Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Sci ; 2024 May 04.
Article in English | MEDLINE | ID: mdl-38702996

ABSTRACT

The study of anticancer immune responses and in particular the action of immune checkpoint inhibitors that overcome T cell inhibition has revolutionized metastatic patients' care. Unfortunately, many patients are resistant to these innovative immunotherapies. Over the last decade, several immune checkpoint inhibitors, currently available in the clinic, have been developed, such as anti-PD-1/PD-L1 or anti-CTLA-4. More recently, other immune checkpoints have been characterized, among them lymphocyte activation gene 3 (LAG-3). LAG-3 has been the subject of numerous therapeutic studies and may be involved in cancer-associated immune resistance phenomena. This review summarizes the latest knowledge on LAG-3 as an immunotherapeutic target, particularly in combination with standard or innovative therapies. Indeed, many studies are looking at combining LAG-3 inhibitors with chemotherapeutic, immunotherapeutic, radiotherapeutic treatments, or adoptive cell therapies to potentiate their antitumor effects and/or to overcome patients' resistance. We will particularly focus on the association therapies that are currently in phase III clinical trials and innovative combinations in preclinical phase. These new discoveries highlight the possibility of developing other types of therapeutic combinations currently unavailable in the clinic, which could broaden the therapeutic spectrum of personalized medicine.

2.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35091453

ABSTRACT

BACKGROUND: While stimulator of interferon genes (STING) activation in innate immune cells of the tumor microenvironment can result in CD8 T cell-dependent antitumor immunity, whether STING signaling affects CD4 T-cell responses remains elusive. METHODS: Here, we tested whether STING activation modulated the effector functions of CD4 T cells in vivo by analyzing tumor-infiltrating CD4 T cells and evaluating the contribution of the CD4 T cell-derived cytokines in the antitumor activity of the STING ligand 2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) in two mouse tumor models. We performed ex vivo experiments to assess the impact of STING activation on CD4 T-cell differentiation and investigate the underlying molecular mechanisms. Finally, we tested whether STING activation enhances TH9 cell antitumor activity against mouse melanoma upon adoptive transfer. RESULTS: We found that activation of STING signaling cell-intrinsically enhances the differentiation and antitumor functions of TH1 and TH9 cells by increasing their respective production of interferon gamma (IFN-γ) and interleukin-9. IRF3 and type I interferon receptors (IFNARs) are required for the STING-driven enhancement of TH1 cell differentiation. However, STING activation favors TH9 cell differentiation independently of the IFNARs/IRF3 pathway but through mammalian target of rapamycin (mTOR) signaling, underscoring that STING activation differentially affects the fate of distinct CD4 T-cell subsets. The therapeutic effect of STING activation relies on TH1 and TH9-derived cytokines, and STING activation enhances the antitumor activity of TH9 cells upon adoptive transfer. CONCLUSION: Our results reveal the STING signaling pathway as a therapeutic target to boost CD4 T-cell effector functions and antitumor immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-9/physiology , Membrane Proteins/physiology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Female , Interferon Regulatory Factor-3/physiology , Mice , Mice, Inbred C57BL , Nucleotides, Cyclic/pharmacology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Th1 Cells/cytology
3.
FASEB J ; 35(12): e21997, 2021 12.
Article in English | MEDLINE | ID: mdl-34719814

ABSTRACT

The deadliest complication of infection by Plasmodium parasites, cerebral malaria, accounts for the majority of malarial fatalities. Although our understanding of the cellular and molecular mechanisms underlying the pathology remains incomplete, recent studies support the contribution of systemic and neuroinflammation as the cause of cerebral edema and blood-brain barrier (BBB) dysfunction. All Plasmodium species encode an orthologue of the innate cytokine, Macrophage Migration Inhibitory Factor (MIF), which functions in mammalian biology to regulate innate responses. Plasmodium MIF (PMIF) similarly signals through the host MIF receptor CD74, leading to an enhanced inflammatory response. We investigated the PMIF-CD74 interaction in the onset of experimental cerebral malaria (ECM) and liver stage Plasmodium development by using a combination of CD74 deficient (Cd74-/- ) hosts and PMIF deficient parasites. Cd74-/- mice were found to be protected from ECM and the protection was associated with the inability of brain microvessels to present parasite antigen to sequestered and pathogenic Plasmodium-specific CD8+ T cells. Infection of WT hosts with PMIF-deficient sporozoites or infection of Cd74-/- hosts with WT sporozoites impacted the survival of infected hepatocytes and subsequently reduced blood-stage associated inflammation, contributing to protection from ECM. We recapitulated these finding with a novel pharmacologic PMIF-selective antagonist that reduced PMIF/CD74 signaling and fully protected mice from ECM. These findings reveal a conserved mechanism for Plasmodium usurpation of host CD74 signaling and suggest a tractable approach for new pharmacologic intervention.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/chemistry , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/chemistry , Inflammation/prevention & control , Liver/pathology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Malaria, Cerebral/prevention & control , Plasmodium berghei/physiology , Animals , Antigens, Differentiation, B-Lymphocyte/physiology , Histocompatibility Antigens Class II/physiology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Liver/immunology , Liver/parasitology , Macrophage Migration-Inhibitory Factors/metabolism , Malaria, Cerebral/etiology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Nat Commun ; 9(1): 2714, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30006528

ABSTRACT

Plasmodium species produce an ortholog of the cytokine macrophage migration inhibitory factor, PMIF, which modulates the host inflammatory response to malaria. Using a novel RNA replicon-based vaccine, we show the impact of PMIF immunoneutralization on the host response and observed improved control of liver and blood-stage Plasmodium infection, and complete protection from re-infection. Vaccination against PMIF delayed blood-stage patency after sporozoite infection, reduced the expression of the Th1-associated inflammatory markers TNF-α, IL-12, and IFN-γ during blood-stage infection, augmented Tfh cell and germinal center responses, increased anti-Plasmodium antibody titers, and enhanced the differentiation of antigen-experienced memory CD4 T cells and liver-resident CD8 T cells. Protection from re-infection was recapitulated by the adoptive transfer of CD8 or CD4 T cells from PMIF RNA immunized hosts. Parasite MIF inhibition may be a useful approach to promote immunity to Plasmodium and potentially other parasite genera that produce MIF orthologous proteins.


Subject(s)
Adaptive Immunity/drug effects , Antibodies, Protozoan/biosynthesis , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Protozoan Proteins/antagonists & inhibitors , Vaccines, DNA/administration & dosage , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Female , Gene Expression , Germinal Center/drug effects , Germinal Center/immunology , Germinal Center/parasitology , Immunologic Memory/drug effects , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/biosynthesis , Mice , Mice, Inbred BALB C , Plasmodium berghei/drug effects , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , RNA, Protozoan/genetics , RNA, Protozoan/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccines, DNA/biosynthesis
5.
J Med Chem ; 57(20): 8652-6, 2014 10 23.
Article in English | MEDLINE | ID: mdl-25268646

ABSTRACT

We report the crystal structures of two inhibitors of Plasmodium falciparum macrophage migration inhibitory factor (PfMIF) with nanomolar Ki's, analyze their interactions with the active site of PfMIF, and provide explanations regarding their selectivity of PfMIF versus human MIF. These inhibitors were also found to selectively inhibit interactions between PfMIF and the human MIF receptor CD74. The results of this study provide the framework for the development of new therapeutics that target PfMIF.


Subject(s)
Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/chemistry , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Antigens, Differentiation, B-Lymphocyte/metabolism , Antimalarials/chemistry , Antimalarials/pharmacology , Catalytic Domain , Crystallography, X-Ray , Histocompatibility Antigens Class II/metabolism , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Models, Molecular , Protein Stability , Protozoan Proteins/metabolism
6.
J Med Chem ; 55(22): 10148-59, 2012 11 26.
Article in English | MEDLINE | ID: mdl-23067344

ABSTRACT

The Plasmodium falciparum orthologue of the human cytokine, macrophage migratory inhibitory factor (PfMIF), is produced by the parasite during malaria infection and modulates the host's immune response. As for other MIF orthologues, PfMIF has tautomerase activity, whose inhibition may influence the cytokine activity. To identify small-molecule inhibitors of the tautomerase activity of PfMIF, virtual screening has been performed by docking 2.1 million compounds into the enzymatic site. Assaying of 17 compounds identified four as active. Substructure search for the most potent of these compounds, a 4-phenoxypyridine analogue, identified four additional compounds that were purchased and also shown to be active. Thirty-one additional analogues were then designed, synthesized, and assayed. Three were found to be potent PfMIF tautomerase inhibitors with K(i) of ∼40 nM; they are also highly selective with K(i) > 100 µM for human MIF.


Subject(s)
Antimalarials/pharmacology , High-Throughput Screening Assays , Isomerases/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Malaria/drug therapy , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Antimalarials/chemistry , Humans , Hydrogen Bonding , Malaria/metabolism , Malaria/parasitology , Models, Molecular , Molecular Structure , Protein Binding , Structure-Activity Relationship
7.
PLoS Pathog ; 6(9): e1001115, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886098

ABSTRACT

We have identified and characterized a Macrophage Migration Inhibitory Factor (MIF) family member in the Lophotrochozoan invertebrate, Biomphalaria glabrata, the snail intermediate host of the human blood fluke Schistosoma mansoni. In mammals, MIF is a widely expressed pleiotropic cytokine with potent pro-inflammatory properties that controls cell functions such as gene expression, proliferation or apoptosis. Here we show that the MIF protein from B. glabrata (BgMIF) is expressed in circulating immune defense cells (hemocytes) of the snail as well as in the B. glabrata embryonic (Bge) cell line that has hemocyte-like features. Recombinant BgMIF (rBgMIF) induced cell proliferation and inhibited NO-dependent p53-mediated apoptosis in Bge cells. Moreover, knock-down of BgMIF expression in Bge cells interfered with the in vitro encapsulation of S. mansoni sporocysts. Furthermore, the in vivo knock-down of BgMIF prevented the changes in circulating hemocyte populations that occur in response to an infection by S. mansoni miracidia and led to a significant increase in the parasite burden of the snails. These results provide the first functional evidence that a MIF ortholog is involved in an invertebrate immune response towards a parasitic infection and highlight the importance of cytokines in invertebrate-parasite interactions.


Subject(s)
Biomphalaria/immunology , Hemocytes/physiology , Host-Parasite Interactions , Macrophage Migration-Inhibitory Factors/metabolism , Schistosoma mansoni/pathogenicity , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Amino Acid Sequence , Animals , Apoptosis , Biomphalaria/embryology , Biomphalaria/parasitology , Blotting, Western , Cell Proliferation , Cells, Cultured , Cricetinae , Humans , Liver/parasitology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/genetics , Molecular Sequence Data , Oocysts/metabolism , Oocysts/pathology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...