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.
ChemMedChem ; 17(21): e202200350, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-35929380

ABSTRACT

Natural Killer Group 2D (NKG2D) is a homo-dimeric transmembrane protein which is typically expressed on the surface of natural killer (NK) cells, natural killer T (NKT) cells, gamma delta T (γδT) cells, activated CD8 positive T-cells and activated macrophages. Bispecific molecules, capable of bridging NKG2D with a target protein expressed on the surface of tumor cells, may be used to redirect the cytotoxic activity of NK-cells towards antigen-positive malignant T-cells. In this work, we report the discovery of a novel NKG2D small molecule binder [KD =(410±60) nM], isolated from a DNA-Encoded Chemical Library (DEL). The discovery of small organic NKG2D ligands may facilitate the generation of fully synthetic bispecific adaptors, which may serve as an alternative to bispecific antibody products and which may benefit from better tumor targeting properties.


Subject(s)
NK Cell Lectin-Like Receptor Subfamily K , Small Molecule Libraries , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Ligands , Small Molecule Libraries/pharmacology , Small Molecule Libraries/metabolism , Killer Cells, Natural , DNA/metabolism
2.
J Med Chem ; 64(23): 17496-17510, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34821503

ABSTRACT

Interleukin-2 (IL2) is a pro-inflammatory cytokine that plays a crucial role in immunity, which is increasingly being used for therapeutic applications. There is growing interest in developing IL2-based therapeutics which do not interact with the alpha subunit of the IL2 receptor (CD25) as this protein is primarily found on immunosuppressive regulatory T cells (Tregs). Screenings of a new DNA-encoded library, comprising 669,240 members, provided a novel series of IL2 ligands, subsequently optimized by medicinal chemistry. One of these molecules (compound 18) bound to IL2 with a dissociation constant of 0.34 µM was able to form a kinetically stable complex with IL2 in size-exclusion chromatography and recognized the CD25-binding site as evidenced by competition experiments with the NARA1 antibody. Compound 18 and other members of the series may represent the starting point for the discovery of potent small-molecule modulators of IL2 activity, abrogating the binding to CD25.


Subject(s)
DNA/metabolism , Interleukin-2/metabolism , Humans , Ligands
3.
Int J Mol Sci ; 22(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34576184

ABSTRACT

Antibody-cytokine fusion proteins (immunocytokines) are gaining importance for cancer therapy, but those products are often limited by systemic toxicity related to the activity of the cytokine payload in circulation and in secondary lymphoid organs. Tumor necrosis factor (TNF) is used as a pro-inflammatory payload to trigger haemorrhagic necrosis and boost anti-cancer immunity at the tumor site. Here we describe a depotentiated version of TNF (carrying the single point mutation I97A), which displayed reduced binding affinity to its cognate receptor tumor necrosis factor receptor 1 (TNFR-1) and lower biocidal activity. The fusion of the TNF(I97A) mutant to the L19 antibody promoted restoration of anti-tumor activity upon accumulation on the cognate antigen, the alternatively spliced EDB domain of fibronectin. In vivo administration of high doses (375 µg/Kg) of the fusion protein showed a potent anti-tumor effect without apparent toxicity compared with the wild type protein. L19-TNFI97A holds promise for the targeted delivery of TNF activity to neoplastic lesions, helping spare normal tissues.


Subject(s)
Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , Cricetulus , Cytokines/genetics , Cytokines/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Fluorescent Antibody Technique , Immunotherapy , Mice, Inbred BALB C , Mutation , Protein Structure, Secondary , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/genetics
4.
Oncotarget ; 10(62): 6678-6690, 2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31803362

ABSTRACT

INTRODUCTION: Cytokine-based products are gaining importance for cancer immunotherapy. L19-TNF is a clinical-stage antibody-cytokine fusion protein that selectively accumulates to tumors and displays potent anticancer activity in preclinical models. Here, we describe an innovative approach to transiently inhibit off-target toxicity of L19-TNF, while maintaining antitumor activity. METHODS: GSK'963, a potent small molecule inhibitor of RIPK1, was tested in tumor-bearing mice for its ability to reduce acute toxicity associated with TNF signaling. The biological effects of L19-TNF on tumor cells, lymphocytes and tumor vessels were investigated with the aim to enable the administration of TNF doses, which would otherwise be lethal. RESULTS: Transient inhibition of RIPK1 allowed to increase the maximal tolerated dose of L19-TNF. The protective effect of GSK'963 did not affect the selective localization of the immunocytokine to tumors as evidenced by quantitative biodistribution analysis and allowed to reach high local TNF concentrations around tumor blood vessels, causing diffused vascular shutdown and hemorrhagic necrosis within the neoplastic mass. CONCLUSIONS: The selective inhibition of RIPK1 with small molecule inhibitors can be used as a pharmaceutical tool to transiently mask TNF activity and improve the therapeutic window of TNF-based biopharmaceuticals. Similar approaches may be applicable to other pro-inflammatory cytokines.

5.
Biochem Biophys Res Commun ; 446(1): 404-9, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24613850

ABSTRACT

Secreted by tumor and stromal cells, S100 proteins exert their biological functions via the interaction with surface receptors. The most described receptor is the receptor for advanced glycation end-products (RAGE), thereby participating in the S100-dependent cell migration, invasion, tumor growth, angiogenesis and metastasis. Several approaches have been described for determining this interaction. Here we describe an easy, specific and highly reproducible ELISA-based method, by optimizing several parameters such as the binding and blocking buffer, interaction time and concentrations, directed to screen chemical and biological inhibitors of this interaction for S100A4, S100A7 and S100P proteins. The efficiency of the protocol was validated by using well described neutralizing agents of the RAGE receptor and of the S100A4 activity. The methodology described here will allow future works with other members of the S100 protein family and their receptors.


Subject(s)
Receptors, Immunologic/antagonists & inhibitors , S100 Proteins/antagonists & inhibitors , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Communication/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Glycation End Products, Advanced/metabolism , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S100 Calcium Binding Protein A7 , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , S100 Proteins/metabolism
6.
Biochem Pharmacol ; 86(11): 1541-54, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24070653

ABSTRACT

As a new approach for gene therapy, we recently developed a new type of molecule called polypurine reverse Hoogsteen hairpins (PPRHs). We decided to explore the in vitro and in vivo effect of PPRHs in cancer choosing survivin as a target since it is involved in apoptosis, mitosis and angiogenesis, and overexpressed in different tumors. We designed four PPRHs against the survivin gene, one of them directed against the template strand and three against different regions of the coding strand. These PPRHs were tested in PC3 prostate cancer cells in an in vitro screening of cell viability and apoptosis. PPRHs against the promoter sequence were the most effective and caused a decrease in survivin mRNA and protein levels. We confirmed the binding between the selected PPRHs and their target sequences in the survivin gene. In addition we determined that both the template- and the coding-PPRH targeting the survivin promoter were interfering with the binding of transcription factors Sp1 and GATA-3, respectively. Finally, we conducted two in vivo efficacy assays using the Coding-PPRH against the survivin promoter and performing two routes of administration, namely intratumoral and intravenous, in a subcutaneous xenograft tumor model of PC3 prostate cancer cells. The results showed that the chosen Coding-PPRH proved to be effective in decreasing tumor volume, and reduced the levels of survivin protein and the formation of blood vessels. These findings represent the preclinical proof of principle of PPRHs as a new silencing tool for cancer gene therapy.


Subject(s)
Base Pairing , DNA/genetics , Genetic Therapy/methods , Inhibitor of Apoptosis Proteins/genetics , Prostatic Neoplasms/therapy , Purine Nucleotides/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Gene Targeting , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Survivin , Xenograft Model Antitumor Assays
7.
PLoS One ; 8(9): e72480, 2013.
Article in English | MEDLINE | ID: mdl-24023743

ABSTRACT

S100A4, a member of the S100 calcium-binding protein family secreted by tumor and stromal cells, supports tumorigenesis by stimulating angiogenesis. We demonstrated that S100A4 synergizes with vascular endothelial growth factor (VEGF), via the RAGE receptor, in promoting endothelial cell migration by increasing KDR expression and MMP-9 activity. In vivo overexpression of S100A4 led to a significant increase in tumor growth and vascularization in a human melanoma xenograft M21 model. Conversely, when silencing S100A4 by shRNA technology, a dramatic decrease in tumor development of the pancreatic MiaPACA-2 cell line was observed. Based on these results we developed 5C3, a neutralizing monoclonal antibody against S100A4. This antibody abolished endothelial cell migration, tumor growth and angiogenesis in immunodeficient mouse xenograft models of MiaPACA-2 and M21-S100A4 cells. It is concluded that extracellular S100A4 inhibition is an attractive approach for the treatment of human cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Animals , Blotting, Western , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Real-Time Polymerase Chain Reaction , S100 Calcium-Binding Protein A4 , S100 Proteins/antagonists & inhibitors , S100 Proteins/immunology , Surface Plasmon Resonance , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...