Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Hematol Oncol ; 14(1): 155, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34579739

ABSTRACT

BACKGROUND: Acute myeloid leukaemia (AML) stem cells (LSCs) cause disease relapse. The CD47 "don't eat me signal" is upregulated on LSCs and contributes to immune evasion by inhibiting phagocytosis through interacting with myeloid-specific signal regulatory protein alpha (SIRPα). Activation of macrophages by blocking CD47 has been successful, but the ubiquitous expression of CD47 on healthy cells poses potential limitations for such therapies. In contrast, CD123 is a well-known LSC-specific surface marker utilized as a therapeutic target. Here, we report the development of SIRPα-αCD123 fusion antibodies that localize the disruption of CD47/SIRPα signalling to AML while specifically enhancing LSC clearance. METHODS: SIRPα-αCD123 antibodies were generated by fusing the extracellular domain of SIRPα to an αCD123 antibody. The binding properties of the antibodies were analysed by flow cytometry and surface plasmon resonance. The functional characteristics of the fusion antibodies were determined by antibody-dependent cellular phagocytosis and antibody-dependent cellular cytotoxicity assays using primary AML patient cells. Finally, an in vivo engraftment assay was utilized to assess LSC targeting. RESULTS: SIRPα-αCD123 fusion antibodies exhibited increased binding and preferential targeting of CD123+ CD47+ AML cells even in the presence of CD47+ healthy cells. Furthermore, SIRPα-αCD123 fusion antibodies confined disruption of the CD47-SIRPα axis locally to AML cells. In vitro experiments demonstrated that SIRPα-αCD123 antibodies greatly enhanced AML cell phagocytosis mediated by allogeneic and autologous macrophages. Moreover, SIRPα-αCD123 fusion antibodies efficiently targeted LSCs with in vivo engraftment potential. CONCLUSIONS: SIRPα-αCD123 antibodies combine local CD47 blockade with specific LSC targeting in a single molecule, minimize the risk of targeting healthy cells and efficiently eliminate AML LSCs. These results validate SIRPα-αCD123 antibodies as promising therapeutic interventions for AML.


Subject(s)
Antigens, Differentiation/immunology , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/immunology , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Receptors, Immunologic/immunology , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Leukemia, Myeloid, Acute/immunology , Male , Middle Aged , Neoplastic Stem Cells/immunology
2.
Front Immunol ; 11: 602802, 2020.
Article in English | MEDLINE | ID: mdl-33281829

ABSTRACT

Conventional dendritic cell (DC) vaccine strategies, in which DCs are loaded with antigens ex vivo, suffer biological issues such as impaired DC migration capacity and laborious GMP production procedures. In a promising alternative, antigens are targeted to DC-associated endocytic receptors in vivo with antibody-antigen conjugates co-administered with toll-like receptor (TLR) agonists as adjuvants. To combine the potential advantages of in vivo targeting of DCs with those of conjugated TLR agonists, we generated a multifunctional antibody construct integrating the DC-specific delivery of viral- or tumor-associated antigens and DC activation by TLR ligation in one molecule. We validated its functionality in vitro and determined if TLR ligation might improve the efficacy of such a molecule. In proof-of-principle studies, an αCD40 antibody containing a CMV pp65-derived peptide as an antigen domain (αCD40CMV) was genetically fused to the TLR5-binding D0/D1 domain of bacterial flagellin (αCD40.FlgCMV). The analysis of surface maturation markers on immature DCs revealed that fusion of flagellin to αCD40CMV highly increased DC maturation (3.4-fold elevation of CD80 expression compared to αCD40CMV alone) by specifically interacting with TLR5. Immature DCs loaded with αCD40.FlgCMV induced significantly higher CMVNLV-specific T cell activation and proliferation compared to αCD40CMV in co-culture experiments with allogeneic and autologous T cells (1.8-fold increase in % IFN-γ/TNF-α+ CD8+ T cells and 3.9-fold increase in % CMVNLV-specific dextramer+ CD8+ T cells). More importantly, we confirmed the beneficial effects of flagellin-dependent DC stimulation using a tumor-specific neoantigen as the antigen domain. Specifically, the acute myeloid leukemia (AML)-specific mutated NPM1 (mNPM1)-derived neoantigen CLAVEEVSL was delivered to DCs in the form of αCD40mNPM1 and αCD40.FlgmNPM1 antibody constructs, making this study the first to investigate mNPM1 in a DC vaccination context. Again, αCD40.FlgmNPM1-loaded DCs more potently activated allogeneic mNPM1CLA-specific T cells compared to αCD40mNPM1. These in vitro results confirmed the functionality of our multifunctional antibody construct and demonstrated that TLR5 ligation improved the efficacy of the molecule. Future mouse studies are required to examine the T cell-activating potential of αCD40.FlgmNPM1 after targeting of dendritic cells in vivo using AML xenograft models.


Subject(s)
Antibodies/pharmacology , CD40 Antigens/immunology , Cancer Vaccines/pharmacology , Dendritic Cells/drug effects , Flagellin/pharmacology , Lymphocyte Activation , Nuclear Proteins/pharmacology , T-Lymphocytes/immunology , Toll-Like Receptor 5/agonists , Viral Matrix Proteins/pharmacology , Antibodies/genetics , Antibodies/immunology , CD40 Antigens/genetics , Cancer Vaccines/immunology , Cell Communication , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes , Filaggrin Proteins , Flagellin/genetics , Flagellin/immunology , HEK293 Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nucleophosmin , Proof of Concept Study , Recombinant Fusion Proteins/pharmacology , Signal Transduction , T-Lymphocytes/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
3.
Sci Rep ; 8(1): 12136, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108253

ABSTRACT

Coordination of DNA replication and cellular redox homeostasis mechanisms is essential for the sustained genome stability due to the sensitivity of replicating DNA to oxidation. However, substantial gaps remain in our knowledge of underlying molecular pathways. In this study, we characterise the interaction of Keap1, a central antioxidant response regulator in Metazoa, with the replicative helicase subunit protein MCM3. Our analysis suggests that structural determinants of the interaction of Keap1 with its critical downstream target - Nrf2 master transactivator of oxidative stress response genes - may have evolved in evolution to mimic the conserved helix-2-insert motif of MCM3. We show that this has led to a competition between MCM3 and Nrf2 proteins for Keap1 binding, and likely recruited MCM3 for the competitive binding dependent modulation of Keap1 controlled Nrf2 activities. We hypothesise that such mechanism could help to adjust the Keap1-Nrf2 antioxidant response pathway according to the proliferative and replicative status of the cell, with possible reciprocal implications also for the regulation of cellular functions of MCM3. Altogether this suggests about important role of Keap1-MCM3 interaction in the cross-talk between replisome and redox homeostasis machineries in metazoan cells.


Subject(s)
DNA Replication , Kelch-Like ECH-Associated Protein 1/metabolism , Minichromosome Maintenance Complex Component 3/metabolism , Oxidative Stress/physiology , Amino Acid Motifs , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , Evolution, Molecular , Humans , Kelch-Like ECH-Associated Protein 1/chemistry , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/isolation & purification , Keratinocytes , Minichromosome Maintenance Complex Component 3/chemistry , Minichromosome Maintenance Complex Component 3/genetics , Minichromosome Maintenance Complex Component 3/isolation & purification , NF-E2-Related Factor 2/metabolism , Primary Cell Culture , Protein Binding/physiology , Protein Conformation, alpha-Helical , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sf9 Cells , Spodoptera , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...