Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38893156

ABSTRACT

Immune check-point blockade (ICB) has revitalized cancer immunotherapy, showing unprecedented efficacy despite only a narrow number of indications and with limited long-term protection. Cancer vaccines are promising combination partners for ICB to widen the patient population profiting from these treatments. Therapeutic heterologous prime-boost vaccination with KISIMATM protein vaccine and VSV-GP-TAg oncolytic virus was shown to inflame the tumor microenvironment, promoting significant infiltration of antigen-specific CD8 T cells resulting in robust antitumoral efficacy in mouse tumor models, and clinical trials are currently ongoing. Here, we report the impact of NKG2A blockade on antitumoral CD8 T cell immune response elicited by KISIMA-VSV-GP-TAg vaccination in tumor mouse models. Combination therapy significantly reduced the amount of vaccine-induced exhausted CD8 T cells infiltrating the tumor, resulting in short-term improved tumor growth control and prolonged mouse survival, while it also influenced the establishment of systemic effector memory CD8 T cell response. Taken together, these data show a compartment-dependent effect of NKG2A blockade on cancer vaccine-induced T cell immunity, increasing intratumoral T cell efficacy and attenuating the development of peripheral effector memory CD8 T cell response.

2.
Cancers (Basel) ; 13(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34885215

ABSTRACT

Heterologous prime-boost settings with a protein vaccine and the viral vector vesicular stomatitis virus, both expressing tumor-associated antigens (KISIMA-TAA and VSV-GP-TAA), have been previously shown to generate potent antitumor immunity. In the cold TC-1 model (HPV antigen) and the immune-infiltrate MC-38 model (Adpgk, Reps1 and Rpl18 neo-antigens), we further investigated pivotal immune cells that educate CD8+ T cells. Heterologous prime-boost vaccination induced a superior antitumor response characterized by the increase in number and functionality of antigen-specific CD8+ T cells, recruitment of cross-presenting dendritic cells, and polarization of CD4+ T cells towards an antitumor Th1 phenotype within the tumor and tumor-draining lymph nodes, turning the cold TC-1 tumor into a hot, inflamed tumor. In the inflamed MC-38 tumor model, treatment combination markedly prolonged the overall survival of mice. Treatment with multi-epitope vaccines also induced high frequencies of multiple antigen specificities in the periphery and in the tumor. Prime-boost treatment reduced tumor-infiltrating regulatory CD4+ T cells whilst increasing cross-presenting dendritic cells in tumor-draining lymph nodes. In conclusion, heterologous prime-boost vaccination possesses the ability to induce a potent anti-tumor response in both immune-excluded and immune-infiltrated mouse tumor models. Additionally, this study highlights the design of a multi-epitope vaccine for cancer immunotherapy.

3.
Front Immunol ; 12: 695056, 2021.
Article in English | MEDLINE | ID: mdl-34276686

ABSTRACT

Combining different immunotherapy approaches is currently building the future of immunotherapy, with the view to maximize anti-tumoral efficacy for larger patient population. The KISIMA™ platform allows the development of protein-based cancer vaccines able to induce tumor-specific T cell response resulting in anti-tumoral efficacy in various mouse models. Intra-tumoral administration of stimulator of interferon gene agonists (STINGa) was shown to induce a potent inflammatory response leading to the development of tumor-specific immunity. Here, we explored the efficacy and mechanisms of action of subcutaneous STINGa treatment combined with therapeutic vaccination in various mouse tumor models. This combinatory treatment highly enhanced frequency and effector function of both peripheral and intra-tumoral antigen-specific CD8 T cells, promoting potent IFNγ and TNFα production along with increased cytotoxicity. Moreover, combination therapy favorably modulated the tumor microenvironment by dampening immune-suppressive cells and increasing CD4 T cell infiltration together with their polarization toward Th1 phenotype. Combination with STINGa treatment improved the effect of therapeutic vaccination, resulting in a prolonged control and slower growth of B16-OVA and TC-1 tumors. Altogether, the results presented here highlight the potential of combining STINGa with a therapeutic protein vaccine for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/pharmacology , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/drug therapy , Membrane Proteins/agonists , Skin Neoplasms/drug therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Female , Interferon-gamma/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Phenotype , Signal Transduction , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Burden/drug effects , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism , Vaccines, Subunit/pharmacology
4.
Cancers (Basel) ; 13(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671373

ABSTRACT

Novel immunopreventive strategies are emerging that show great promise for conferring long-term protection to individuals at high risk of developing colorectal cancer. The KISIMA vaccine platform utilizes a chimeric protein comprising: (1) a selected tumor antigen; (2) a cell-penetrating peptide to improve antigen delivery and epitope presentation, and (3) a TLR2/4 agonist to serve as a self-adjuvant. This study examines the ability of a KISIMA vaccine against achaete-scute family bHLH transcription factor 2 (Ascl2), an early colon cancer antigen, to reduce colon tumor formation by stimulating an anti-tumor immune response. Vaccine administrations were well-tolerated and led to circulating antibodies and antigen-specific T cells in a mouse model of colorectal cancer. To assess preventive efficacy, the vaccine was administered to mice either alone or in combination with the immune checkpoint inhibitor anti-PD-1. When delivered to animals prior to colon tumor formation, the combination strategy significantly reduced the development of colon microadenomas and adenomas, as compared to vehicle-treated controls. This response was accompanied by an increase in the intraepithelial density of CD3+ T lymphocytes. Together, these data indicate that the KISIMA-Ascl2 vaccine shows great potential to be a safe and potent immunopreventive intervention for individuals at high risk of developing colorectal cancer.

5.
JCI Insight ; 52019 04 23.
Article in English | MEDLINE | ID: mdl-31013258

ABSTRACT

Induction of a potent CD4 and CD8 T-cell response against tumor-specific and tumor-associated antigen is critical for eliminating tumor cells. Recent vaccination strategies have been hampered by an inefficacious and low amplitude immune response. Here we describe a self-adjuvanted chimeric protein vaccine platform to address these challenges, characterized by a multidomain construction incorporating (i) a cell penetrating peptide (CPP) allowing internalization of several multiantigenic Major Histocompatibility Complex (MHC)-restricted peptides within (ii) the multiantigenic domain (Mad) and (iii) a TLR2/4 agonist domain (TLRag). Functionality of the resulting chimeric protein is based on the combined effect of the above-mentioned three different domains for simultaneous activation of antigen presenting cells and antigen cross-presentation, leading to an efficacious multiantigenic and multiallelic cellular immune response. Helper and cytotoxic T-cell responses were observed against model-, neo- and self-antigens, and were highly potent in several murine tumor models. The safety and the immunogenicity of a human vaccine candidate designed for colorectal cancer treatment was demonstrated in a non-human primate model. This newly engineered therapeutic vaccine approach is promising for the treatment of poorly infiltrated tumors that do not respond to currently marketed immunotherapies.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell-Penetrating Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptors/agonists , Adjuvants, Immunologic , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Dendritic Cells/immunology , HEK293 Cells , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunity, Cellular , Immunologic Memory/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macaca fascicularis , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Mice , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Toll-Like Receptors/immunology
6.
Mol Ther ; 24(9): 1675-85, 2016 09.
Article in English | MEDLINE | ID: mdl-27377043

ABSTRACT

Cell penetrating peptides (CPPs) from the protein ZEBRA are promising candidates to exploit in therapeutic cancer vaccines, since they can transport antigenic cargos into dendritic cells and induce tumor-specific T cells. Employing CPPs for a given cancer indication will require engineering to include relevant tumor-associated epitopes, administration with an appropriate adjuvant, and testing for antitumor immunity. We assessed the importance of structural characteristics, efficiency of in vitro transduction of target cells, and choice of adjuvant in inducing the two key elements in antitumor immunity, CD4 and CD8 T cells, as well as control of tumor growth in vivo. Structural characteristics associated with CPP function varied according to CPP truncations and cargo epitope composition, and correlated with in vitro transduction efficiency. However, subsequent in vivo capacity to induce CD4 and CD8 T cells was not always predicted by in vitro results. We determined that the critical parameter for in vivo efficacy using aggressive mouse tumor models was the choice of adjuvant. Optimal pairing of a particular ZEBRA-CPP sequence and antigenic cargo together with adjuvant induced potent antitumor immunity. Our results highlight the irreplaceable role of in vivo testing of novel vaccine constructs together with adjuvants to select combinations for further development.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Cell-Penetrating Peptides/immunology , Neoplasms/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Circular Dichroism , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Mice , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Trans-Activators/chemistry , Trans-Activators/immunology , Treatment Outcome , Vaccination
7.
ChemMedChem ; 10(4): 688-714, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25735812

ABSTRACT

Sphingosine-1-phosphate (S1P) receptor agonists have shown promise as therapeutic agents for multiple sclerosis (MS) due to their regulatory roles within the immune, central nervous system, and cardiovascular system. Here, the design and optimization of novel [1,2,4]oxadiazole derivatives as selective S1P receptor agonists are described. The structure-activity relationship exploration was carried out on the three dominant segments of the series: modification of the polar head group (P), replacement of the oxadiazole linker (L) with different five-membered heterocycles, and the use of diverse 2,2'-disubstituted biphenyl moieties as the hydrophobic tail (H). All three segments have a significant impact on potency, S1P receptor subtype selectivity, physicochemical properties, and in vitro absorption, distribution, metabolism, excretion and toxicity (ADMET) profile of the compounds. From these optimization studies, a selective S1P1 agonist, N-methyl-N-(4-{5-[2-methyl-2'-(trifluoromethyl)biphenyl-4-yl]-1,2,4-oxadiazol-3-yl}benzyl)glycine (45), and a dual S1P1,5 agonist, N-methyl-N-(3-{5-[2'-methyl-2-(trifluoromethyl)biphenyl-4-yl]-1,2,4-oxadiazol-3-yl}benzyl)glycine (49), emerged as frontrunners. These compounds distribute predominantly in lymph nodes and brain over plasma and induce long lasting decreases in lymphocyte count after oral administration. When evaluated head-to-head in an experimental autoimmune encephalomyelitis mouse model, together with the marketed drug fingolimod, a pan-S1P receptor agonist, S1P1,5 agonist 49 demonstrated comparable efficacy while S1P1 -selective agonist 45 was less potent. Compound 49 is not a prodrug, and its improved property profile should translate into a safer treatment of relapsing forms of MS.


Subject(s)
Drug Design , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/chemistry , Immunologic Factors/therapeutic use , Oxadiazoles/chemistry , Oxadiazoles/therapeutic use , Receptors, Lysosphingolipid/agonists , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Immunologic Factors/pharmacokinetics , Mice , Mice, Inbred C57BL , Models, Molecular , Multiple Sclerosis/drug therapy , Oxadiazoles/pharmacokinetics , Receptors, Lysosphingolipid/immunology , Structure-Activity Relationship
8.
Methods Mol Biol ; 1013: 67-92, 2013.
Article in English | MEDLINE | ID: mdl-23625494

ABSTRACT

This chapter describes assays that focus on the characterization of compounds identified in high--throughput screening campaigns, and the subsequent medicinal chemistry programs. They cover methods to determine potency in buffer, the effect of whole blood on the compounds' activity and finally the pharmacokinetic (PK)/pharmacodynamic (PD) -relationship of the compounds in a rodent species.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays , Receptors, Chemokine/antagonists & inhibitors , Animals , Automation, Laboratory , Cell Culture Techniques , Cell Migration Assays , Cells, Cultured , Chemokines/metabolism , Chemotaxis/drug effects , Dielectric Spectroscopy , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Ligands , Pharmacokinetics , Protein Binding , Receptors, Chemokine/metabolism , Signal Transduction/drug effects
9.
PLoS One ; 7(7): e40457, 2012.
Article in English | MEDLINE | ID: mdl-22792334

ABSTRACT

Multiple sclerosis (MS) is a neuroinflammatory disease characterized by a progressive loss of myelin and a failure of oligodendrocyte (OL)-mediated remyelination, particularly in the progressive phases of the disease. An improved understanding of the signaling mechanisms that control differentiation of OL precursors may lead to the identification of new therapeutic targets for remyelination in MS. About 100 mammalian Protein Tyrosine Phosphatases (PTPs) are known, many of which are involved in signaling both in health and disease. We have undertaken a systematic genomic approach to evaluate PTP gene activity in multiple sclerosis autopsies and in related in vivo and in vitro models of the disease. This effort led to the identification of Dusp15/VHY, a PTP previously believed to be expressed only in testis, as being transcriptionally regulated during OL differentiation and in MS lesions. Subsequent RNA interference studies revealed that Dusp15/VHY is a key regulator of OL differentiation. Finally, we identified PDGFR-beta and SNX6 as novel and specific Dusp15 substrates, providing an indication as to how this PTP might exert control over OL differentiation.


Subject(s)
Cell Differentiation , Dual-Specificity Phosphatases/genetics , Multiple Sclerosis/enzymology , Oligodendroglia/enzymology , Aged , Animals , Brain/enzymology , Cells, Cultured , Cerebellum/enzymology , Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Female , Gene Knockdown Techniques , Genomics , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/pathology , Myelin Basic Protein/metabolism , Oligodendroglia/physiology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptor, Platelet-Derived Growth Factor beta/chemistry , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Sorting Nexins/chemistry , Sorting Nexins/metabolism , Spinal Cord/enzymology , Substrate Specificity , Transcriptome
10.
J Med Chem ; 49(13): 3857-71, 2006 Jun 29.
Article in English | MEDLINE | ID: mdl-16789742

ABSTRACT

Class I phosphoinositide 3-kinases (PI3Ks), in particular PI3Kgamma, have become attractive drug targets for inflammatory and autoimmune diseases. Here, we disclose a novel series of furan-2-ylmethylene thiazolidinediones as selective, ATP-competitive PI3Kgamma inhibitors. Structure-based design and X-ray crystallography of complexes formed by inhibitors bound to PI3Kgamma identified key pharmacophore features for potency and selectivity. An acidic NH group on the thiazolidinedione moiety and a hydroxy group on the furan-2-yl-phenyl part of the molecule play crucial roles in binding to PI3K and contribute to class IB PI3K selectivity. Compound 26 (AS-252424), a potent and selective small-molecule PI3Kgamma inhibitor emerging from these efforts, was further profiled in three different cellular PI3K assays and shown to be selective for class IB PI3K-mediated cellular effects. Oral administration of 26 in a mouse model of acute peritonitis led to a significant reduction of leukocyte recruitment.


Subject(s)
Furans/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Thiazolidinediones/chemical synthesis , Acute Disease , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cells, Cultured , Chemotaxis/drug effects , Class Ib Phosphatidylinositol 3-Kinase , Crystallography, X-Ray , Furans/chemistry , Furans/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Models, Molecular , Molecular Structure , Monocytes/drug effects , Monocytes/physiology , Neutrophils/immunology , Peritonitis/chemically induced , Peritonitis/drug therapy , Peritonitis/immunology , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship , Thiazolidinediones/chemistry , Thiazolidinediones/pharmacology , Thioglycolates
11.
J Neuroimmunol ; 145(1-2): 1-11, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14644025

ABSTRACT

We investigated the role of the CD134 (also named OX40) molecule in experimental allergic encephalomyelitis (EAE) and multiple sclerosis (MS). We examined the susceptibility of Cd134(-/-) mice to EAE, an autoimmune murine model that is dependent on infiltrating CD4+ T lymphocytes reactive to myelin proteins. EAE induced by myelin oligodendrocyte glycoprotein (MOG) injection in Cd134(-/-) mice showed less severe clinical signs of disease and markedly reduced inflammatory infiltrates within the central nervous system (CNS). Resistance was associated with a strong reduction of pathogenic IFNgamma-producing T cells infiltrating the CNS of Cd134(-/-) mice. Furthermore, analysis of CNS tissue sections from EAE animals and MS patients revealed the presence of CD134+ cells that were localized in active lesions, mainly in perivascular infiltrates. The presence of CD134-expressing T cells in brain tissue of MS patients and EAE affected mice, together with the functional evidence provided by the significant decrease in disease score obtained in Cd134(-/-) mice, indicate that interfering with the CD134 molecule in T cells may be an appropriate target for therapeutic intervention in active MS.


Subject(s)
Brain/immunology , Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Receptors, Tumor Necrosis Factor/physiology , Spinal Cord/immunology , Spinal Cord/metabolism , Up-Regulation/immunology , Amino Acid Sequence , Animals , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Division/genetics , Cell Division/immunology , Cell Movement/genetics , Cell Movement/immunology , Down-Regulation/genetics , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Receptors, OX40 , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Spinal Cord/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Up-Regulation/genetics
12.
J Biomol Screen ; 8(5): 522-32, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14567779

ABSTRACT

To take advantage of the growing knowledge of cellular signaling pathways, modern-day drug discovery faces an increasing challenge to develop assays to screen for compounds that modulate protein-protein interactions. One bottleneck in achieving this goal is a lack of suitable and robust assay technologies amenable to a high-throughput format. In this report, we describe how we utilized Alphascreen trade mark technology to develop a high-throughput assay to monitor ligand binding to a member of the tumor necrosis factor receptor superfamily. We expressed a fusion protein consisting of the extracellular domain of the OX40 receptor with the constant domains of human IgG. In the presence of OX40 ligand, we determined a binding affinity constant consistent with reported values and optimized the protocol to develop a simple, homogeneous, and sensitive binding assay in a 384-well format. Finally, we assessed if this system could identify small peptides capable of inhibiting the OX40 receptor and ligand interaction. The results showed that the assay was able to detect such peptides and could be used to launch a high-throughput screening campaign for small molecules able to prevent OX40 receptor activation.


Subject(s)
Biochemistry/methods , Drug Evaluation, Preclinical/methods , Receptors, Tumor Necrosis Factor/metabolism , Biochemistry/instrumentation , CD8 Antigens/metabolism , Dimethyl Sulfoxide/chemistry , Drug Evaluation, Preclinical/instrumentation , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Membrane Glycoproteins/metabolism , OX40 Ligand , Peptides/metabolism , Peptides/pharmacology , Reagent Kits, Diagnostic , Receptors, OX40 , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...