Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 3(10): 100783, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36260981

ABSTRACT

Harnessing innate immunity is emerging as a promising therapeutic approach in cancer. We report here the design of tetraspecific molecules engaging natural killer (NK) cell-activating receptors NKp46 and CD16a, the ß-chain of the interleukin-2 receptor (IL-2R), and a tumor-associated antigen (TAA). In vitro, these tetraspecific antibody-based natural killer cell engager therapeutics (ANKETs) induce a preferential activation and proliferation of NK cells, and the binding to the targeted TAA triggers NK cell cytotoxicity and cytokine and chemokine production. In vivo, tetraspecific ANKETs induce NK cell proliferation and their accumulation at the tumor bed, as well as the control of local and disseminated tumors. Treatment of non-human primates with CD20-directed tetraspecific ANKET leads to CD20+ circulating B cell depletion, with minimal systemic cytokine release and no sign of toxicity. Tetraspecific ANKETs, thus, constitute a technological platform for harnessing NK cells as next-generation cancer immunotherapies.


Subject(s)
Interleukin-2 , Neoplasms , Animals , Interleukin-2/genetics , Killer Cells, Natural , Receptors, Interleukin-2/metabolism , Cytokines , Neoplasms/genetics , Chemokines/metabolism
2.
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.

4.
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
5.
Int J Cancer ; 136(5): 1085-94, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25046660

ABSTRACT

Dendritic cells (DCs) cross-present antigen (Ag) to initiate T-cell immunity against most infections and tumors. Natural killer (NK) cells are innate cytolytic lymphocytes that have emerged as key modulators of multiple DC functions. Here, we show that human NK cells promote cross-presentation of tumor cell-derived Ag by DC leading to Ag-specific CD8(+) T-cell activation. Surprisingly, cytotoxic function of NK cells was not required. Instead, we highlight a critical and nonredundant role for IFN-γ and TNF-α production by NK cells to enhance cross-presentation by DC using two different Ag models. Importantly, we observed that NK cells promote cell-associated Ag cross-presentation selectively by monocytes-derived DC (Mo-DC) and CD34-derived CD11b(neg) CD141(high) DC subsets but not by myeloid CD11b(+) DC. Moreover, we demonstrate that triggering NK cell activation by monoclonal antibodies (mAbs)-coated tumor cells leads to efficient DC cross-presentation, supporting the concept that NK cells can contribute to therapeutic mAbs efficiency by inducing downstream adaptive immunity. Taken together, our findings point toward a novel role of human NK cells bridging innate and adaptive immunity through selective induction of cell-associated Ag cross-presentation by CD141(high) DC, a process that could be exploited to better harness Ag-specific cellular immunity in immunotherapy.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Antibody-Dependent Cell Cytotoxicity , Dendritic Cells/pathology , Humans , Killer Cells, Natural/pathology , Neoplasms/pathology , Tumor Cells, Cultured
6.
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
7.
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
8.
Proc Natl Acad Sci U S A ; 104(19): 8047-52, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17463087

ABSTRACT

TRIF is an adaptor protein associated with the signaling by Toll-like receptor (TLR)3 and TLR4 for the induction of type I IFNs. Here, we demonstrate a mechanism by which TLR signaling controls cell proliferation and survival. We show that TLR3 and TLR4 can induce cell cycle entry via TRIF, which targets the cell cycle inhibitor p27(kip1) for relocalization, phosphorylation by cyclin/cdk complexes, and proteasome degradation. These events are antagonized by type I IFN induced by the TRIF pathway. Furthermore, in human dendritic cells treated with TLR3, TLR4, or TLR5 ligands, we demonstrate that IFN signaling modulates p27(kip1) degradation and apoptosis, identifying an immunoregulatory "switching" function of type I IFNs. These findings reveal a previously uncharacterized function of TLR signaling in cell proliferation and survival.


Subject(s)
Interferon Type I/physiology , Signal Transduction/physiology , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/physiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Apoptosis/drug effects , Cell Proliferation , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/physiology , Dendritic Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , NF-kappa B/genetics , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/physiology , Rats
9.
J Immunol ; 178(5): 2763-9, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17312119

ABSTRACT

The efficacy of immune response to control human cancer remains controversial. It is particularly debated whether and to what extent the capacity of tumor-infiltrating dendritic cells (DC) to drive immunization can be turned off by transformed cells, leading to tumor-specific tolerance rather than immunization. To address this issue, we have characterized the DC isolated from human non-small cell lung cancer (NSCLC). These biopsy specimens contained CD11c(high) myeloid DC (mDC), but also CD11c(-) plasmacytoid DC (pDC) and a third DC subset expressing intermediate level of CD11c. Compared with peripheral blood, CD11c(high) tumor-infiltrating DC (TIDC) displayed a "semi-mature" phenotype, and TLR4 or TLR8 stimulation drove them to mature partially and to secrete limited amounts of cytokines. In contrast, most tumor-infiltrating pDC were immature but underwent partial maturation after TLR7 activation, whereas TLR9 ligation triggered low secretion of IFN-alpha. CD11c(int) mDC represented approximately 25% of total DC in tumoral and peritumoral tissues and expressed low levels of costimulatory molecules contrasting with high levels of the immunoinhibitory molecule B7-H1. Finally, the poor APC function of total TIDC even after TLR stimulation and the migratory response of both tumor-infiltrating mDC and pDC toward CCL21 and SDF-1 in vitro suggested their ability to compromise the tumor-specific immune response in draining lymph nodes in vivo. Further studies will be required to establish the specific role of the three TIDC subsets in tumor immunity and to draw conclusions for the design of therapeutic strategies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Myeloid Cells/immunology , Tumor Escape , CD11c Antigen/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Chemokine CCL21 , Chemokine CXCL12 , Chemokines, CC/immunology , Chemokines, CXC/immunology , Humans , Toll-Like Receptors/immunology , Tumor Cells, Cultured , Vaccination
10.
J Immunol ; 176(9): 5293-8, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16621995

ABSTRACT

Human CD4(+)CD25(+) regulatory T cells (Treg) play an essential role in the prevention of autoimmune diseases. However, the mechanisms of immune suppression and the spectrum of cells they target in vivo remain incompletely defined. In particular, although Treg directly suppress conventional T cells in vitro, they have been shown to inhibit the Ag-presenting functions of macrophage- and monocyte-derived dendritic cells (DC). We have now studied the maturation of human blood-derived myeloid DC and plasmacytoid DC activated with TLR ligands in the presence of Treg. Preactivated Treg suppressed strongly TLR-triggered myeloid DC maturation, as judged by the blocking of costimulatory molecule up-regulation and the inhibition of proinflammatory cytokines secretion that resulted in poor Ag presentation capacity. Although IL-10 played a prominent role in inhibiting cytokines secretion, suppression of phenotypic maturation required cell-cell contact and was independent of TGF-beta and CTLA-4. In contrast, the acquisition of maturation markers and production of cytokines by plasmacytoid DC triggered with TLR ligands were insensitive to regulatory T cells. Therefore, human Treg may enlist myeloid, but not plasmacytoid DC for the initiation and the amplification of tolerance in vivo by restraining their maturation after TLR stimulation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Antigen Presentation , Cell Communication , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Phenotype , T-Lymphocytes, Regulatory/cytology , Toll-Like Receptors/metabolism
11.
Eur J Immunol ; 34(2): 418-26, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14768046

ABSTRACT

This work aims to further characterize the newly described leukemic plasmacytoid dendritic cells (LPDC), for which we had previously demonstrated their normal, PDC-like ability to produce IFN-alpha. In addition, LPDC also express the specific antigens BDCA-2 and BDCA-4. Importantly, they become fully competent antigen-presenting cells (APC) after a short maturation induced by IL-3 + CD40L or virus, exhibiting a characteristic APC phenotype (high expression of CD83 and of the costimulatory molecules CD40, CD80, CD86). Whereas IL-3 + CD40L-activated LPDC prime naive CD4(+) T cells towards a Th2 pathway (IL-4-secreting T cells), virus-activated LPDC drive a Th1 profile (IFN-gamma-secreting T cells). Moreover, we show in one case that LPDC are able to capture, process and present exogenous antigens, leading to the activation of both CD4(+) and CD8(+) T cell clones in an antigen-specific manner. This study further characterizes the phenotype and immunological functions of LPDC.


Subject(s)
Dendritic Cells/immunology , Leukemia/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antigen Presentation , Antigens, Surface/immunology , CD40 Ligand/immunology , Cell Differentiation/immunology , Child , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Female , Flow Cytometry , Humans , Immunophenotyping , Interleukin-3/immunology , Leukemia/pathology , Male , Middle Aged , Orthomyxoviridae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL