Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34599020

ABSTRACT

BACKGROUND: T cell checkpoint immunotherapies have shown promising results in the clinic, but most patients remain non-responsive. CD47-signal regulatory protein alpha (SIRPα) myeloid checkpoint blockade has shown early clinical activity in hematologic malignancies. However, CD47 expression on peripheral blood limits αCD47 antibody selectivity and thus efficacy in solid tumors. METHODS: To improve the antibody selectivity and therapeutic window, we developed a novel affinity-tuned bispecific antibody targeting CD47 and programmed death-ligand 1 (PD-L1) to antagonize both innate and adaptive immune checkpoint pathways. This PD-L1-targeted CD47 bispecific antibody was designed with potent affinity for PD-L1 and moderate affinity for CD47 to achieve preferential binding on tumor and myeloid cells expressing PD-L1 in the tumor microenvironment (TME). RESULTS: The antibody design reduced binding on red blood cells and enhanced selectivity to the TME, improving the therapeutic window compared with αCD47 and its combination with αPD-L1 in syngeneic tumor models. Mechanistically, both myeloid and T cells were activated and contributed to antitumor activity of αCD47/PD-L1 bispecific antibody. Distinct from αCD47 and αPD-L1 monotherapies or combination therapies, single-cell RNA sequencing (scRNA-seq) and gene expression analysis revealed that the bispecific treatment resulted in unique innate activation, including pattern recognition receptor-mediated induction of type I interferon pathways and antigen presentation in dendritic cells and macrophage populations. Furthermore, treatment increased the Tcf7+ stem-like progenitor CD8 T cell population in the TME and promoted its differentiation to an effector-like state. Consistent with mouse data, the compounds were well tolerated and demonstrated robust myeloid and T cell activation in non-human primates (NHPs). Notably, RNA-seq analysis in NHPs provided evidence that the innate activation was mainly contributed by CD47-SIRPα but not PD-L1-PD-1 blockade from the bispecific antibody. CONCLUSION: These findings provide novel mechanistic insights into how myeloid and T cells can be uniquely modulated by the dual innate and adaptive checkpoint antibody and demonstrate its potential in clinical development (NCT04881045) to improve patient outcomes over current PD-(L)1 and CD47-targeted therapies.


Subject(s)
Antibodies, Bispecific/therapeutic use , CD47 Antigen/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Disease Models, Animal , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunity, Innate , Immunotherapy/methods , Macaca fascicularis , Mice , Tumor Microenvironment
2.
J Biol Chem ; 297(4): 101102, 2021 10.
Article in English | MEDLINE | ID: mdl-34419446

ABSTRACT

CD27 is a tumor necrosis factor (TNF) receptor, which stimulates lymphocytes and promotes their differentiation upon activation by TNF ligand CD70. Activation of the CD27 receptor provides a costimulatory signal to promote T cell, B cell, and NK cell activity to facilitate antitumor and anti-infection immunity. Aberrant increased and focused expression of CD70 on many tumor cells renders CD70 an attractive therapeutic target for direct tumor killing. However, despite their use as drug targets to treat cancers, the molecular basis and atomic details of CD27 and CD70 interaction remain elusive. Here we report the crystal structure of human CD27 in complex with human CD70. Analysis of our structure shows that CD70 adopts a classical TNF ligand homotrimeric assembly to engage CD27 receptors in a 3:3 stoichiometry. By combining structural and rational mutagenesis data with reported disease-correlated mutations, we identified the key amino acid residues of CD27 and CD70 that control this interaction. We also report increased potency for plate-bound CD70 constructs compared with solution-phase ligand in a functional activity to stimulate T-cells in vitro. These findings offer new mechanistic insight into this critical costimulatory interaction.


Subject(s)
CD27 Ligand/chemistry , Multiprotein Complexes/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry , CD27 Ligand/genetics , CD27 Ligand/immunology , Crystallography, X-Ray , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Protein Structure, Quaternary , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
3.
MAbs ; 13(1): 1871171, 2021.
Article in English | MEDLINE | ID: mdl-33557687

ABSTRACT

T-cell engaging biologics is a class of novel and promising immune-oncology compounds that leverage the immune system to eradicate cancer. Here, we compared and contrasted a bispecific diabody-Fc format, which displays a relatively short antigen-binding arm distance, with our bispecific IgG platform. By generating diverse panels of antigen-expressing cells where B cell maturation antigen is either tethered to the cell membrane or located to the juxtamembrane region and masked by elongated structural spacer units, we presented a systematic approach to investigate the role of antigen epitope location and molecular formats in immunological synapse formation and cytotoxicity. We demonstrated that diabody-Fc is more potent for antigen epitopes located in the membrane distal region, while bispecific IgG is more efficient for membrane-proximal epitopes. Additionally, we explored other parameters, including receptor density, antigen-binding affinity, and kinetics. Our results show that molecular format and antigen epitope location, which jointly determine the intermembrane distance between target cells and T cells, allow decoupling of cytotoxicity and cytokine release, while antigen-binding affinities appear to be positively correlated with both readouts. Our work offers new insight that could potentially lead to a wider therapeutic window for T-cell engaging biologics in general.


Subject(s)
Antibodies, Bispecific/pharmacology , B-Cell Maturation Antigen/metabolism , Biological Products/pharmacology , Epitopes , Immunoglobulin G/pharmacology , Protein Engineering , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Antibody-Dependent Cell Cytotoxicity , Antigen-Antibody Reactions , B-Cell Maturation Antigen/immunology , Binding Sites, Antibody , Biological Products/immunology , Biological Products/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line, Tumor , Cytokines/metabolism , Epitope Mapping , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunological Synapses/drug effects , Immunological Synapses/immunology , Immunological Synapses/metabolism , Kinetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , fms-Like Tyrosine Kinase 3/immunology , fms-Like Tyrosine Kinase 3/metabolism
4.
Dev Dyn ; 248(12): 1195-1210, 2019 12.
Article in English | MEDLINE | ID: mdl-31444829

ABSTRACT

BACKGROUND: Atrioventricular valve development relies upon the precisely defined dimensions of the atrioventricular canal (AVC). Current models suggest that Wnt signaling plays an important role atop a pathway that promotes AVC development. The factors that confine AVC differentiation to the appropriate location, however, are less well understood. RESULTS: Transmembrane protein 2 (Tmem2) is a key player in restricting AVC differentiation: in zebrafish, tmem2 mutants display an expansion of AVC characteristics, but the molecular mechanism of Tmem2 function in this context remains unclear. Through structure-function analysis, we demonstrate that the extracellular portion of Tmem2 is crucial for its role in restricting AVC boundaries. Importantly, the Tmem2 ectodomain contains regions implicated in the depolymerization of hyaluronic acid (HA). We find that tmem2 mutant hearts exhibit excess HA deposition alongside broadened distribution of Wnt signaling. Moreover, addition of ectopic hyaluronidase can restore the restriction of AVC differentiation in tmem2 mutants. Finally, we show that alteration of a residue important for HA depolymerization impairs the efficacy of Tmem2 function during AVC development. CONCLUSIONS: Taken together, our data support a model in which HA degradation, regulated by Tmem2, limits the distribution of Wnt signaling and thereby confines the differentiation of the AVC.


Subject(s)
Heart Septal Defects/genetics , Heart Septum/embryology , Heart Ventricles/embryology , Hyaluronic Acid/metabolism , Membrane Proteins/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Carbohydrate Metabolism/genetics , Embryo, Nonmammalian , Heart/embryology , Heart Septal Defects/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organogenesis/genetics , Signal Transduction/genetics , Wnt Signaling Pathway/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Development ; 143(16): 2965-72, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27471259

ABSTRACT

Skeletal muscle morphogenesis depends upon interactions between developing muscle fibers and the extracellular matrix (ECM) that anchors fibers to the myotendinous junction (MTJ). The pathways that organize the ECM and regulate its engagement by cell-matrix adhesion complexes (CMACs) are therefore essential for muscle integrity. Here, we demonstrate the impact of transmembrane protein 2 (tmem2) on cell-matrix interactions during muscle morphogenesis in zebrafish. Maternal-zygotic tmem2 mutants (MZtmem2) exhibit muscle fiber detachment, in association with impaired laminin organization and ineffective fibronectin degradation at the MTJ. Similarly, disorganized laminin and fibronectin surround MZtmem2 cardiomyocytes, which could account for their hindered movement during cardiac morphogenesis. In addition to ECM defects, MZtmem2 mutants display hypoglycosylation of α-dystroglycan within the CMAC, which could contribute to the observed fiber detachment. Expression of the Tmem2 ectodomain can rescue aspects of the MZtmem2 phenotype, consistent with a possible extracellular function of Tmem2. Together, our results suggest that Tmem2 regulates cell-matrix interactions by affecting both ECM organization and CMAC activity. These findings evoke possible connections between the functions of Tmem2 and the etiologies of congenital muscular dystrophies, particularly dystroglycanopathies.


Subject(s)
Embryo, Nonmammalian/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Zebrafish Proteins/metabolism , Animals , Cell-Matrix Junctions/metabolism , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Female , Fluorescent Antibody Technique , In Situ Hybridization , Membrane Proteins/genetics , Muscle Development/genetics , Muscle Development/physiology , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...