody (mAb), displayed potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) of MICA/B-expressing tumor cells in vitro. However, it showed insufficient efficiency against solid tumors in vivo, which prompted the development of antibody-drug conjugates (ADC). Indeed, optimal tumor control was achieved with MICAB1-ADC format in several solid tumor models, including patient-derived xenografts (PDX) and carcinogen-induced tumors in immunocompetent MICAgen transgenic mice. Conclusions: These data indicate that MICA and MICB are promising targets for cytotoxic immunotherapy.

11.
J Exp Med ; 195(7): 953-8, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11927638

ABSTRACT

Plasmacytoid predendritic cells or type 1 interferon (IFN)-producing cells (IPCs) have recently been identified in mice. Although culture systems giving rise to different murine dendritic cell subsets have been established, the developmental regulation of murine plasmacytoid IPCs and the culture conditions leading to their generation remain unknown. Here we show that large numbers of over 40% pure CD11c(+)CD11b(-)B220(+)Gr-1(+) IPCs can be generated from mouse bone marrow cultures with FLT3-ligand. By contrast GM-CSF or TNF-alpha, which promote the generation of CD11c(+)CD11b(+)B220(-) myeloid DCs, block completely the development of IPCs. IPCs generated display similar features to human IPCs, such as the plasmacytoid morphology, the ability to produce large amounts of IFN-alpha in responses to herpes simplex virus, and the capacity to respond to ligands for Toll-like receptor 9 (TLR-9; CpG ODN 1668), but not to ligands for TLR-4 (lipopolysaccharide [LPS]). Unlike human IPCs which produce little IL-12p70, mouse IPCs produce IL-12p70 in response to CpG ODN 1668 and herpes simplex virus. This study demonstrates that the development of murine CD11c(+)CD11b(-)B220(+)Gr-1(+) IPCs and CD11c(+)CD11b(+)B220(-) myeloid DCs is differentially regulated by FLT3-ligand and granulocyte/macrophage colony-stimulating factor. Human IPCs and mouse IPCs display different ability to produce IL-12p70. Large numbers of mouse IPCs can now be obtained from total bone marrow culture.


Subject(s)
Dendritic Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Animals , Antigens, Differentiation/analysis , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Differentiation , Cells, Cultured , Dendritic Cells/drug effects , Female , Gene Expression Regulation , Hematopoiesis , Humans , Kinetics , Macrophage-1 Antigen/analysis , Membrane Proteins , Mice , Mice, Inbred BALB C , Species Specificity , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
12.
J Exp Med ; 195(4): 507-16, 2002 Feb 18.
Article in English | MEDLINE | ID: mdl-11854363

ABSTRACT

An effective type I interferon (IFN-alpha/beta) response is critical for the control of many viral infections. Here we show that in vesicular stomatitis virus (VSV)-infected mouse embryonic fibroblasts (MEFs) the production of IFN-alpha is dependent on type I IFN receptor (IFNAR) triggering, whereas in infected mice early IFN-alpha production is IFNAR independent. In VSV-infected mice type I IFN is produced by few cells located in the marginal zone of the spleen. Unlike other dendritic cell (DC) subsets, FACS((R))-sorted CD11c(int)CD11b(-)GR-1(+) DCs show high IFN-alpha expression, irrespective of whether they were isolated from VSV-infected IFNAR-competent or -deficient mice. Thus, VSV preferentially activates a specialized DC subset presumably located in the marginal zone to produce high-level IFN-alpha largely independent of IFNAR feedback signaling.


Subject(s)
Dendritic Cells/metabolism , Dendritic Cells/virology , Interferon-alpha/biosynthesis , Receptors, Interferon/deficiency , Receptors, Interferon/metabolism , Vesicular stomatitis Indiana virus/physiology , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Dendritic Cells/drug effects , Feedback, Physiological , Fibroblasts/metabolism , Fibroblasts/virology , Flow Cytometry , Gene Deletion , Gene Expression Regulation/drug effects , In Situ Hybridization , Interferon Regulatory Factor-7 , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-beta/metabolism , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Poly I-C/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Signal Transduction/drug effects , Spleen/metabolism , Spleen/pathology , Time Factors , Ultraviolet Rays
13.
J Exp Med ; 195(4): 517-28, 2002 Feb 18.
Article in English | MEDLINE | ID: mdl-11854364

ABSTRACT

Interferon (IFN)-alpha/beta and interleukin (IL)-12 are cytokines critical in defense against viruses, but their cellular sources and mechanisms of regulation for in vivo expression remain poorly characterized. The studies presented here identified a novel subset of dendritic cells (DCs) as major producers of the cytokines during murine cytomegalovirus (MCMV) but not lymphocytic choriomeningitis virus (LCMV) infections. These DCs differed from those activated by Toxoplasma antigen but were related to plasmacytoid cells, as assessed by their CD8alpha(+)Ly6G/C(+)CD11b(-) phenotype. Another DC subset (CD8alpha(2)Ly6G/C(-)CD11b(+)) also contributed to IL-12 production in MCMV-infected immunocompetent mice, modestly. However, it dramatically increased IL-12 expression in the absence of IFN-alpha/beta functions. Conversely, IFN-alpha/beta production was greatly reduced under these conditions. Thus, a cross-regulation of DC subset cytokine responses was defined, whereby secretion of type I IFNs by CD8alpha(+) DCs resulted in responses limiting IL-12 expression by CD11b(+) DCs but enhancing overall IFN-alpha/beta production. Taken together, these data indicate that CD8alpha(+)Ly6G/C(+)CD11b(-) DCs play important roles in limiting viral replication and regulating immune responses, through cytokine production, in some but not all viral infections. They also illustrate the plasticity of cellular sources for innate cytokines in vivo and provide new insights into the roles of IFNs in shaping immune responses to viruses.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Interferon-alpha/immunology , Interferon-beta/immunology , Interleukin-12/immunology , Lymphocytic choriomeningitis virus/immunology , Muromegalovirus/immunology , Animals , Antigens, Viral/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/cytology , Flow Cytometry , Immunohistochemistry , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interleukin-12/biosynthesis , Interleukin-12/genetics , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Receptors, Interferon/metabolism , STAT1 Transcription Factor , Signal Transduction , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Trans-Activators/genetics , Trans-Activators/metabolism
14.
J Exp Med ; 197(1): 101-9, 2003 Jan 06.
Article in English | MEDLINE | ID: mdl-12515817

ABSTRACT

Distinct dendritic cell (DC) subsets have been suggested to be preprogrammed to direct either T helper cell (Th) type 1 or Th2 development, although more recently different pathogen products or stimuli have been shown to render these DCs more flexible. It is still unclear how distinct mouse DC subsets cultured from bone marrow precursors, blood, or their lymphoid tissue counterparts direct Th differentiation. We show that mouse myeloid and plasmacytoid precursor DCs (pDCs) cultured from bone marrow precursors and ex vivo splenic DC subsets can induce the development of both Th1 and Th2 effector cells depending on the dose of antigen. In general, high antigen doses induced Th1 cell development whereas low antigen doses induced Th2 cell development. Both cultured and ex vivo splenic plasmacytoid-derived DCs enhanced CD4(+) T cell proliferation and induced strong Th1 cell development when activated with the Toll-like receptor (TLR)9 ligand CpG, and not with the TLR4 ligand lipopolysaccharide (LPS). The responsiveness of plasmacytoid pDCs to CpG correlated with high TLR9 expression similarly to human plasmacytoid pDCs. Conversely, myeloid DCs generated with granulocyte/macrophage colony-stimulating factor enhanced Th1 cell development when stimulated with LPS as a result of their high level of TLR4 expression. Polarized Th1 responses resulting from high antigen dose were not additionally enhanced by stimulation of DCs by TLR ligands. Thus, the net effect of antigen dose, the state of maturation of the DCs together with the stimulation of DCs by pathogen-derived products, will determine whether a Th1 or Th2 response develops.


Subject(s)
Antigens/immunology , Cell Differentiation , Dendritic Cells/immunology , Drosophila Proteins , Membrane Glycoproteins/metabolism , Plasma Cells/immunology , Receptors, Cell Surface/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Division , CpG Islands/immunology , Dendritic Cells/cytology , Dose-Response Relationship, Immunologic , Flow Cytometry , Gene Expression Regulation , Lipopolysaccharides/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Plasma Cells/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Stem Cells/cytology , Stem Cells/immunology , Th1 Cells/cytology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism , Toll-Like Receptor 4 , Toll-Like Receptors
15.
BMC Cancer ; 10: 327, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20576118

ABSTRACT

BACKGROUND: There is increasing evidence that the toll-like receptor 3 (TLR3) is an interesting target for anti-cancer therapy. Unfortunately, most laboratory investigations about the impact of TLR3 stimulation on human malignant cells have been performed with very high concentrations--5 to 100 microg/ml--of the prototype TLR3 ligand, poly(I:C). In a previous study focused on a specific type of human carcinoma - nasopharyngeal carcinoma - we have shown that concentrations of poly(I:C) as low as 100 ng/ml are sufficient to induce apoptosis of malignant cells when combined to a pharmacological antagonist of the IAP family based on Smac mimicry. METHODS: This observation prompted us to investigate the contribution of the IAP family in cell response to poly(I:C) in a variety of human malignant cell types. RESULTS: We report a rapid, intense and selective increase in c-IAP2 protein expression observed under stimulation by poly(I:C)(500 ng/ml) in all types of human malignant cells. In most cell types, this change in protein expression is underlain by an increase in c-IAP2 transcripts and dependent on the TLR3/TRIF pathway. When poly(I:C) is combined to the IAP inhibitor RMT 5265, a cooperative effect in apoptosis induction and/or inhibition of clonogenic growth is obtained in a large fraction of carcinoma and melanoma cell lines. CONCLUSIONS: Currently, IAP inhibitors like RMT 5265 and poly(I:C) are the subject of separate therapeutic trials. In light of our observations, combined use of both types of compounds should be considered for treatment of human malignancies including carcinomas and melanomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Neoplasms/pathology , Adaptor Proteins, Vesicular Transport/metabolism , Adult , Baculoviral IAP Repeat-Containing 3 Protein , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Male , Middle Aged , Neoplasms/genetics , Neoplasms/metabolism , Poly I-C/pharmacology , RNA Interference , RNA, Messenger/metabolism , Time Factors , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Up-Regulation
16.
Cell Rep ; 27(8): 2411-2425.e9, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31116985

ABSTRACT

Immune checkpoint inhibitors have revolutionized cancer treatment. However, many cancers are resistant to ICIs, and the targeting of additional inhibitory signals is crucial for limiting tumor evasion. The production of adenosine via the sequential activity of CD39 and CD73 ectoenzymes participates to the generation of an immunosuppressive tumor microenvironment. In order to disrupt the adenosine pathway, we generated two antibodies, IPH5201 and IPH5301, targeting human membrane-associated and soluble forms of CD39 and CD73, respectively, and efficiently blocking the hydrolysis of immunogenic ATP into immunosuppressive adenosine. These antibodies promoted antitumor immunity by stimulating dendritic cells and macrophages and by restoring the activation of T cells isolated from cancer patients. In a human CD39 knockin mouse preclinical model, IPH5201 increased the anti-tumor activity of the ATP-inducing chemotherapeutic drug oxaliplatin. These results support the use of anti-CD39 and anti-CD73 monoclonal antibodies and their combination with immune checkpoint inhibitors and chemotherapies in cancer.


Subject(s)
5'-Nucleotidase/immunology , Antibodies, Blocking/immunology , Antigens, CD/immunology , Apyrase/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Antibodies, Blocking/therapeutic use , Antigens, CD/genetics , Antineoplastic Agents/therapeutic use , Apyrase/deficiency , Apyrase/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Knock-In Techniques , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Melanoma/drug therapy , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxaliplatin/therapeutic use , Survival Rate , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment
17.
Oncotarget ; 9(25): 17675-17688, 2018 Apr 03.
Article in English | MEDLINE | ID: mdl-29707140

ABSTRACT

PURPOSE: Anti-KIR monoclonal antibodies (mAbs) can enhance the antitumor responses of natural killer (NK) cells. We evaluated the safety of the anti-KIR2D mAb lirilumab in patients with various cancers. EXPERIMENTAL DESIGN: Thirty-seven patients with hematological malignancies (n = 22) or solid tumors (n = 15) were included in the study. Dose escalation (0.015 to 10 mg/kg) was conducted following a 3 + 3 design. Patients were scheduled to receive four cycles of treatment. In a second (extension) phase 17 patients were treated at 0.015 (n = 9) or 3 mg/kg (n = 8). RESULTS: No dose-limiting toxicity was recorded. The most frequent lirilumab-related adverse events were pruritus (19%), asthenia (16%), fatigue (14%), infusion-related reaction (14%), and headache (11%), mostly mild or moderate. Pharmacokinetics was dose-dependent and linear, with minimal accumulation resulting from the 4-weekly repeated administrations. Full KIR occupancy (>95%) was achieved with all dosages, and the duration of occupancy was dose-related. No significant changes were observed in the number or distribution of lymphocyte subpopulations, nor was any reduction in the distribution of KIR2D-positive NK cells. CONCLUSIONS: This phase 1 trial demonstrated the satisfactory safety profile of lirilumab up to doses that enable full and sustained blockade of KIR.

19.
Cancer Res ; 74(21): 6060-70, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25361998

ABSTRACT

Advanced cutaneous T-cell lymphoma (CTCL) remains an unmet medical need, which lacks effective targeted therapies. In this study, we report the development of IPH4102, a humanized monoclonal antibody that targets the immune receptor KIR3DL2, which is widely expressed on CTCL cells but few normal immune cells. Potent antitumor properties of IPH4102 were documented in allogeneic human CTCL cells and a mouse model of KIR3DL2(+) disease. IPH4102 antitumor activity was mediated by antibody-dependent cell cytotoxicity and phagocytosis. IPH4102 improved survival and reduced tumor growth in mice inoculated with KIR3DL2(+) tumors. Ex vivo efficacy was further evaluated in primary Sézary patient cells, sorted natural killer-based autologous assays, and direct spiking into Sézary patient peripheral blood mononuclear cells. In these settings, IPH4102 selectively and efficiently killed primary Sézary cells, including at unfavorable effector-to-target ratios characteristic of unsorted PBMC. Together, our results offer preclinical proof of concept for the clinical development of IPH4102 to treat patients with advanced CTCL.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/immunology , Receptors, KIR3DL2/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Humans , Lymphoma, T-Cell, Cutaneous/pathology , Mice , Neoplasm Staging , Receptors, KIR3DL2/biosynthesis
20.
PLoS One ; 8(8): e72112, 2013.
Article in English | MEDLINE | ID: mdl-23977222

ABSTRACT

Using a yeast-based assay, a previously unsuspected antiprion activity was found for imiquimod (IQ), a potent Toll-like receptor 7 (TLR7) agonist already used for clinical applications. The antiprion activity of IQ was first detected against yeast prions [PSI (+) ] and [URE3], and then against mammalian prion both ex vivo in a cell-based assay and in vivo in a transgenic mouse model for prion diseases. In order to facilitate structure-activity relationship studies, we conducted a new synthetic pathway which provides a more efficient means of producing new IQ chemical derivatives, the activity of which was tested against both yeast and mammalian prions. The comparable antiprion activity of IQ and its chemical derivatives in the above life forms further emphasizes the conservation of prion controlling mechanisms throughout evolution. Interestingly, this study also demonstrated that the antiprion activity of IQ and IQ-derived compounds is independent from their ability to stimulate TLRs. Furthermore, we found that IQ and its active chemical derivatives inhibit the protein folding activity of the ribosome (PFAR) in vitro.


Subject(s)
Aminoquinolines/pharmacology , Glutathione Peroxidase/metabolism , Peptide Termination Factors/metabolism , Prion Diseases/drug therapy , Prions/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Aminoquinolines/chemical synthesis , Animals , Cell Line , Drug Evaluation, Preclinical , Guanosine/analogs & derivatives , Guanosine/pharmacology , Humans , Imidazoles/pharmacology , Imiquimod , Membrane Glycoproteins/agonists , Membrane Glycoproteins/metabolism , Mice , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Protein Folding , Saccharomyces cerevisiae/drug effects , Structure-Activity Relationship , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL