WO2023172699A1 - Traitement d'une maladie coronarienne - Google Patents

Traitement d'une maladie coronarienne Download PDF

Info

Publication number
WO2023172699A1
WO2023172699A1 PCT/US2023/014924 US2023014924W WO2023172699A1 WO 2023172699 A1 WO2023172699 A1 WO 2023172699A1 US 2023014924 W US2023014924 W US 2023014924W WO 2023172699 A1 WO2023172699 A1 WO 2023172699A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
plaque
agent
areg
Prior art date
Application number
PCT/US2023/014924
Other languages
English (en)
Inventor
Patricia NGUYEN
Charles K.F. Chan
Mark M. Davis
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2023172699A1 publication Critical patent/WO2023172699A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2318/00Antibody mimetics or scaffolds
    • C07K2318/20Antigen-binding scaffold molecules wherein the scaffold is not an immunoglobulin variable region or antibody mimetics

Definitions

  • Coronary artery disease CAD
  • atherosclerotic plaque build-up can cause ischemia, heart attack and even death.
  • CAD Coronary artery disease
  • Recent studies using single cell transcriptomics have shown that the atherosclerotic plaque contains distinct subpopulations of non-immune cells and immune cells in various states of differentiation and activation. Nevertheless, how cells within the plaque interact with each other as well as the tissue microenvironment to contribute to plaque maturity and possible rupture remains elusive. Understanding these dynamics is especially critical considering reports of vascular thrombosis and myocarditis associated with viruses including influenza and SARS-Cov-2, and their associated vaccines, a complication that may be mediated by cross reactivity between viral antigens and self-peptides.
  • compositions and methods are provided for treating coronary artery disease by blocking the activity of immune cell proteins that are undesirably associated with atherosclerotic plaque.
  • the immune cell protein is a cytolytic protein, e.g. granulysin and perforin.
  • the cytolytic protein is human granulysin.
  • an effective dose of an anti-cytolytic protein agent including without limitation, one or both of an anti-granulysin agent and an anti-perforin agent, to an individual in need thereof to reduce the adverse effects of cytolytic proteins in coronary artery disease.
  • pro-inflammatory, cytolytic T cells are associated with atherosclerotic plaque.
  • Tem2 cells referred to herein as Tem2 were identified as expressing high levels of pro- inflammatory cytokines, e.g., CCL3, CCL4, CCL5, and IFNG, and cytolytic markers such as GZMA, GZMH, GZMM, NKG7.
  • This T cell subset also expressed high levels of perforin and granulysin, two pore-forming proteins that mediate cell killing by enabling granzyme entry. Granulsyin itself may also be able to directly cause cellular injury and death.
  • This sub-population was shown to track with plaque progression, increasing as plaques mature from lipid-rich to more complex lesions, including fibroatheroma, then declining as plaques became more stable and calcified post- rupture. This allows targeted intervention to reduce the adverse effects of cytolytic proteins, by blocking activity during vulnerable stages of plaque progression.
  • an effective dose of an anti-cytolytic agent is administered to an individual determined to have a vulnerable plaque, e.g. at the fibroatheroma stage. In some embodiments an effective dose of an anti-cytolytic agent is administered to an individual determined to have a vulnerable plaque at the complex stage, as defined herein. In some embodiments an individual is determined to have a vulnerable plaque by imaging, prior to treatment.
  • An anti-cytolytic protein agent is a molecule that blocks the activity of, for example, granulysin or perforin. In some embodiments the agent specifically binds to granulysin or perforin and thereby reduces cytolysis. In some embodiments the agent is an antibody. In some embodiments the agent comprises a cytolytic protein-specific binding domain selected from a nanobody, a DARPin (designed ankyrin repeat protein), an antibody, an antibody fragment, a heavy chain only antibody, a single-chain variable fragment (scFv), an immunoglobulin single variable domain (ISV, or nanobody), a receptor extracellular domain (ECD) etc. In some embodiments the binding domain is a nanobody.
  • the first and the second binding domains are of the same type, e.g. a first binding domain comprising an ISV and a second signal activator binding domain comprising an ISV.
  • the first and second binding domains are of a different type, e.g. a first binding domain comprising a scFv and a second signal activator binding domain comprising an ISV and the like.
  • the agent further comprises a second binding domain, which is a targeting domain that specifically binds to vascular markers upregulated in atherosclerosis as detailed below.
  • Target binding domains can be specific for smooth muscle markers associated with plaque, particularly cell surface markers including, for example, PDGFR-a and -
  • Other markers of interest included 6 transmembrane epithelial antigen of the prostate 2, phosphoprotein-glycosphingolipid microdomains 1 , prostaglandin E2 receptor, thombospondin 2, and sushi domain-containing protein 2.
  • domains can include connexins expressed on diseased vascular walls (e.g., Cx37, 0x40, and Cx43) and endothelial surface markers that are upregulated during inflammation including ICAM-1 , VCAM-1 , P- selectin and E-selectin.
  • connexins expressed on diseased vascular walls e.g., Cx37, 0x40, and Cx43
  • endothelial surface markers that are upregulated during inflammation including ICAM-1 , VCAM-1 , P- selectin and E-selectin.
  • the first and second binding domains may be contiguous within one domain, or separated by a linker, e.g. a polypeptide linker, or a non-peptidic linker, etc.
  • the length of the linker, and therefore the spacing between the binding domains, can be used to modulate binding.
  • the enforced distance between binding domains can vary, but in certain embodiments may be less than about 100 angstroms, less than about 90 angstroms, less than about 80 angstroms, less than about 70 angstroms, less than about 60 angstroms, or less than about 50 angstroms.
  • the linker is a rigid linker, in other embodiments the linker is a flexible linker.
  • the linker is a peptide linker, it may be from about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30 or more amino acids in length, and is of sufficient length and amino acid composition to enforce the distance between binding domains.
  • the linker comprises or consists of one or more glycine and/or serine residues.
  • an anti-cytolytic agent is provided, optionally in a formulation comprising a pharmaceutically acceptable excipient.
  • the agent may be specific for human granulysin, or human perforin.
  • the agent may further comprise a binding domain specific for a smooth muscle cell marker.
  • the agent may comprise one or two ISV domains.
  • the immune cell protein is amphiregulin (AREG), and an AREG specific agent an agent is administered to modulate AREG activity.
  • the desired effect i.e. upregulation or down-regulation of AREG, can vary with the stage of the plaque.
  • the first stage of plaque formation is referred to the lipid stage, which transitions to the second stage, fibroatheroma.
  • the activity of AREG can be detrimental, for example, by increasing proliferation of vascular smooth muscle cells and endothelial cells and promoting the development of irreversible plaque.
  • Administration of an anti-AREG agent that decreases activity of AREG is therapeutic at this stage.
  • AREG pro-fibrotic protein amphiregulin
  • AREG vascular smooth muscle cells
  • an effective dose of an anti-AREG protein agent is administered to an individual in need thereof to reduce the adverse effects of AREG in the development of coronary artery disease, in a dose effective to stabilize or reduce the proliferation of cells in the plaque.
  • Administration may be performed at an early stage of plaque progression, to reduce proliferation of responsive cells, e.g. VSMC, endothelial cells, etc.
  • An anti-AREG protein agent is a molecule that blocks the biological activity of AREG, e.g. by interfering with the binding of AREG to EGFR.
  • the agent specifically binds to AREG.
  • the agent is an antibody.
  • the agent comprises a AREG protein-specific binding domain selected from an immunoglobulin single variable domain (ISV, or nanobody), a DARPin (designed ankyrin repeat protein), an antibody, an antibody fragment, a heavy chain only antibody, a single-chain variable fragment (scFv),, a receptor extracellular domain (ECD) etc.
  • the binding domain is a nanobody (ISV).
  • the agent further comprises a second binding domain, which is a targeting domain that specifically binds to a VSMC or endothelial cell surface marker.
  • the first and the second binding domains are of the same type, e.g. a first binding domain comprising an ISV and a second signal activator binding domain comprising an ISV.
  • the first and second binding domains are of a different type, e.g. a first binding domain comprising a scFv and a second signal activator binding domain comprising an ISV and the like.
  • the second binding domain specifically binds to Lectin-like oxidized LDL (oxLDL) receptor-1 (LOX-1 ), which mediates the uptake of oxLDL by vascular cells.
  • LOX-1 is involved in endothelial dysfunction, monocyte adhesion, the proliferation, migration, and apoptosis of smooth muscle cells, foam cell formation, platelet activation, as well as plaque instability. These LOX-1 -dependent biological processes contribute to plaque instability and the ultimate clinical sequelae of plaque rupture and life-threatening tissue ischemia.
  • a benefit of LOX-1 binding is that the agent provides both for targeting to affected tissues, and can further inhibit the undesirable activity of LOX-1 .
  • Target binding domains can be specific for any cell surface marker associated with plaque, particularly cell surface markers including, for example, PDGFR-a and -p (platelet- derived growth factor receptor), which are typical receptors expressed on smooth muscle cells, LRP1 (low-density lipoprotein receptor-related protein), interleukin 1 receptor, type II, CPM (carboxypeptidase M), CA12 (carbonic anhydrase 12), and RAMP1 (receptor activity- modifying protein 1).
  • Other markers of interest included 6 transmembrane epithelial antigen of the prostate 2, phosphoprotein-glycosphingolipid microdomains 1 , prostaglandin E2 receptor, thombospondin 2, and sushi domain-containing protein 2.
  • Other domains can include connexins expressed on diseased vascular walls (e.g., Cx37, 0x40, and 0x43) and endothelial surface markers that are upregulated during inflammation including ICAM-1 , VCAM-1 , P- selectin and E-selectin.
  • Target binding domains can also be specific for, for example, serum albumin, which can provide for an extended serum half-life of the agent.
  • the first and second binding domains may be contiguous within one domain, or separated by a linker, e.g. a polypeptide linker, or a non-peptidic linker, etc.
  • the length of the linker, and therefore the spacing between the binding domains, can be used to modulate binding.
  • the enforced distance between binding domains can vary, but in certain embodiments may be less than about 100 angstroms, less than about 90 angstroms, less than about 80 angstroms, less than about 70 angstroms, less than about 60 angstroms, or less than about 50 angstroms.
  • the linker is a rigid linker, in other embodiments the linker is a flexible linker.
  • the linker is a peptide linker, it may be from about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30 or more amino acids in length, and is of sufficient length and amino acid composition to enforce the distance between binding domains.
  • the linker comprises or consists of one or more glycine and/or serine residues.
  • an anti-AREG agent is provided, optionally in a formulation comprising a pharmaceutically acceptable excipient.
  • the agent may comprise one or two ISV domains.
  • the agent may further comprise a binding domain specific for LOX-1.
  • the agent may further comprise a binding domain specific for a smooth muscle cell surface marker.
  • the agent may further comprise a binding domain specific for serum albumin.
  • an effective dose of an AREG agent e.g. AREG protein or an AREG-expressing cell
  • an individual determined to have a vulnerable plaque e.g. at the late fibroatheroma stage or complex stage, in a dose effective to reduce plaque rupture.
  • an effective dose of an anti-AREG agent is administered to an individual determined to have a early, lipid laden plaques, as defined herein, to prevent conversion to irreversible plaques.
  • an individual is determined to have the stage of plaque determined by imaging, prior to treatment.
  • Suitable AREG agents include, without limitation, AREG protein, which may be modified, e.g. by the addition of a targeting domain or other moiety to localize the protein at the site of a plaque.
  • T cells expressing AREG are also suitable agents. T cells for this purpose may be expanded T-myeloid like cell subset containing a high proportion of activated T cells expressing high levels of AREG. Such T cells can be obtained from the individual and expanded ex vivo. Alternatively such T cells can be obtained from an allogeneic source. T cells can be genetically modified during ex vivo expansion, e.g. by introducing genes for upregulation of AREG, introducing a “suicide” switch, introducing a chimeric antigen contract (CAR-T) and other modifications as known in the art.
  • CAR-T chimeric antigen contract
  • FIGS. 1 A-1 E Antigen-experienced T cells predominate in the plaque immune landscape.
  • TCR repertoire was analyzed for clonal diversity, clone size, and complementarity-determining region 3 (CDR3) motif enrichment (using GLIPH), and their association with pathological disease stages.
  • CDR3 motif enrichment using GLIPH
  • TCR sequences from our data were compared to sequences with known specificities from public and private databases. Selected TCR sequences were expressed as transfectants to confirm antigen-specificity.
  • B Cluster analysis.
  • Plaque immune cells left and the T cell subset from the plaque transcriptome (right) from 12 donors were plotted by uniform manifold approximation and projection (UMAP) dimensionality reduction of the scRNA-seq data and colored by cluster assignment.
  • C 2D visualization of selected gene markers defining each immune cluster.
  • D 2D visualization of selected gene markers defining each T cell cluster.
  • E Heatmap for expression (normalized, z-scored) of selected marker genes (rows) between cells belonging to different clusters (columns).
  • FIGS. 2A-2D Clonally expanded T cells are shared across arterial segments and track with disease stage.
  • A Mapping TCR clonality of coronary plaques at various stages of maturity;
  • Each color across all pie charts for a donor represents a unique clonotype, and the area of each pie is proportional to the clone size.
  • individual clonotypes were colored if they appeared in 3 or more locations. Clonotypes that did not meet this criterion but had an overlap in at least one other location were colored black.
  • Clonality of plaque CD4 and CDS T cells based on coronary arterial segment.
  • D Clonality of plaque CD4 and CD8 T cells measured across pathological disease stages. Clonality is calculated as the proportion that the multiplets occupy in the total repertoire per sample. P-values were derived using one-way ANOVA followed by Tukey’s multiple comparisons test. Mean anderror bars representing the 95% confidence intervals are shown for all comparisons.
  • FIGS. 3A-3F Flu, CMV and EBV specificities are enriched in plaque CD8 T cells, especially in fibroatheromas.
  • A Diagram of steps for matching the TCR plaque repertoire to databases of TCRs with known MHC restricted HLA specificities to identify candidate antigen epitopes.
  • B Clonotypes from our data were matched against a compiled database of known antigen specificities, including sequences from public databases (VDJdb, McPAS-TCR, TBAdb, 10X Genomics), and an internally generated Flu database. The matching criteria required CDR3P and HLA identity.
  • the fraction of matching clonotypes in plaque showing specificity to influenza FLU
  • CMV cytomegalovirus
  • EBV Epstein-Barr virus
  • C The prevalence of viral-antigen-specific T cells (identified in B) by donor is shown (18 out of 20 donors showed matches).
  • D The prevalence of viral-antigen-specific T cells (identified in B) across plaque disease phenotypes.
  • E The number of FLU, CMV, and EBV epitope specificities among matching clonotypes in plaque.
  • F The number of FLU, CMV, and EBV epitope specificities among matching clonotypes across plaque disease phenotypes.
  • FIGS. 4A-4F conserved coronavirus epitope specificities and CDR3P motifs are found in coronary plaque with the highest fraction in the fibroatheroma stage.
  • A Matching the TCR plaque repertoire to a publicly available databases of TCRs with knownspecificities to SARS- CoV-2 (Adaptive Biotechnologies resources). Because the database is not restricted by MHC HLA type, the matching criteria required CDR3 identity only.
  • B The fraction of matching clonotypes in plaque showing specificity to coronavirus.
  • C The prevalence of viral-antigenspecific T cells (identified in B) across plaque disease phenotypes.
  • FIGS. 5A-5C Viral epitopes share similar amino acid and nucleotide sequences with self-proteins.
  • A Schematic of the application of BLAST and gene expression commons to identify self-proteins with similar amino acid sequences to viral epitopes.
  • B Heatmap showing the expression of self-proteins with similar sequences to viral epitopes across different cell types.
  • C Expression of selected ubiquitous proteins that share similar sequences with viral epitopes stratified by different cell subtypes.
  • A atria
  • Ao aorta
  • C coronary
  • CMs cardiomyocytes
  • ECs endothelial cells
  • SMCs smooth muscle cells
  • V1 ventricle normal
  • V2 ventricle with CAD
  • O other
  • U umbilical.
  • FIGS. 6A-6B Jurkat stimulation assay confirm plaque T cell cross reactivity between viral and self-epitopes.
  • A Schematic of Jurkat stimulation assay.
  • B Representative FACS dot plots showing the CD69 expression on a Jurkat transfectant with and without stimulation.
  • FIGS. 7A-7D A pro-inflammatory, cytolytic T cell signature characterizes the complex disease stage.
  • A Schematic of the evaluation of plaque T cell transcriptome with scRNAseq.
  • B Pro-inflammatory and cytolytic genes are upregulated in plaque residing T cells in CD8 Tem2 relative to other clusters, identified using the FindMarkers function with the default nonparametric Wilcoxon rank sum test. Selected genes with Iog2 fold- change (log2FC) > 0.5 (light pink), 1.0 (dark pink) and 1 .5 (purple) are annotated.
  • C Frequencies of plaque T cell clusters showing significant alteration in the CD8 Tem2 cluster with plaque progression
  • D Dot plot for differentially expressed genes (columns) across 4 pathological disease stages (rows) in the cells in the CD8 Tem2 cluster that do and do not express the activation marker HLA-DRA (rows). Color represents maximum normalized mean expression of genes in each disease stage, and size indicates the proportion of cells expressing the selected genes.
  • FIGS. 8A-8F The transition from lipid-rich plaques to more mature plaques is marked by increasing proportion of T cells expressing the pro-fibrotic protein AREG.
  • A Schematic of evaluation of AREG expression in coronary plaque using scRNAseq.
  • B, C Comparison of AREG expression in lipid and more advanced plaque plotted by uniform manifold approximation and projection (UMAP) dimensionality reduction (B) and by bar graph stratified by sample (C). The P-values were derived using ANOVA followed by the unpaired t test post the Shapiro-Wilk test for normal distribution.
  • D Chord diagram showing significant ligand receptor relationships between T cells and smooth muscle cells (left) and between T cells and macrophages (right).
  • FIGS. 9A-9C Clinical and histological classification of coronary arteries.
  • A Categorization of clinical disease stages based on coronary artery imaging using cardiac catheterization. Views of the right coronary artery with different degrees of stenosis are shown.
  • B Categorization of coronary arteries by histology. Coronary arteries from the four major branches (left main, left anterior descending artery, left circumflex, and right coronary artery) were dissected and examined for plaques with defining features. Lesions were binned into 4 categories (based on stages defined by the American Heart Association), dissected from the remainder of the arterial specimen and digested
  • FIGS. 10A-10F Coronary plaques contain a large proportion of CD4 and CD8 T cells.
  • A Schematic of flow cytometric analyses of the distribution of T cells in blood and across coronary plaque phenotypes.
  • B Representative dot plots showing the gating strategy for identifying CD4 and CD8 T cells in blood (top panel) and coronary plaque (bottom panel) from an individual donor for sorting.
  • C Percentage of total (CD3+) ap T cells, CD4 and CD8 T cell subsets, and CD4/CD8 ratio in blood and coronary plaques. Data normality was confirmed by the Shapiro-Wilk test. P-values were derived using the unpaired t test.
  • D Percentage of total (CD45+) immune cells and ab T cells in normal arteries and across plaque disease phenotypes.
  • E Percentage of CD4 and CD8 T cells, and CD4/CD8 ratio in normal arteries and across plaque disease phenotypes.
  • F Percentage of total immune cells and ab T cells in 4 major coronary arterial segments. Mean and error bars representing the 95% confidence intervals are shown for all comparisons.
  • FIGS. 11 A-1 1 D The coronary plaque immune landscape contains diverse subsets of ab T cells.
  • A Diagram of scRNAseq analysis of immune subsets in plaque of 12 patients with coronary artery disease.
  • B 2D visualization of showing successful exclusion of doublets from the analysis of the alpha beta T cell subset.
  • C Expression (molecules/1 ,000 unique molecular identifiers (UMIs) of select genes across the T cell landscape in patients with coronary artery disease.
  • UMIs unique molecular identifiers
  • FIGS. 12A-12E Coronary plaque T cells appear to be activated in antigen-dependent manner.
  • A scRNAseq analysis of HLA-DRA expression in the plaque T cell transcriptome.
  • C Proportion of HLA-DRA T cells by sample stratified by cluster.
  • D Violin plot showing gene expression of HLA-DRA in T cells in each cluster at the single cell level.
  • E Gene ontology pathway analysis of the genes preferentially upregulated in activated and non-activated CD4+ and CD8+ plaque T cells defined by HLA-DRA expression using the Enrichr pathway analysis tool. The top 10 pathways in enriched in activated and non-activated cells (Adjusted P ⁇ 0.05) are listed.
  • FIGS. 13A-13E Parallel expansion of clonotypes across coronary arterial segments.
  • A Schematic showing the workflow for single cell TCR determination and repertoire analysis (shown in FIG. 13A are TCRa sequence CAVGNFNKFYK (SEQ ID NO:385) and TCR
  • B, C Pie charts showing clonal expansion of paired CDR3a/b sequences obtained by single cell analysis of CD4 T cells (B) and CD8 T cells (C) in coronary plaques by arterial segment. Each color across all pie charts for a donor represents a unique clonotype, and the area of each pie is proportional to the clone size.
  • FIGS. 14A-14F Flu and EBV CDR3b motifs in plaques are shared across the majority of patients, suggesting common antigen-epitopes activating TCRs.
  • A Clustering of shared CDR3b motifs from the plaque TCR repertoires from donors using the GLIPH2 algorithm. The CDR3b region of the TCR has the most touch points with the antigenic epitope. GLIPH2 analysis was performed by co-clustering CDR3b sequences from our data with known Flu, CMV, and EBV specific sequences.
  • B Publicly available CDR3b sequences with known antigen specificities (Flu, CMV, and EBV) were analyzed for the presence of donor-derived plaque-motifs.
  • C Prevalence of plaque-motifs that include Flu-specificities (SLNN, DSPS, LAVN, RFDG, YSTS, SQEK) across plaque disease phenotypes. P-values were derived using Z statistic followed by the Bonferroni correction.
  • D The percentage of input plaque-derived CDR3b sequences within each donor that could be assigned a GLIPH2 inferred antigenspecificity is shown.
  • E For each individual, the percentage of GLIPH2 inferred specificities that could be assigned to FLU, CMV, and EBV is shown.
  • F Network analysis comparing similarities in GLIPH2 clusters with inferred viral specificities across donors.
  • Nodes correspond to donors, while edges correspond to the Jaccard coefficient, which represents the extent of sharing of antigen-specific T-cell response between two subjects. Edges were calculated as the number of GLIPH2 clusters shared between the donors normalized by the total number of GLIPH2 clusters found between them.
  • FIGS. 15A-15D Jurkat stimulation assay and tetramer staining confirm viral specificities of coronary plaque T cells.
  • A Schematic of Jurkat stimulation assay.
  • B Two TCR a/b pairs that showed identical CDR3b (but not CDR3a) match to known viral antigenspecific clonotypes were expressed as transfectants in Jurkat a-b- cells and tested for reactivity to the putative viral antigen.
  • C Representative FACS dot plots showing the CD69 expression on a Jurkat transfectant with and without stimulation.
  • D Representative FACS dot plots of Flu (M1 peptide) tetramer staining.
  • FIGS. 16A-16E Plaque T cells express a cytokine signature that characterizes disease stages and mediates interaction with other cells within the plaque.
  • A Schematic of analysis of plaque tissue by scRNAseq and targeted scRNAseq.
  • B Pro-inflammatory and cytolytic genes are upregulated in plaque residing T cells in CD8 Tem1 relative to other clusters, identified using the FindMarkers function with the default non-parametric Wilcoxon rank sum test. Selected genes with Iog2 fold-change (log2FC) > 0.5 (light pink), 1 .0 (dark pink) and 1 .5 (purple) are annotated.
  • C Expression of selected cytokine genes by non-clonal and clonal ab T cells derived from coronary plaques. P-values were derived using the Wilcoxon matched- pairs signed rank test. Lower and upper hinges of boxes represent 25th to 75th percentiles, the central line represents the median, and the whiskers extend to 10th and 90th percentiles.
  • D Violin plot showing gene expression of amphiregulin (AREG) in T cells by phenotype at the single cell level.
  • E Dot plot showing representative ligand-receptor and receptor-ligand interactions between T cells and other cells in the plaque microenvironment (e.g., smooth muscle and myeloid cells). The color and size of the dots represent the communication score and its significance, respectively.
  • FIGS. 17A-17C Unique and parallel expansion of clonotypes observed between the plaque and blood T cell repertoire.
  • A Clonal composition and expansion of T cell subsets in blood by targeted scRNAseq.
  • C Clonal expansion scatter plots for CD4 (orange) and CD8 (yellow) ab T cells. Scatter plots of expanded clonotypes (multiplets) are shown for donors with paired blood and plaque samples. Clonotypes that exhibit dual expansion in blood and plaque are colored purple and pink for CD4 and CD8 ab T cells, respectively. All dual expanded clonotypes are numbered and their corresponding CDR3a/b sequences are tabulated. Dually expanded clonotypes with matching CDR3b sequences to known viral antigens are labeled and the corresponding sequence is colored (red) and highlighted in bold. One CDR3b sequence showing cross-specificity for FLU and coronavirus is colored in blue and highlighted in bold.
  • FIGS. 18A-18D TCRs that are dually expanded in blood and plaque may serve as a proxy for disease progression.
  • A Schematic of targeted scRNAseq of blood and plaque TCRs.
  • B Bubble plot showing the relationship between clonality of dual expanded TCRs (identified in Supplemental Fig. 7c) and plaque pathological disease stage. Clonality is defined as the number of times a specific CDR3a/b sequence appears relative to the total number sequenced per sample.
  • C Percent clonality of dual expanded TCRs in lipid-rich versus fibroatheroma/complex plaques in donors who had at least one plaque with early and one plaque with advanced disease.
  • FIGS. 19A-19F TCRs in plaque and blood display differences in epitope specificities.
  • A Clonotypes from each donor were matched against a compiled database of known antigen specificities, including sequences from public databases (VDJdb, McPAS-TCR, TBAdb, 10X Genomics and Adaptive Biotechnologies resources) and an internally generated Flu database. The matching criteria required CDR3b and HLA identity.
  • (B) The fraction of matching clonotypes in blood and plaque showing specificity to influenza (FLU), cytomegalovirus (CMV), Epstein-Barr virus (EBV), and other antigens is shown.
  • C The number of FLU, CMV, and EBV epitope specificities among matching clonotypes in blood and plaque.
  • D Clonotypes from our data were matched against a SARS-CoV2-specific publicly available database (Adaptive Biotechnologies). The fraction of matching clonotypes in blood and plaque is shown.
  • (E) The number of conserved coronavirus epitope specificities among matching clonotypes in blood and plaque.
  • PCA Principal Component Analysis
  • FIGS. 20A-20B Viral antigen specific CD8 T cells are enriched in coronary atherosclerotic plaques.
  • A Comparison of bulk RNAseq of blood TCRs and sRNAseq of plaque TCRs in a subset of patients with samples at both sites;
  • B Scatter plots showing the correlation between TCR sequences in blood (determined by bulkTCR-seq) and plaque (determined by scTCR-seq) in 7 donors. The dotted line represents the line of identity.
  • Individual clonotypes were matched against sequences of known viral specificities. The matching criteria required CDR3b and HLA identity. The matching clonotypes identified are colored and their specificities are shown. The CDR3b sequences of the matching clonotypes and their clone size in plaques are tabulated.
  • FIGS. 21 A-21 D A higher prevalence of viral motifs in plaque compared to blood.
  • C Publicly available CDR3b sequences with known antigen specificities (FLU, CMV, EBV and other) were analyzed for the presence of donor-derived GLIPH2-motifs, which are shared between blood and plaque.
  • FIGS. 22A-22D Distinct ap T cell subsets enrich in the plaque immune landscape.
  • B) Changes in frequencies of specific T cell clusters between blood and plaque (n 3). P-values were derived using the paired Student’s t test. Data normality was confirmed by the Shapiro- Wilk test.
  • Coronary artery disease is a narrowing or blockage of the arteries and vessels that provide oxygen and nutrients to the heart. It is associated with vascular inflammation, which causes atherosclerosis, an accumulation of fatty materials on the inner linings of arteries. The resulting blockage restricts blood flow to the heart. When the blood flow is completely cut off, the result is a heart attack. CAD is the leading cause of death for both men and women in the United States.
  • Atherosclerosis also referred to as arteriosclerosis, atheromatous vascular disease, arterial occlusive disease
  • arteriosclerosis also referred to as arteriosclerosis, atheromatous vascular disease, arterial occlusive disease
  • the plaque consists of accumulated intracellular and extracellular lipids, smooth muscle cells, connective tissue, inflammatory cells, and glycosaminoglycans.
  • Vascular inflammation occurs in combination with lipid accumulation in the vessel wall, and vascular inflammation is a hallmark of the atherosclerosis disease process.
  • Myocardial infarction is an ischemic myocardial necrosis usually resulting from abrupt reduction in coronary blood flow to a segment of myocardium.
  • an acute thrombus often associated with plaque rupture, occludes the artery that supplies the damaged area. Plaque rupture occurs generally in vessels previously partially obstructed by an atherosclerotic plaque enriched in inflammatory cells. Altered platelet function induced by endothelial dysfunction and vascular inflammation in the atherosclerotic plaque presumably contributes to thrombogenesis.
  • Myocardial infarction can be classified into ST-elevation and non-ST elevation Ml (also referred to as unstable angina).
  • myocardial infarction In both forms of myocardial infarction, there is myocardial necrosis. In ST-elevation myocardial infraction there is transmural myocardial injury which leads to ST-elevations on electrocardiogram. In non-ST elevation myocardial infarction, the injury is sub-endocardial and is not associated with ST segment elevation on electrocardiogram. Myocardial infarction (both ST and non-ST elevation) represents an unstable form of atherosclerotic cardiovascular disease. Acute coronary syndrome encompasses all forms of unstable coronary artery disease. Heart failure can occur as a result of myocardial dysfunction caused by myocardial infraction. [0056] Angina refers to chest pain or discomfort resulting from inadequate blood flow to the heart.
  • Angina can be a symptom of atherosclerotic cardiovascular disease.
  • Angina may be classified as stable, which follows a regular chronic pattern of symptoms, unlike the unstable forms of atherosclerotic vascular disease.
  • the pathophysiological basis of stable atherosclerotic cardiovascular disease is also complicated but is biologically distinct from the unstable form.
  • stable angina is not myocardial necrosis.
  • Stages of atherosclerotic plaque may be characterized herein by stages: 1. lipid, 2. fibroatheroma, 3. complex, and 4. calcified. As described below and in the examples, within these stages there can be a progression, e.g. where the fibrous cap in fibroatheroma thins and becomes vulnerable to rupture.
  • stage 1 the fatty streak and pre-existing lesions of adaptive intimal thickening.
  • Fatty streak is a yellow discoloration on the surface of the artery lumen, which is flat or slightly elevated in the intima and contains accumulations of intracellular and extracellular lipid.
  • the fatty streak doesn’t protrude substantially into the artery wall nor impedes blood flow.
  • Inflammation begins when the endothelial cells become activated and secrete adhesion molecules, and the smooth muscle cells secrete chemokines and chemoattractants, which together draw monocytes, lymphocytes, mast cells, and neutrophils into the arterial wall. Intimal smooth muscle cells also secrete into the extracellular matrix proteoglycans, collagen, and elastic fibers.
  • Atherosclerosis is believed to start when the lipid accumulation appears as confluent extracellular lipid pools and extracellular lipid cores with decreased cellularity. Increasing accumulation of extracellular lipid coalesces into pools and causes cell necrosis. This progressively distorts the normal architecture of the intima until it is completely disrupted. These enlarging pools form lipid-rich necrotic cores that dominate the central part of the intima, ultimately occupying 30% to 50% of arterial wall volume.
  • fibrous tissue forms a fibrous cap over the lipid-rich necrotic cores and just under the endothelium at the blood interface. This forms the fibrous plaque lesions that develop to become the dominant lesion.
  • the fibrous cap at a few sites becomes thin and weakened when proteolytic enzyme activity continues unchecked and dissolves the fibrous tissue.
  • This thin cap is susceptible to rupture, which exposes the thrombogenic interior arterial wall and produces a thrombus that extends into the arterial lumen.
  • This lesion usually is labeled a vulnerable plaque because of the risk of rupture and life-threatening thrombosis.
  • the plaque may grow into adjacent media and adventitia and distort them. As the plaque grows, the local segment of the arterial wall may enlarge its caliber, thus compensating for threatened reduction of the lumen by the plaque. This compensation, which is seen as remodeling, stops when the plaque occupies about 40% of the area of the artery. Any further plaque enlargement reduces the arterial lumen and may become hemodynamically significant. New vaso vasorum with thin walls invade the diseased intima from the media. These fragile vessels of endothelium, lacking pericytes for support, may leak, producing hemorrhage within the arterial wall. These intramural hemorrhages provoke increased fibrous tissue.
  • stage 3 ruptures of thin fibrous caps are clinically silent in that they heal by forming fibrous tissue matrices of cells, collagen fibers, and extracellular space but may rupture again with thrombus formation. These cyclic changes of rupture, thrombosis, and healing may recur as many as 4 times at a single site in the arterial wall, resulting in multiple layers of healed tissue.
  • stage 4 calcium deposits in the wall occur throughout all these steps, initially as small aggregates, and later as large nodules. Plaques may rupture into the lumen and expose the nodules, which become sites for thrombosis. Erosion of endothelium, underlain by some of the changes described previously or with no underlying histologic abnormality, may occur, resulting in thrombosis. The increasing mass of some plaques alone may become sufficient to form significant stenosis that may cause lethal ischemia simply through flow restriction.
  • a vulnerable plaque is an atheromatous plaque that is particularly unstable and prone to produce sudden major problems such as a heart attack or stroke.
  • the defining characteristics of a vulnerable plaque include but are not limited to: a thin fibrous cap, large lipid-rich necrotic core, increased plaque inflammation, positive vascular remodeling, increased vasa-vasorum neovascularization, and intra-plaque hemorrhage. These characteristics together with the usual hemodynamic pulsating expansion during systole and elastic recoil contraction during diastole contribute to a high mechanical stress zone on the fibrous cap of the atheroma, making it prone to rupture. Tearing of the cover is called plaque rupture.
  • Measurements of vascular disease include, for example, echocardiography and angiography, which have traditionally been the primary imaging modalities for diagnosing cardiac disease.
  • Computed tomography (CT) and magnetic resonance (MR) imaging are used with increasing frequency because they improve tissue characterization.
  • Intravascular ultrasonography, CT, and MR imaging are frequently used to detect atherosclerotic plaque, wall thickening, and luminal stenosis or enlargement; quantify the extent of the disease; and identify complications such as aneurysm, dissection, and thrombus.
  • PET Positron emission tomography
  • FDG fluorine 18 fluorodeoxyglucose
  • PET/CT computed tomography
  • FDG PET/CT has become a valuable imaging modality for diagnosing various conditions in patients who present with systemic symptoms that are difficult to localize and diagnose with a clinical examination and routine imaging procedures. These methods can be used in both preclinical and clinical studies for the evaluation of inflammation in the arterial wall.
  • Technical progress to extend the CV applications of 18 F-FDG PET/CT include improved image acquisition, measurements, and reconstruction protocols. This has allowed a number of clinical trials to provide results of 18 F- FDG PET/CT in detecting atherosclerotic plaque inflammation, discriminating stable from unstable plaques, predicting CV prognosis, and monitoring response to CV-related therapies.
  • 18 F-FDG PET has been used to assess the impact of statin treatment on arterial wall inflammation in interventional studies.
  • arterial 18 F-FDG uptake is expressed as the Target-to-Background Ratio (TBR), that is a measure of the blood-normalized standardized uptake value (SUV).
  • TBR Target-to-Background Ratio
  • SUV blood-normalized standardized uptake value
  • 18 F-FDG is taken up mostly by macrophages within the atherosclerotic plaques, although other cells (i.e., endothelial cells, vascular smooth muscle cells, neutrophils, lymphocytes) may participate in tracer uptake.
  • TBR as a measure of SUV, has been demonstrated to be a reproducible index for quantification of 18 F-FDG uptake in the inflamed arterial wall.
  • intimal smooth muscle cells in native atherosclerotic plaque derive mainly from the medial arterial layer. During this process, SMCs undergo complex structural and functional changes giving rise to a broad spectrum of phenotypes.
  • intimal SMCs are described as dedifferentiated/synthetic SMCs, a phenotype characterized by reduced expression of contractile proteins. Intimal SMCs are considered to have a beneficial role by contributing to the fibrous cap and thereby stabilizing atherosclerotic plaque.
  • intimal SMCs can lose their properties to such an extent that they become hard to identify, contribute significantly to the foam cell population, and acquire inflammatory-like cell features.
  • SMCs Smooth muscle cells
  • DIT diffuse intimal thickening
  • SMCs are not terminally differentiated and can change their phenotype in response to environmental cues including growth factors/inhibitors, mechanical influences, cell-cell and cell-matrix interactions, extracellular lipids and lipoproteins, and various inflammatory mediators in the injured artery wall.
  • DIT diffuse intimal thickening
  • SMCs exhibit a low proliferative rate and maintain a stable phenotype.
  • phenotypic modulation of intimal SMCs occurs, including a more proliferative state, loss of contractility, increased synthesis of proteoglycans, and reduced expression of SMC markers including a-smooth muscle actin (a-SMA) and smooth muscle myosin heavy chains (SMMHCs).
  • SMC markers including a-smooth muscle actin (a-SMA) and smooth muscle myosin heavy chains (SMMHCs).
  • SMCs express variations in their phenotype including foam cell formation, while SMCs forming the fibrous cap retain SMC markers including a-SMA.
  • SMCs constituting the medial layer of healthy arteries are primarily quiescent and highly differentiated. However, in contrast to skeletal and heart muscle cells, they retain a high degree of dedifferentiation potential and plasticity and can shift from a contractile to a so-called synthetic phenotype. This phenomenon is known as SMC phenotypic modulation or switching, typical of SMCs accumulating in the intima during the formation of atherosclerotic plaques, as well as in restenotic lesions following angioplasty/stent placement in humans. SMCs accumulated in the intima during pre-atherosclerotic DIT also exhibit phenotypic dedifferentiation from medial SMCs.
  • a-SMA corresponding to the gene ACTA2
  • actin isoform typical of vascular SMCs and most general marker of SMC lineage from early stages of development
  • Additional SMC markers lost during dedifferentiation include SMMHC (corresponding to the gene MYH1 1 ) isoforms SM1 and SM2, SM22a (or transgelin corresponding to the gene TAGLN), serum response factor (SRF), calponin (corresponding to the gene CNN1 ), h- caldesmon and meta-vinculin. Proteins known to be up- or downregulated in contractile and synthetic SMCs are summarized below.
  • Cell surface markers are of particular interest, and include, for example, PDGFR-a and -p (platelet-derived growth factor receptor), which are typical receptors expressed on smooth muscle cells, LRP1 (low-density lipoprotein receptor-related protein), interleukin 1 receptor, type II, CPM (carboxypeptidase M), CA12 (carbonic anhydrase 12), and RAMP1 (receptor activity-modifying protein 1 ).
  • PDGFR-a and -p platelet-derived growth factor receptor
  • LRP1 low-density lipoprotein receptor-related protein
  • interleukin 1 receptor type II
  • CPM carboxypeptidase M
  • CA12 carbonic anhydrase 12
  • RAMP1 receptor activity-modifying protein 1
  • Other markers of interest included 6 transmembrane epithelial antigen of the prostate 2, phosphoprotein-glycosphingolipid microdomains 1 , prostaglandin E2 receptor, thombospondin 2, and sushi domain-containing protein 2.
  • Granulysin is a cytolytic and proinflammatory molecule expressed by activated human cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Based on sequence homologies with sphingolipid hydrolase activators of the central nervous system, saposins A-D, it is a member of the saposin-like protein (SAPLIP) family of protein.
  • SAPLIP saposin-like protein
  • Granulysin is found in cytolytic granules in CTL and NK cells along with the poreforming protein, perforin, and granzymes. It is synthesized as a 15-kDa molecule, and portions are then cleaved at the amino and carboxy termini to produce a 9-kDa form.
  • agents of the present disclosure target the 9 kDa form. Equivalent amounts of these two forms of granulysin are found in CTL and NK cells. However, the 9-kDa form is sequestered in cytolytic granules, while the 15-kDa form is constitutively secreted.
  • Recombinant 9 kDa granulysin is dependent on perforin for killing intracellular pathogens.
  • Recombinant 9 kDa granulysin is tumoricidal and broadly antimicrobial, killing gram-positive and gram-negative bacteria, yeast, fungi and parasites.
  • Granulysin appears to scissor through the cell membrane, causing ion fluxes. Cytosolic calcium levels increase as a result of an influx of extracellular calcium and a release of calcium from intracellular stores, while cytosolic potassium levels decrease as a result of activation of a calcium-dependent potassium pump. Addition of agents that block either the increase in intracellular calcium or the decrease in intracellular potassium prevents activation of subsequent cell death pathways, and no tumor lysis occurs. The increase in intracellular calcium, decrease in potassium and the presence of granulysin (if ATP is present) all contribute to mitochondrial damage. Mitochondrial calcium overload disrupts the Krebs cycle and oxidative phosphorylation.
  • cytochrome c cytochrome c
  • AIF apoptosis-inducing factor
  • Perforin is a glycoprotein responsible for pore formation in cell membranes of target cells. Perforin is able to polymerize and form a channel in target cell membrane. Polymerized perforin molecules form channels enabling free, non-selective, passive transport of ions, water, small-molecule substances and enzymes. In consequence, the channels disrupt protective barrier of cell membrane and destroy integrity of the target cell. Natural killer (NK) cells and CD8-positive T-cells are the main source of perforin.
  • Perforin is a 60-70-kDa glycoprotein. It has 555 amino acids.
  • Single perforin molecule consists of four domains, including two (N-terminal and C-terminal) typical for perforin and related to its biological functions. The other two domains, which are located in the center of the molecule, are 20%-homologous to analogous domains in complement molecules (C6, 07, C8 and 09).
  • One of the homologous domains contains a sequence allowing formation of two p-sheets and one a-helix structures. It is a hydrophobic domain able to incorporate into the lipid membrane of the target cell.
  • a perforin molecule contains a cysteine-rich domain homologous to low density lipoprotein (LDL) receptor type B and epithelial growth factor (EGF) precursor.
  • N-terminal domain of perforin contains Ca2+ ions binding site that is related to its biological function.
  • the reference sequence for human perforin may be accessed at Genbank NM_005041 (mRNA), and NP_005032 (protein).
  • the pores formed by perforin in the target cell are 5-20 nm in diameter.
  • a 14-nm channel is formed by approximately 20 perforin molecules; however, it was shown that even 3-4 perforin molecules are able to form an efficient pore.
  • Polymerized perforin molecules form cylindrical, hydrophobic channels enabling free, non-selective, passive transport of ions, water, small-molecule substances and enzymes. In consequence, the channels disrupt protective barrier of cell membrane and destroy integrity of the target cell.
  • anti-cytolytic agenf' refers to an agent that blocks the activity of cytolytic proteins secreted by CD8+ T cells, including specifically granulysin and perforin.
  • suitable agents include small molecules, antibodies including nanobodies and derivatives thereof that bind to cytolytic proteins and block their activity.
  • the efficacy of a suitable anti-cytolytic agent can be assessed by assaying the agent, for example by measuring cytolysis of target cells in the presence and absence of the agent.
  • target cells are incubated in the presence or absence of the candidate agent and in the presence of an effector cell, e.g. an activated CD8+ T cell.
  • An agent for use in the methods of the invention will decrease killing by at least 5% (e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 120%, at least 140%, at least 160%, at least 180%, at least 200%, at least 500%, at least 1000%) compared to cytolysis in the absence of the agent.
  • at least 5% e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 120%, at least 140%, at least 160%, at least 180%, at least 200%, at least 500%, at least 1000.
  • Antibodies specific for human granulysin include, without limitation, polyclonal antibodies (as described by Pena et al. (1997) J. Immunol 158:2680-2688).
  • Commercially available monoclonal antibodies include, without limitation, eBioDH2 (DH2) (Invitrogen);
  • MAB31381 (R&D Systems); Clone RB1 (BD Biosciences); Clone B-L38 (Medix Biochemica); etc.
  • Antibodies specific for human perforin are known and used in the art, for example as disclosed in Hameed et al. (1992) Am J Pathol. 140(5): 1025-1030; clones B-D48 and deltaG9 described by Hersperger et al. (2008) Cytometry A 73(1 1):1050-7; polyclonal antibodies disclosed by Fujinaka et al. (2007) Kidney Int 72(7):823-30; and commercially available antibodies including clone dG9 (LS Bio); eBioOMAK-D (Invitrogen); delta G9 (Invitrogen); 5B10 (invitrogen); MAB801 1 (R&D Systems), etc.
  • Amphiregulin is a transmembrane glycoprotein of 252 amino acids.
  • the refseq for human AREG can be accessed at Genbank NM 001657 (mRNA) and NP 001648 (protein). It is a ligand of the epidermal growth factor receptor (EGFR).
  • AREG is synthesized as a membrane-anchored precursor protein that can engage in juxtacrine signaling on adjacent cells.
  • AREG is secreted and behaves as an autocrine or paracrine factor.
  • AREG gene expression and release is induced by stimuli including inflammatory lipids, cytokines, hormones, growth factors and xenobiotics.
  • stimuli including inflammatory lipids, cytokines, hormones, growth factors and xenobiotics.
  • AREG activates major intracellular signaling cascades governing cell survival, proliferation and motility.
  • a number of cells types have been shown to express AREG, including epithelial cells and group 2 innate lymphoid cells. It is thought that AREG ameliorates injury by binding to epithelial EGFR and stimulating proliferation and repair.
  • anti-AREG agent refers to an agent that blocks the activity of amphiregulin.
  • suitable agents include small molecules, antibodies, including nanobodies and derivatives thereof that bind to AREG and block its activity.
  • the efficacy of a suitable anti-AREG agent can be assessed by assaying the agent, for example by measuring proliferation of target cells, e.g. VSMC, endothelial cells, etc. in the presence and absence of the agent.
  • An agent for use in the methods of the invention will decrease responses by at least 5% (e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 120%, at least 140%, at least 160%, at least 180%, at least 200%, at least 500%, at least 1000%) compared to the response in the absence of the agent.
  • at least 5% e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 120%, at least 140%, at least 160%, at least 180%, at least 200%, at least 500%, at least 1000.
  • Antibodies that bind to, and neutralize, AREG are known in the art, for example see Carvalho et al. (2016) Oncogene 35(4):438-47. Commercially available antibodies include AREG559 (Invitrogen); Amphiregulin Monoclonal Antibody (A3) (Invitrogen); clone 3E4 (LS Bio), etc. [0084] As used herein, "antibody” includes reference to an immunoglobulin molecule immunologically reactive with a particular antigen, and includes both polyclonal and monoclonal antibodies. The term also includes genetically engineered forms such as chimeric antibodies (e.g., humanized murine antibodies) and heteroconjugate antibodies.
  • antibody also includes antigen binding forms of antibodies, including fragments with antigenbinding capability (e.g., Fab', F(ab')2, Fab, Fv and rlgG.
  • fragments with antigenbinding capability e.g., Fab', F(ab')2, Fab, Fv and rlgG.
  • the term also refers to recombinant single chain Fv fragments (scFv).
  • scFv single chain Fv fragments
  • antibody also includes bivalent or bispecific molecules, diabodies, triabodies, and tetrabodies.
  • Selection of antibodies may be based on a variety of criteria, including selectivity, affinity, cytotoxicity, etc.
  • the specified antibodies bind to a particular protein sequences at least two times the background and more typically more than 10 to 100 times background.
  • antibodies of the present invention bind antigens on the surface of target cells in the presence of effector cells (such as natural killer cells or macrophages). Fc receptors on effector cells recognize bound antibodies.
  • An antibody immunologically reactive with a particular antigen can be generated by recombinant methods such as selection of libraries of recombinant antibodies in phage or similar vectors, or by immunizing an animal with the antigen or with DNA encoding the antigen.
  • Methods of preparing polyclonal antibodies are known to the skilled artisan.
  • the antibodies may, alternatively, be monoclonal antibodies.
  • Monoclonal antibodies may be prepared using hybridoma methods. In a hybridoma method, an appropriate host animal is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes may be immunized in vitro. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell.
  • a suitable fusing agent such as polyethylene glycol
  • Human antibodies can be produced using various techniques known in the art, including phage display libraries. Similarly, human antibodies can be made by introducing of human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire.
  • Antibodies also exist as a number of well-characterized fragments produced by digestion with various peptidases.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)' 2 , a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond.
  • the F(ab)' 2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)' 2 dimer into an Fab' monomer.
  • the Fab' monomer is essentially Fab with part of the hinge region.
  • antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology.
  • antibody also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries.
  • a "humanized antibody” is an immunoglobulin molecule which contains minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementary determining region
  • donor antibody such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework (FR) regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • a ISV, or nanobody provided herein is an anti-AREG agent and may be, e.g. conjugated or fused, to a targeting moiety.
  • a targeting moiety can be joined to a nanobody through a linker sequence, e.g. a polypeptide linker sequence. The moiety targets the nanobody to specific organs, tissues, tissue compartments, and cell types of interest.
  • Immunoglobulin sequences such as antibodies and antigen binding fragments derived there from (e.g., immunoglobulin single variable domains or ISVs) are used to specifically target the respective antigens disclosed herein.
  • the generation of immunoglobulin single variable domains such as e.g., VHHS or ISV may involve selection from phage display or yeast display, for example ISV can be selected by utilizing surface display platforms where the cell or phage surface display a synthetic library of ISV, in the presence of tagged antigen.
  • a fluorescent secondary antibody directed to the tagged antigen is added to the solution thereby labeling cells bound to antigen.
  • Cells are then sorted using any cell sorting platform of interest e.g., magnetic-activated cell sorting (MACS) or fluorescence-activated cell sorting (FACS). Sorted clones are amplified, resulting in an enriched library of clones expressing ISV that bind antigen. The enriched library is then re-screened with antigen to further enrich for surface displayed antigen binding ISV. These clones can then be sequenced to identify the sequences of the ISV of interest and further transferred to other heterologous systems for large scale protein production.
  • MCS magnetic-activated cell sorting
  • FACS fluorescence-activated cell sorting
  • similar immunoglobulin single variable domains can be generated and selected by the immunization of an experimental animal such as a llama, construction of phage libraries from immune tissue, and
  • immunoglobulin single variable domain or "ISV” is used as a general term to include but not limited to antigen-binding domains or fragments such as VHH domains or V H or V domains, respectively. VHH domains are of interest for the present disclosure.
  • antigen-binding molecules or antigen-binding protein are used interchangeably and include also the term NANOBODIES®.
  • the immunoglobulin single variable domains can be light chain variable domain sequences [e.g., a V -sequence), or heavy chain variable domain sequences (e.g., a VH-sequence); more specifically, they can be heavy chain variable domain sequences that are derived from a conventional four-chain antibody or heavy chain variable domain sequences that are derived from a heavy chain antibody. Accordingly, the immunoglobulin single variable domains can be single domain antibodies, or immunoglobulin sequences that are suitable for use as single domain antibodies, "dAbs", or immunoglobulin sequences that are suitable for use as dAbs, or NANOBODIESTM, including but not limited to VHH sequences.
  • the invention includes immunoglobulin sequences of different origin, comprising mouse, rat, rabbit, donkey, human and camelid immunoglobulin sequences.
  • the immunoglobulin single variable domain includes fully human, humanized, otherwise sequence optimized or chimeric immunoglobulin sequences.
  • the immunoglobulin single variable domain and structure of an immunoglobulin single variable domain can be considered - without however being limited thereto - to be comprised of four framework regions or "PR's", which are referred to in the art and herein as “Framework region 1" or “FR1”; as “Framework region 2" or “FR2”; as “Framework region 3" or “FR3”; and as “Framework region 4" or “FR4", respectively; which framework regions are interrupted by three complementary determining regions or "CDR's”, which are referred to in the art as “Complementarity Determining Region 1" or “CDR1”; as “Complementarity Determining Region 2" or “CDR2”; and as “Complementarity Determining Region 3" or “CDR3", respectively.
  • CDR's complementary determining regions or “CDR's”
  • Nanobody or Nanobodies are registered trademarks of Ablynx N.V. and thus may also be referred to as
  • An amino acid sequence such as e.g. an immunoglobulin single variable domain or polypeptide according to the invention is said to be a "VHH1 type immunoglobulin single variable domain" or "VHH type 1 sequence", if said VHH1 type immunoglobulin single variable domain or VHH type 1 sequence has 85% identity (using the VHH1 consensus sequence as the query sequence and use the blast algorithm with standard setting, i.e., blosom62 scoring matrix) to the VHH1 consensus sequence and mandatorily has a cysteine in position 50, i.e., C50 (using Kabat numbering). See, for example, VHH domains from Camelids in the article of Riechmann and Muyldermans, J. Immunol. Methods 2000 Jun 23; 240 (1 -2): 185-195.
  • polypeptides of the invention that comprise or essentially consist of (i) a first building block consisting essentially of a first immunoglobulin single variable domain, (ii) a second building block consisting essentially of a second immunoglobulin single variable domain, and optionally (iii) a third building block consisting essentially of a third immunoglobulin single variable domain, linked via a linker.
  • Such immunoglobulin single variable domains may be derived in any suitable manner and from any suitable source, and may for example be naturally occurring VHH sequences (i.e. , from a suitable species of Camelid, e.g., llama) or synthetic or semi-synthetic VHs or VLs (e.g., from human).
  • VHH sequences i.e. , from a suitable species of Camelid, e.g., llama
  • VHs or VLs e.g., from human
  • immunoglobulin single variable domains may include "humanized” or otherwise “sequence optimized” VHHs, “camelized” immunoglobulin sequences (and in particular camelized heavy chain variable domain sequences, i.e., camelized VHs), as well as human VHs, human VLs, camelid VH Hs that have been altered by techniques such as affinity maturation (for example, starting from synthetic, random or naturally occurring immunoglobulin sequences), CDR grafting, veneering, combining fragments derived from different immunoglobulin sequences, PCR assembly using overlapping primers, and similar techniques for engineering immunoglobulin sequences well known to the skilled person; or any suitable combination of any of the foregoing as further described herein.
  • Immunoglobulin single variable domains may comprise an amino acid sequence that corresponds to the amino acid sequence of a naturally occurring VHH domain, but that has been "humanized", i.e. by replacing one or more amino acid residues in the amino acid sequence of said naturally occurring VHH sequence (and in particular in the framework sequences) by one or more of the amino acid residues that occur at the corresponding position(s) in a VH domain from a conventional 4-chain antibody from a human being (e.g. indicated above).
  • This can be performed in a manner known per se, which will be clear to the skilled person, for example on the basis of the further description herein and the prior art on humanization referred to herein.
  • humanized immunoglobulin single variable domains of the invention can be obtained in any suitable manner known per se and thus are not strictly limited to polypeptides that have been obtained using a polypeptide that comprises a naturally occurring VHH domain as a starting material.
  • Another class of immunoglobulin single variable domains of the invention comprises immunoglobulin single variable domains with an amino acid sequence that corresponds to the amino acid sequence of a naturally occurring VH domain, but that has been "camelized", i.e. by replacing one or more amino acid residues in the amino acid sequence of a naturally occurring VH domain from a conventional 4-chain antibody by one or more of the amino acid residues that occur at the corresponding position(s) in a VHH domain of a heavy chain antibody.
  • This can be performed in a manner known per se, which will be clear to the skilled person, for example on the basis of the description herein.
  • V H sequence that is used as a starting material or starting point for generating or designing the camelized immunoglobulin single variable domains is preferably a V H sequence from a mammal, more preferably the V H sequence of a human being, such as a V H 3 sequence.
  • camelized immunoglobulin single variable domains of the invention can be obtained in any suitable manner known per se and thus are not strictly limited to polypeptides that have been obtained using a polypeptide that comprises a naturally occurring V H domain as a starting material.
  • the term "AREG agenf' refers to an agent that provides amphiregulin activity, including AREG protein, and cells that express AREG.
  • the efficacy of a suitable AREG agent can be assessed by assaying the agent, for example by measuring proliferation of target cells, e.g. VSMC, endothelial cells, etc. in the presence and absence of the agent.
  • An agent for use in the methods of the invention will increase responses by at least 5% (e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 120%, at least 140%, at least 160%, at least 180%, at least 200%, at least 500%, at least 1000%) compared to the response in the absence of the agent.
  • at least 5% e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 120%, at least 140%, at least 160%, at least 180%, at least 200%, at least 500%, at least 1000.
  • AREG protein can be modified, e.g. by the addition of a targeting domain or other moiety to localize the protein at the site of a plaque.
  • T cells expressing AREG are also suitable agents. T cells for this purpose may be expanded T-myeloid like cell subset containing a high proportion of activated T cells expressing high levels of AREG. Such T cells can be obtained from the individual and expanded ex vivo. Alternatively such T cells can be obtained from an allogeneic source. T cells can be genetically modified during ex vivo expansion, e.g. by introducing genes for upregulation of AREG, introducing a “suicide” switch, introducing a chimeric antigen contract (CAR-T) and other modifications as known in the art.
  • CAR-T chimeric antigen contract
  • T cells refers to mammalian immune effector cells that may be characterized by expression of CD3 and/or T cell antigen receptor, which cells may be engineered to express AREG, a chimeric antigen receptor, etc.
  • the T cells are expanded T-myeloid like cell subset containing a high proportion of activated T cells expressing high levels of AREG.
  • T-cells useful for expansion and optional engineering include T myeloid-like cells, naive T-cells, activated T cells, CD8+ T cells, CD4+ T cells, central memory T-cells, effector memory T-cells or combination thereof.
  • T cells for engineering as described above are collected from a subject or a donor may be separated from a mixture of cells by techniques that enrich for desired cells or may be engineered and cultured without separation.
  • An appropriate solution may be used for dispersion or suspension.
  • Such solution will generally be a balanced salt solution, e.g.
  • fetal calf serum or other naturally occurring factors in conjunction with an acceptable buffer at low concentration, generally from 5-25 mM.
  • Convenient buffers include HEPES, phosphate buffers, lactate buffers, etc.
  • Techniques for affinity separation may include magnetic separation, using antibody-coated magnetic beads, affinity chromatography, cytotoxic agents joined to a monoclonal antibody or used in conjunction with a monoclonal antibody, e.g., complement and cytotoxins, and "panning" with antibody attached to a solid matrix, e.g., a plate, or other convenient technique.
  • the cells may be selected against dead cells by employing dyes associated with dead cells (e.g., propidium iodide). Any technique may be employed which is not unduly detrimental to the viability of the selected cells.
  • the affinity reagents may be specific receptors or ligands for the cell surface molecules indicated above. In addition to antibody reagents, peptide-MHC antigen and T cell receptor pairs may be used; peptide ligands and receptor; effector and receptor molecules, and the like.
  • the separated cells may be collected in any appropriate medium that maintains the viability of the cells, usually having a cushion of serum at the bottom of the collection tube.
  • Various media are commercially available and may be used according to the nature of the cells, including dMEM, HBSS, dPBS, RPMI, Iscove’s medium, etc., frequently supplemented with fetal calf serum (FCS).
  • FCS fetal calf serum
  • FCS fetal calf serum
  • the collected and optionally enriched cell population may be used immediately for genetic modification, or may be frozen at liquid nitrogen temperatures and stored, being thawed and capable of being reused.
  • the cells will usually be stored in 10% DMSO, 50% FCS, 40% RPMI 1640 medium.
  • Ex vivo T-cell activation may be achieved by procedures well established in the art including cell-based T-cell activation, antibody-based activation or activation using a variety of bead-based activation reagents.
  • Cell-based T-cell activation may be achieved by exposure of the T-cells to antigen presenting cells, such as dendritic cells or artificial antigen presenting cells such as irradiated K562 cells.
  • Antibody based activation of T-cell surface CD3 molecules with soluble anti-CD3 monoclonal antibodies also supports T-cell activation in the presence of IL-2.
  • the T-cells of the invention are expanded by culturing the cells in contact with a surface providing an agent that stimulates a CD3 TOR complex associated signal (e.g., an anti-CD3 antibody) and an agent that stimulates a co-stimulatory molecule on the surface of the T-cells (e.g an anti-CD28 antibody).
  • a CD3 TOR complex associated signal e.g., an anti-CD3 antibody
  • an agent that stimulates a co-stimulatory molecule on the surface of the T-cells e.g an anti-CD28 antibody.
  • Bead-based T-cell activation has gained acceptance in the art for the preparation of CAR-T cells for clinical use. Bead-based activation of T-cells may be achieved using commercially available T-cell activation reagents including but not limited to the Invitrogen® GTS Dynabeads® CD3/28 (Life Technologies, Inc.
  • T-cell culture Conditions appropriate for T-cell culture are well known in the art. Lin, et al. (2009) Cytotherapy 1 1 (7):912-922; Smith, et al. (2015) Clinical & Translational Immunology 4:e31 published online 16 January 2015.
  • the target cells are maintained under conditions necessary to support growth, for example, an appropriate temperature (e.g., 37°C) and atmosphere (e.g., air plus 5% CO 2 ).
  • a T cell is allogeneic with respect to the individual that is treated, e.g. see clinical trials NCT03121625; NCT03016377; NCT02476734; NCT02746952; NCT02808442. See for review Graham et al. (2016) Cells. 7(10) E155.
  • an allogeneic T cell is fully HLA matched. However not all patients have a fully matched donor and a cellular product suitable for all patients independent of HLA type provides an alternative.
  • a universal ‘off the shelf’ T cell product provides advantages in uniformity of harvest and manufacture.
  • Allogeneic T cells used in the practice of the present invention may be genetically modified to reduce graft versus host disease.
  • the engineered cells of the present invention may be TCRap receptor knock-outs achieved by gene editing techniques.
  • TCRap is a heterodimer and both alpha and beta chains need to be present for it to be expressed.
  • a single gene codes for the alpha chain (TRAC), whereas there are 2 genes coding for the beta chain, therefore TRAC loci KO has been deleted for this purpose.
  • a number of different approaches have been used to accomplish this deletion, e.g. CRISPR/Cas9; meganuclease; engineered l-Crel homing endonuclease, etc.
  • the preparation of T-cells useful in the practice of the present invention may include transforming isolated T-cells with an expression vector comprising a nucleic acid sequence encoding AREG, kill switch, etc. as desired; optionally in combination with a nucleic acid sequence encoding a CAR polypeptide.
  • Nucleic acid sequences may be under the control of a single promoter with intervening or downstream control elements that facilitate coexpression of the two sequences from the vector.
  • the T-cell has been modified to surface express a chimeric antigen receptor (a ‘CAR-T’ cell).
  • a chimeric antigen receptor a ‘CAR-T’ cell.
  • the terms “chimeric antigen receptor T-cell” and “CAR-T cell” are used interchangeably to refer to a T-cell that has been recombinantly modified to express a chimeric antigen receptor.
  • chimeric antigen receptor and “CAR” are used interchangeably to refer to a polypeptide comprising multiple functional domains arranged from amino to carboxy terminus in the sequence: (a) an antigen binding domain (ABD), (b) a transmembrane domain (TD); and (c) one or more cytoplasmic signaling domains (CSDs) wherein the foregoing domains may optionally be linked by one or more spacer domains.
  • the CAR may also further comprise a signal peptide sequence which is conventionally removed during post-translational processing and presentation of the CAR on the cell surface.
  • CARs useful in the practice of the present invention are prepared in accordance with principles well known in the art.
  • the ABD is a single chain Fv (ScFv).
  • ScFv is a polypeptide comprised of the variable regions of the immunoglobulin heavy and light chain of an antibody covalently connected by a peptide linker (Bird, et al. (1988) Science 242:423-426; Huston, et al. (1988) PNAS(USA) 85:5879-5883; S-z Hu, et al. (1996) Cancer Research, 56, 3055-3061 .
  • the generation of ScFvs based on monoclonal antibody sequences is well known in the art. See, e.g. The Protein Protocols Handbook, John M. Walker, Ed.
  • Antibodies used in the preparation of scFvs may be optimized to select for those molecules which possess particular desirable characteristics (e.g. enhanced affinity) through techniques well known in the art such as phage display and directed evolution.
  • the ABD comprises an anti-CD19 scFv, an anti-PSA scFv, an anti- HER2 scFv, an anti-CEA scFv, an anti-EGFR scFv, an anti-EGFRvI II scFv, an anti-NY-ESO- 1 scFv, an anti-MAGE scFv, an anti-5T4 scFv, or an anti-Wnt1 scFv.
  • the ABD is a single domain antibody obtained through immunization of a camel or llama with a target cell derived antigen, in particular a tumor antigen. See, e.g.
  • the ABD may be generated wholly synthetically through the generation of peptide libraries and isolating compounds having the desired target cell antigen binding properties in substantial accordance with the teachings or Wigler, et al. United States Patent No. 6303313 B1 issued November 12, 1999; Knappik, et al., United States Patent No 6,696,248 B1 issued February 24, 2004, Binz, et al. (2005) Nature Biotechnology 23:1257-1268, and Bradbury, et al. (2011 ) Nature Biotechnology 29:245-254.
  • the ABD may have affinity for more than one target antigen.
  • an ABD of the present invention may comprise chimeric bispecific binding members, i.e. have capable of providing for specific binding to a first target cell expressed antigen and a second target cell expressed antigen.
  • a linker polypeptide molecule is optionally incorporated into the CAR between the antigen binding domain and the transmembrane domain to facilitate antigen binding.
  • the linker is the hinge region from an immunoglobulin, e.g. the hinge from any one of lgG1 , lgG2a, lgG2b, lgG3, lgG4, particularly the human protein sequences. Alternatives include the CH2CH3 region of immunoglobulin and portions of CD3. In those instances where the ABD is an scFv, an IgG hinge may be employed.
  • the linker comprises the amino acid sequence (G4S) n where n is 1 , 2, 3, 4, 5, etc., and in some embodiments n is 3.
  • CARs useful in the practice of the present invention further comprise a transmembrane domain joining the ABD (or linker, if employed) to the intracellular cytoplasmic domain of the CAR.
  • the transmembrane domain is comprised of any polypeptide sequence which is thermodynamically stable in a eukaryotic cell membrane.
  • the transmembrane spanning domain may be derived from the transmembrane domain of a naturally occurring membrane spanning protein or may be synthetic. In designing synthetic transmembrane domains, amino acids favoring alpha-helical structures are preferred.
  • Transmembrane domains useful in construction of CARs are comprised of approximately 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 22, 23, or 24 amino acids favoring the formation having an alpha-helical secondary structure.
  • Amino acids having a to favor alpha-helical conformations are well known in the art. See, e.g Pace, etal. (1998) Biophysical Journal 75: 422-427.
  • Amino acids that are particularly favored in alpha helical conformations include methionine, alanine, leucine, glutamate, and lysine.
  • the CAR transmembrane domain may be derived from the transmembrane domain from type I membrane spanning proteins, such as CD3 ⁇ , CD4, CD8, CD28, etc.
  • the cytoplasmic domain of the CAR polypeptide comprises one or more intracellular signal domains.
  • the intracellular signal domains comprise the cytoplasmic sequences of the T-cell receptor (TCR) and co-receptors that initiate signal transduction following antigen receptor engagement and functional derivatives and sub-fragments thereof.
  • TCR T-cell receptor
  • a cytoplasmic signaling domain such as those derived from the T cell receptor ⁇ -chain, is employed as part of the CAR in order to produce stimulatory signals for T lymphocyte proliferation and effector function following engagement of the chimeric receptor with the target antigen.
  • cytoplasmic signaling domains include but are not limited to the cytoplasmic domain of CD27, the cytoplasmic domain S of CD28, the cytoplasmic domain of CD137 (also referred to as 4-1 BB and TNFRSF9), the cytoplasmic domain of CD278 (also referred to as IGOS), p110a, p, or 0 catalytic subunit of PI3 kinase, the human chain, cytoplasmic domain of CD134 (also referred to as 0X40 and TNFRSF4), FCER1 y and p chains, MB1 (Iga) chain, B29 (IgP) chain, etc.), CD3 polypeptides (0, A and E), syk family tyrosine kinases (Syk, ZAP 70, etc.), src family tyrosine kinases (Lek, Fyn, Lyn, etc.) and other molecules involved in T-cell transduction, such as CD2, CD5 and CD
  • the CAR may also provide a co-stimulatory domain.
  • co-stimulatory domain refers to a stimulatory domain, typically an endodomain, of a CAR that provides a secondary non-specific activation mechanism through which a primary specific stimulation is propagated.
  • the co-stimulatory domain refers to the portion of the CAR which enhances the proliferation, survival or development of memory cells. Examples of costimulation include antigen nonspecific T cell co-stimulation following antigen specific signaling through the T cell receptor and antigen nonspecific B cell co-stimulation following signaling through the B cell receptor. Co-stimulation, e.g., T cell co-stimulation, and the factors involved have been described in Chen & Flies.
  • the CSD comprises one or more of members of the TNFR superfamily, CD28, CD137 (4-1 BB), CD134 (0X40), Dap10, CD27, CD2, CD5, ICAM- 1 , LFA-1 (CD1 1 a/CD18), Lek, TNFR-I, TNFR-II, Fas, CD30, CD40 or combinations thereof.
  • Dosage and frequency may vary depending on the half-life of the agent in the patient. It will be understood by one of skill in the art that such guidelines will be adjusted for the molecular weight of the active agent, the clearance from the blood, the mode of administration, and other pharmacokinetic parameters.
  • the dosage may also be varied for localized administration, e.g. intranasal, inhalation, etc., or for systemic administration, e.g. i.m., i.p., i.v., oral, and the like.
  • An active agent can be administered by any suitable means, including topical, oral, parenteral, intrapulmonary, and intranasal.
  • Parenteral infusions include intramuscular, intravenous (bolus or slow drip), intraarterial, intraperitoneal, intrathecal or subcutaneous administration.
  • An agent can be administered in any manner which is medically acceptable. This may include injections, by parenteral routes such as intravenous, intravascular, intraarterial, subcutaneous, intramuscular, intraperitoneal, intraventricular, intraepidural, or others as well as oral, nasal, ophthalmic, rectal, or topical. Sustained release administration is also specifically included in the disclosure, by such means as depot injections or erodible implants.
  • an agent can be formulated with an a pharmaceutically acceptable carrier (one or more organic or inorganic ingredients, natural or synthetic, with which a subject agent is combined to facilitate its application).
  • a suitable carrier includes sterile saline although other aqueous and non-aqueous isotonic sterile solutions and sterile suspensions known to be pharmaceutically acceptable are known to those of ordinary skill in the art.
  • An "effective amount” refers to that amount which is capable of ameliorating or delaying progression of the diseased, degenerative or damaged condition. An effective amount can be determined on an individual basis and will be based, in part, on consideration of the symptoms to be treated and results sought. An effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • compositions comprising a pharmaceutically acceptable excipient.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • the diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution.
  • the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like.
  • compounds which are "commercially available” may be obtained from commercial sources including but not limited to Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc.
  • the active agents of the invention and/or the compounds administered therewith are incorporated into a variety of formulations for therapeutic administration.
  • the agents are formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and are formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • administration of the active agents and/or other compounds can be achieved in various ways, usually by oral administration.
  • the active agents and/or other compounds may be systemic after administration or may be localized by virtue of the formulation, or by the use of an implant that acts to retain the active dose at the site of implantation.
  • the active agents and/or other compounds may be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination with other pharmaceutically active compounds.
  • the agents may be combined, as previously described, to provide a cocktail of activities.
  • the following methods and excipients are exemplary and are not to be construed as limiting the invention.
  • the agents can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • Formulations are typically provided in a unit dosage form, where the term "unit dosage form,” refers to physically discrete units suitable as unitary dosages for human subjects, each unit containing a predetermined quantity of active agent in an amount calculated sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects, each unit containing a predetermined quantity of active agent in an amount calculated sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for the unit dosage forms of the present invention depend on the particular complex employed and the effect to be achieved, and the pharmacodynamics associated with each complex in the host.
  • the pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are commercially available.
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are commercially available.
  • Any compound useful in the methods and compositions of the invention can be provided as a pharmaceutically acceptable base addition salt.
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid.
  • Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • compositions can also include large, slowly metabolized macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized SepharoseTM, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes).
  • large, slowly metabolized macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized SepharoseTM, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes).
  • a carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group, and non-covalent associations.
  • Suitable covalent-bond carriers include proteins such as albumins, peptides, and polysaccharides such as aminodextran, each of which have multiple sites for the attachment of moieties.
  • the nature of the carrier can be either soluble or insoluble for purposes of the invention.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine,
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above. Langer, Science 249: 1527, 1990 and Hanes, Advanced Drug Delivery Reviews 28: 97-1 19, 1997.
  • the agents of this invention can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.
  • the pharmaceutical compositions are generally formulated as sterile, substantially isotonic and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration.
  • GMP Good Manufacturing Practice
  • Toxicity of the active agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 (the dose lethal to 50% of the population) or the LD100 (the dose lethal to 100% of the population). The dose ratio between toxic and therapeutic effect is the therapeutic index.
  • the data obtained from these cell culture assays and animal studies can be used in further optimizing and/or defining a therapeutic dosage range and/or a sub-therapeutic dosage range (e.g., for use in humans). The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition.
  • polypeptide As well as to naturally occurring amino acid polymers and non- naturally occurring amino acid polymer.
  • sequence identity refers to the subunit sequence identity between two molecules. When a subunit position in both of the molecules is occupied by the same monomeric subunit (e.g., the same amino acid residue or nucleotide), then the molecules are identical at that position. The similarity between two amino acid or two nucleotide sequences is a direct function of the number of identical positions. In general, the sequences are aligned so that the highest order match is obtained. If necessary, identity can be calculated using published techniques and widely available computer programs, such as the GCS program package (Devereux et al., Nucleic Acids Res. 12:387, 1984), BLASTP, BLASTN, FASTA (Atschul et al., J. Molecular Biol. 215:403, 1990).
  • protein variant or “variant protein” or “variant polypeptide” herein is meant a protein that differs from a wild-type protein by virtue of at least one amino acid modification.
  • the parent polypeptide may be a naturally occurring or wild-type (WT) polypeptide, or may be a modified version of a WT polypeptide.
  • Variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it.
  • the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • parent polypeptide By “parent polypeptide”, “parent protein”, “precursor polypeptide”, or “precursor protein” as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant.
  • a parent polypeptide may be a wild-type (or native) polypeptide, or a variant or engineered version of a wild-type polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, gamma-carboxyglutamate, and O-phosphoserine.
  • amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a- carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Amino acid modifications disclosed herein may include amino acid substitutions, deletions and insertions, particularly amino acid substitutions.
  • Variant proteins may also include conservative modifications and substitutions at other positions of the cytokine and/or receptor (e.g., positions other than those involved in the affinity engineering). Such conservative substitutions include those described by Dayhoff in The Atlas of Protein Sequence and Structure 5 (1978), and by Argos in EMBO J., 8:779-785 (1989).
  • amino acids belonging to one of the following groups represent conservative changes: Group I: Ala, Pro, Gly, Gin, Asn, Ser, Thr; Group II: Cys, Ser, Tyr, Thr; Group III: Vai, lie, Leu, Met, Ala, Phe; Group IV: Lys, Arg, His; Group V: Phe, Tyr, Trp, His; and Group VI : Asp, Glu. Further, amino acid substitutions with a designated amino acid may be replaced with a conservative change.
  • isolated refers to a molecule that is substantially free of its natural environment.
  • an isolated protein is substantially free of cellular material or other proteins from the cell or tissue source from which it is derived.
  • the term refers to preparations where the isolated protein is sufficiently pure to be administered as a therapeutic composition, or at least 70% to 80% (w/w) pure, more preferably, at least 80%-90% (w/w) pure, even more preferably, 90-95% pure; and, most preferably, at least 95%, 96%, 97%, 98%, 99%, or 100% (w/w) pure.
  • a “separated” compound refers to a compound that is removed from at least 90% of at least one component of a sample from which the compound was obtained. Any compound described herein can be provided as an isolated or separated compound.
  • subject is used interchangeably herein to refer to a mammal being assessed for treatment and/or being treated.
  • the mammal is a human.
  • subject encompass, without limitation, individuals having a disease.
  • Subjects may be human, but also include other mammals, particularly those mammals useful as laboratory models for human disease, e.g., mice, rats, etc.
  • sample with reference to a patient encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom and the progeny thereof.
  • the term also encompasses samples that have been manipulated in any way after their procurement, such as by treatment with reagents; washed; or enrichment for certain cell populations, such as diseased cells.
  • the definition also includes samples that have been enriched for particular types of molecules, e.g., nucleic acids, polypeptides, etc.
  • biological sample encompasses a clinical sample, and also includes tissue obtained by surgical resection, tissue obtained by biopsy, cells in culture, cell supernatants, cell lysates, tissue samples, organs, bone marrow, blood, plasma, serum, and the like.
  • a “biological sample” includes a sample obtained from a patient's diseased cell, e.g., a sample comprising polynucleotides and/or polypeptides that is obtained from a patient’s diseased cell (e.g., a cell lysate or other cell extract comprising polynucleotides and/or polypeptides); and a sample comprising diseased cells from a patient.
  • a biological sample comprising a diseased cell from a patient can also include non-diseased cells.
  • diagnosis is used herein to refer to the identification of a molecular or pathological state, disease or condition in a subject, individual, or patient.
  • prognosis is used herein to refer to the prediction of the likelihood of death or disease progression, including recurrence, spread, and drug resistance, in a subject, individual, or patient.
  • prediction is used herein to refer to the act of foretelling or estimating, based on observation, experience, or scientific reasoning, the likelihood of a subject, individual, or patient experiencing a particular event or clinical outcome. In one example, a physician may attempt to predict the likelihood that a patient will survive.
  • treatment refers to administering an agent, or carrying out a procedure, for the purposes of obtaining an effect on or in a subject, individual, or patient.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of effecting a partial or complete cure for a disease and/or symptoms of the disease.
  • Treatment may include treatment of a mammal, particularly in a human, and includes: (a) inhibiting the disease, i.e., arresting its development; and (b) relieving the disease or its symptoms, i.e., causing regression of the disease or its symptoms.
  • Treating may refer to any indicia of success in the treatment or amelioration or prevention of a disease, including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the disease condition more tolerable to the patient; slowing in the rate of degeneration or decline; or making the final point of degeneration less debilitating.
  • the treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of an examination by a physician.
  • treating includes the administration of engineered cells to prevent or delay, to alleviate, or to arrest or inhibit development of the symptoms or conditions associated with disease or other diseases.
  • therapeutic effect refers to the reduction, elimination, or prevention of the disease, symptoms of the disease, or side effects of the disease in the subject.
  • a "therapeutically effective amount” refers to that amount of the therapeutic agent sufficient to treat or manage a disease or disorder.
  • a therapeutically effective amount may refer to the amount of therapeutic agent sufficient to delay or minimize the onset of disease.
  • a therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of a disease.
  • a therapeutically effective amount with respect to a therapeutic agent of the invention means the amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of a disease.
  • the term “dosing regimen” refers to a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time.
  • a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses.
  • a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses.
  • all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen are of different amounts.
  • a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen). [00150] "In combination with”, “combination therapy” and “combination products” refer, in certain embodiments, to the concurrent administration to a patient of the engineered proteins and cells described herein in combination with additional therapies, e.g. surgery, radiation, chemotherapy, and the like. When administered in combination, each component can be administered at the same time or sequentially in any order at different points in time. Thus, each component can be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • Concomitant administration means administration of one or more components, such as engineered proteins and cells, known therapeutic agents, etc. at such time that the combination will have a therapeutic effect. Such concomitant administration may involve concurrent (i.e. at the same time), prior, or subsequent administration of components. A person of ordinary skill in the art would have no difficulty determining the appropriate timing, sequence and dosages of administration.
  • a first prophylactic or therapeutic agent can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second prophylactic or therapeutic agent to a subject with a disorder.
  • the anti-cytolytic agents of the disclosure are effective in treating or reducing adverse effects of plaque progression in cardiovascular disease, and may reduce the rupture of a vulnerable plaque.
  • the agents of the disclosure are useful to treat plaque progression in cardiovascular disease in a human subject by administering the agent of the disclosure in an effective amount to the human subject in need thereof, thereby plaque progression in cardiovascular disease.
  • Any route of administration suitable for achieving the desired effect is contemplated by the disclosure (e.g., intravenous, intramuscular, subcutaneous). Treatment or reduction of plaque progression in cardiovascular disease may result in a decrease in the symptoms associated with the condition, which may be long-term or short-term, or even transient beneficial effect.
  • Efficacy may be monitored, for example, by imaging plaque size, plaque stability, release of plaque components, and the like.
  • methods are provided for treating coronary artery disease by administering an effective dose of an agent that modulates the activity of AREG.
  • the desired effect i.e. up-regulation or down-regulation of AREG, can vary with the stage of the plaque.
  • the first stage of plaque formation is referred to the lipid stage, which transitions to the second stage, fibroatheroma.
  • the activity of AREG can be detrimental, for example, by increasing proliferation of vascular smooth muscle cells and endothelial cells and promoting the development of an irreversible plaque.
  • Administration of an anti-AREG agent that decreases activity of AREG is therapeutic at this stage.
  • increased activity of AREG can stabilize the thinning cap, promote fibrous cap thickening, and reduce rupture.
  • Administration of AREG or an AREG producing cell is therapeutic at this stage.
  • the anti-AREG agents of the disclosure are effective in treating or reducing adverse effects of plaque progression in cardiovascular disease, and may reduce the development of plaque.
  • the agents of the disclosure are useful to treat plaque progression in cardiovascular disease in a human subject by administering the agent of the disclosure in an effective amount to the human subject in need thereof, thereby plaque progression in cardiovascular disease.
  • Any route of administration suitable for achieving the desired effect is contemplated by the disclosure (e.g., intravenous, intramuscular, subcutaneous).
  • Treatment or reduction of plaque progression in cardiovascular disease may result in a decrease in the symptoms associated with the condition, which may be long-term or short-term, or even transient beneficial effect.
  • Efficacy may be monitored, for example, by imaging plaque size, plaque stability, release of plaque components, and the like.
  • an effective dose of an AREG agent e.g. AREG protein or an AREG-expressing cell
  • an individual determined to have a vulnerable plaque e.g. at the late fibroatheroma stage or complex stage, in a dose effective to reduce plaque rupture.
  • the therapeutic effect would be to increase the fibrous cap thickening.
  • an effective dose of an anti-AREG agent is administered to an individual determined to have a vulnerable plaque at the late fibroatheroma or complex stage, as defined herein.
  • an individual is determined to have a vulnerable plaque by imaging, prior to treatment.
  • an anti-cytolytic protein or anti-AREG agent agent can vary with the agent, but will generally range from up to about 50 mg/kg, up to about 40 mg/kg, up to about 30 mg/kg, up to about 20 mg/kg, up to about 10 mg/kg, up to about 5 mg/kg; up to about 1 mg/kg, up to about 0.5 mg/kg; up to about 0.1 mg/kg; up to about 0.05 mg/kg; where the dose may vary with the specific agent, e.g. antibody, and recipient.
  • the agents may be administered one or a plurality of days, and in some embodiments is administered daily, every two days, semi-weekly, weekly, etc. for a period of from about 1 , about 2, about 3, about 4, about 5, about 6, about 7 or more weeks, up to a chronic maintenance level of dosing.
  • Therapeutic entities of the present invention are usually administered on multiple occasions. Intervals between single dosages can be weekly, monthly or yearly. Intervals can also be irregular as indicated by measuring blood levels of the therapeutic entity in the patient. Alternatively, therapeutic entities of the present invention can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the polypeptide in the patient.
  • Human coronary plaque T cells are clonally expanded and display cross-reacting viral/self- antigen specificities
  • coronary artery disease is an incurable, life-threatening disease that is now also characterized by chronic inflammation notable for the build-up of atherosclerotic plaques containing immune cells in various states of activation and differentiation. Understanding how these immune cells contribute to disease progression may lead to the development of novel therapeutic strategies.
  • macrophages we found a high proportion of a
  • TCRs T cell-antigen receptors
  • T cell transcriptome across the different stages of plaque progression
  • scRNA-seq analysis analysis, data processing and clustering. All scRNA-seq analysis were performed using the standard protocols. Briefly, using a computational framework forthe annotation of scRNA-seq by reference to bulk transcriptomes (single R), the clusters containing T-cells were identified by comparing each clusters’ transcriptomic signature to purified immune subsets in the following five cell atlases: 1 ) Blueprint Encode, 2) Database of Immune Cell Expression, 3) Human Primary Cell Atlas, 4) Novershtern, and 5) Monaco Immune Data. Cells that were predicted as T cells by at least three of the five databases were classified as T cells and clusters in which at least 90% of cells were T cells were determined to be T cell clusters.
  • T otal RN A extraction was done according to previously described protocols.
  • GLIPH2 Grouping of Lymphocyte Interactions by Paratope Hotspots
  • TCR a and p chain gene fragments were cloned into lentiviral constructs (nLV Dual Promoter EF-1 a- MCS-PGK-Puro).
  • lentiviral constructs nLV Dual Promoter EF-1 a- MCS-PGK-Puro.
  • TCR expression the TCR a and P chain constructs were transfected into 293X cells separately. The virus was collected after 72 h of transfection and transduced into Jurkat ccp /_ cells, which were selected for highest TCR expressionby FACS sorting.
  • T cell stimulation assays were performed as previously described. All stimulation experiments were done in 96-well flat-bottom plates. 100 l of TCR transduced Jurkat aP /_ cells (10 6 per mL) were co-cultured with 100 pl T2 cells (5x10 5 cells per mL) in the presence of individual peptides (2 jiM). After a 16-hour incubation, cells were collected, washed, and stained with TCRp, human CD3 and CD69 and analyzed on LSR II (BectonDickinson) for activation.
  • LSR II BectonDickinson
  • Study cohort includes donors spanning all clinical stages with a diverse range of plaque phenotypes.
  • Our cohort consisted of 35 living donors with and without a history of heart attack, including patients with no visible disease, non-obstructive disease, and obstructive disease defined by cardiovascular imaging tests (FIG. 1 A).
  • CAD is characterized by distinct disease stages that can be contained within the same artery of the same individual, in some instances, we isolated individual plaques from the three major coronary arteries of individual donors.
  • Our analysis included coronary atherosclerotic plaques with a range of phenotypes as defined in the methods section.
  • Coronary plaque immune landscape revealed a large population of antigen- experienced activated T cells.
  • ⁇ Ne first performed an unbiased survey of the immune landscape in coronary plaque using single cell RNA sequencing (scRNA-seq) on different arterial plaques with a range of disease severities obtained from 12 donors.
  • scRNA-seq single cell RNA sequencing
  • Atherosclerotic plaques contain a high proportion of myeloid cells, which have been shown to engulf lipid to form foamy macrophages, recruit other immune cells into the inflamed area, secrete proteinases that increase plaque instability, and promote smooth muscle survival and proliferation in plaques.
  • T cells have immune memory and become activated through specific interaction with previously encountered antigens to promote ongoing inflammation.
  • Coronary plaque TCR repertoire displays clonal expansion that tracks with disease stage. When T cells engage their cognate antigen-peptide-MHC complex, they become activated and clonally expand.
  • a T cell clone was defined by the presence of identical a[3 pairs, which allowed us to measure clonal expansion and track the presence of specific clones across different arterial locations.
  • CD8 T cells showed an inverted V-shaped clonality. Initially low in fatty streaks, clonality increased with lipid deposition and plaque maturation, then declined significantly in fibrocalcific plaques (FIG. 2D).
  • Coronary plaque CD8 a/3 T cells are specific to Flu, CMV and EBV.
  • VDJdb, McPAS-TCR, TBAdb, 10x Genomics and Adaptive Biotechnologies resources we first compared each clonotype from our data against public TCR databases (VDJdb, McPAS-TCR, TBAdb, 10x Genomics and Adaptive Biotechnologies resources) as well as an internally generated influenza database for identical matches.
  • MHC-class I restricted antigens based on the above findings as summarized here: 1 ) a higher proportion of CD8 T cells express the activation marker HLA-DRA, 2) plaque-derived CD8 T cells show greater clonal expansion than CD4 T cells, and 3) CD8 T cell clonalityassociates with disease progression.
  • Fibroatheromas a progressive lesion, contains the highest proportion of viral specific T cells.
  • Plaque T cell antigenic epitopes share sequence identity with vascular proteins.
  • Our finding that plaque T cells display viral specificities is intriguing, especially considering the reports of plaque rupture and myocarditis in healthy individuals receiving the COVID-19 vaccine and in patients infected with Flu and SARS-CoV-2 who may or may not have known cardiovascular co-morbidities or even symptoms associated with viral infection. Because these patients are not actively infected based on serological testing, they are not producing viral antigens.
  • molecular mimicry has been described as a means by which viruses such as Flu or EBV initiate autoimmune diseases, its role in atherosclerosis remains unclear.
  • Jurkat T cells displaying TCRs identical to plaque T cells are activated by viral and self-epitopes identified by computational analysis.
  • self-epitopes e.g., TSPAN17 and Zip9
  • TCRs exposed to antigen presenting cells displaying the appropriate MHC complex e.g., T2 cells
  • T2 cells the appropriate MHC complex
  • CD69 a protein that is rapidly induced on surface of T cells after their activation.
  • a pro-inflammatory, cytolytic T cell signature predominates in the ruptured phenotype.
  • T cell activation leads to the release of chemokines and cytokines that interact with tissue microenvironment.
  • FIG. 7A To evaluate the function of these activated plaque T cells, we interrogated their transcriptome (FIG. 7A). Of the seven T cell clusters that were described above, two with the highest proportion of activated cells displayed a pro-inflammatory and cytolytic signature (FIG. 7A-B, FIG. 16A-B), a finding that was also observed in the clonal T cells in the TCR repertoire analysis using targeted RNAseq (FIG. 16C).
  • CD8 Tem1 and CD8 Tem2 both express high levels of the pro- inflammatory cytokines (e.g., CCL3, CCL4, CCL5, and IFNG) and cytolytic markers (GZMA, GZMH, GZMM, NKG7).
  • CD8 CTL Teml has higher expression of GZMK, a cytolytic marker recently associated with inflammation.
  • CD8 CTL Tem2 on the other hand, has a higher expression of perforin and granulysin, two pore-forming proteins that mediate cell killing by enabling granzyme entry (FIG. 1 E, FIG. 7A-B, FIG. 16A- B).
  • the transition between lipid and more advanced plaques is characterized by an increasing proportion of T cells expressing the pro-fibrotic protein AREG.
  • the third T cell subset containing a high proportion of activated T cells is a cluster annotated T-myeloid like, which has been described previously in patients with cancer and atherosclerosis. This cluster is distinguished by its high expression of AREG, a cytokine belonging to the family of epidermal growth factors that participates in tissue repair and inflammation regulation (FIG. 1 E). Although most prominent in the T-myeloid cluster, especially in T cells co-expressing HLA-DRA, AREG is seen throughout the plaque transcriptome and increases as plaques mature beyond the lipid rich state (FIG. 8A-C; FIG. 16D).
  • AREG is only secreted after T cell activation.
  • 1 1 .8 ⁇ 4.1 % and 21.6 ⁇ 8.3% of plaque CD4+ and CD8+ AREG were clonal in early disease stages, supporting a possible role for clonal T cells secreting AREG in plaque progression.
  • AREG-expressing T cells have been shown to restore tissue integrity after Flu infection, allergen exposure, muscle trauma, and liver damage via interaction with other cells in the tissue
  • a cell signaling algorithm e.g., CellPhoneDB
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • G-CSF granulocyte colony- stimulating factor
  • GM-CSF GM-CSF
  • hCAVSMCs can release ILS and IL1 A in response to biomechanical stress and in the senescent state, respectively, which in turn can promote chronic inflammation, a key mediator of atherosclerosis progression.
  • plaque T cells with viral specificities are reacting to self-antigens that have similar homologies to viral epitopes, a process known as molecular mimicry, and may cause arthritis, diabetes, multiple sclerosis, and narcolepsy.
  • HLA-A2 restricted Flu-M1 -specific CD8 T cells have demonstrated cross reactivity to other viruses including EBV-BMLF1 and SARS-CoV-2-surface glycoprotein as well as self-proteins.
  • these viral specific T cells found in atherosclerotic plaque may be activated by cytokines and not by viral specific engagement, a process commonly referred to as bystander activation, which is triggered by interferons such as Type I IFN, IFN-15 and IFN- 18.
  • these cytokines are elevated in patients with severe flu and COVID infections, who have also been shown to have a higher incidence of thrombolytic complications.
  • Both TCR-dependent and independent activation of plaque TCRS may act synergistically to mediate athersclerotic plaque rupture.
  • Activated CD8 T cells specific for viral epitopes including influenza, EBV, CMV, and SARS-CoV-2 have also been found in other sites not known to have tropism for these viruses, including the liver and tumors.
  • viral-specific CD8 T cells generated in response to systemic infection outside the liver, are trapped in sinusoids and can trigger T- cell mediated hepatitis in absence of viral antigen.
  • the severity of hepatitis is directly related to the degree of anti-viral CD8 T cell response.
  • the absence of liver resident macrophages attenuates tissue damage, suggesting that crosstalk between macrophages and T cells is required for T cell activation.
  • T cell activation e.g., osteopontin; PTGER477 and ostepontin; CD44 and galectin; CD47; CCL3L3; DPP4
  • oligoclonal expansion rather than a single dominant clone, suggesting multiple TCRs or TCRs with similar binding regions are activated by specific antigen-epitope engagement and/or non-specific cytokine mediated stimulation.
  • TCRs we also identified many TCRs with no known specificities. Previous studies have described the presence of “clonal” T cells with specificity to apolipoprotein B, defined by positive binding to its MHC tetramer, in murine aortic plaque and in the blood of patients with CAD. In a study by Wolf et al, the percentage of “clonal” T cells reactive to apolipoprotein B was 0.7% vs 0.2% in human blood with CAD and without CAD, respectively, which is comparable to the prevalence of viral TCRs found in our plaque apolipoprotein B.
  • T cells secrete their own chemokines and cytokines that interact with the tissue microenvironment and modulate inflammation.
  • T cell plaque transcriptome we find a cluster of CD8 T cells, especially those in the activated state, expressing a trifecta of cytolytic granules including granzymes and pore forming proteins that mediate their entry (e.g., perforin and granulysin).
  • these cytotoxic granules potentially contribute to the growing necrotic core and weaken the fibrous cap, promoting plaque progression and potentially plaque rupture.
  • this T cell signature was most prominent in the complex stage characterized by rupture, thrombosis and erosion and has also been described in coronary blood of patients who are having an acute myocardial infarction.
  • AREG a cytokine that has been associated with tissue repair and the development of fibrosis.
  • AREG-expressing CD4 T cells help restore tissue integrity after damage from Flu infection, allergen exposure, and muscle trauma. Mice deficient in AREG, however, do not resolve inflammation-induced injuries but they have few abnormalities under normal conditions. While the exact mechanisms by which AREG mediates tissue repair remain unclear, previous studies have suggested that AREG+ T cells promote fibrosis, in both antigen-independent and antigen-dependent manner, by increasing smooth muscle proliferation, or by reprograming eosinophils.
  • AREG expression has been described in innate lymphoid cells isolated from murine aortic plaques, neither CD4+ nor CD8+ AREG-expressing T cells have been implicated previously in atherosclerosis until now.
  • Our data suggests that AREG-expressing T cells interact with smooth muscle cells within the plaque via binding to its receptors (e.g., ICAM1 and EGFR) and mediating coronary smooth muscle proliferation in vitro through activation of pathways that promote inflammation, proliferation, and fibrosis.
  • its receptors e.g., ICAM1 and EGFR
  • T cell subsets that express other cytokines, such as interferon gamma and TNF alpha that have been shown previously as important mediators atherosclerotic plaque development we also find these subsets within our dataset but do not highlight them here, given their role has been described previously.
  • Diseased segments were further categorized as: defined by histology and categorized based on criteria established by the American Heart Association as follows: 1 ) lipid-rich lesions that include fatty streaks and lipid pools, 2) fibroatheromas that represent discrete lesions that have a fibrous cap covering a lipid, necrotic core with or without speckled microcalcifications, 3) complex plaques that not only include those with evidence of necrosis, rupture, and/or thrombosis, but may also contain micro/macro-calcifications, and 4) fibrocalcific plaques that appear to be stable, healed lesions with dense calcifications (FIG. 9A). Individual plaque lesions bearing the defined histological categories were isolated from coronary arteries and enzymatically digested into single- cell suspensions.
  • Plaque tissue from each donor was used for different experimental assays. For single cell isolation, fat tissue was carefully removed from the exterior of the coronary arteries. Fresh coronary segments containing the defined plaque stage were chopped in digestion buffer containing 125 U/mL Collagenase XI, 450 U/mL Collagenase I, 60 U/mL Hyaluronidase, and 60 U/mL DNase I in Hanks’ Balanced Salt Solution (unless otherwise stated) and incubated in a 37°C shaker for 1 hour. Digested tissue was neutralized with FACS buffer (2% FBS, 1 % antibiotics, 1% Pluronic acid in PBS), then filtered through a 100-micron cell filter.
  • FACS buffer 2% FBS, 1 % antibiotics, 1% Pluronic acid in PBS
  • PBMCs and plaque-derived cells were blocked with Human TruStain FcX (Biolegend) for 20 minutes on ice. Cells were then stained with antibodies for 30 minutes protected from light on ice. Cells were washed with FACS buffer before being analyzed on the LSRII or sorted using the BD Aria machines. Dead cells were defined as aqua-positive cells and were excluded from the analysis. All antibodies were validated by the manufacturers for flow cytometry application, as indicated on the manufacturer’s website. Data were analyzed using FlowJo version 10.2.
  • scRNA-seq analysis of peripheral blood and plaque cells The scRNA-seq libraries were prepared using the 10x Chromium Single Cell 3’ v2 and v3 Reagent Kits, according to the manufacturer’s instructions. In brief, FACS-sorted CD45 + cells were washed once with PBS containing 0.04% bovine serum albumin (BSA) and resuspended in PBS containing 0.04% BSA to a final concentration of 300 - 1000 cells per pl as determined by the hemocytometer. After cell capture in droplets, followed by reverse transcription and cell barcoding in droplets, cDNA was purified, and the yield determined for each sample using the Agilent Bioanalyzer. Amplified cDNA was then used for library construction using the Chromium i7 Sample Index kit. Samples were multiplexed and sequenced on Illumina HiSeq-4000 machines.
  • scRNA-seq data processing All samples were filtered to remove low-quality cells with very few genes (minimum of 300 genes) and weakly expressed genes (less than 5 cells). Because samples performed with 10X v3 chemistry tended to have higher read counts and lower dropout rates than samples performed with 10X v2 chemistry, separate QC cutoffs were applied to account for differences in technical sensitivities in samples. All v2 samples were filtered using a minimum gene count of 300 and a maximum RNA count of 15000. For v3 samples, the minimum gene count cutoff was 500, and the maximum RNA count cutoff was 20000.
  • v2 and v3 samples were filtered using a maximum mitochondrial proportion of 38%, which was the extreme outlier value (upper quartile + 3 x IQR) of mitochondrial transcriptome proportions for the entire cell population.
  • the minimum gene count aims to remove low-quality cells or empty droplets.
  • the maximum RNA count excludes potential multiplets.
  • the mitochondrial transcriptome cutoff removes dying cells, which tend to have weak biological signals because a large proportion of reads are used by the mitochondrial transcriptome.
  • T cell populations were sub-grouped from each sample, they were merged together using the Seurat v3 integration workflow paired with the SCTransform normalization method to remove batch effects.
  • the function ‘FindlntegrationAnchors’ was used to identify anchors between the different T cell subsets, which were then combined with the function ‘Integrate Data’.
  • PCA was applied to the entire T cell population, selecting the 30 most variable components in order to capture more differences contributed by the different samples and a higher number of cells. Similar to how single samples were processed, UMAP was used for visualization, cells were constructed into a KNN graph, then partitioned into meta clusters by the Leiden algorithm.
  • TCR T cell receptor
  • TRDC TCR delta constant
  • TRGC1 and TRGC2 TCR gamma constants
  • T cells from blood as well as those derived from plaques displaying various disease pathological stages were compared for differences in their gene expression patterns. All differential analysis was performed using the FindMarkers function running the default non-parametric Wilcoxon rank sum test. Differentially expressed genes were further processed for functional analysis using the Enrichr pathway analysis tool. Gene expression values were overlaid on pathway diagrams from the Gene database. CellPhoneDb analysis tool was used to analyze cell to cell interactions between T cells and cells known to be pathogenic in atherosclerosis (e.g., smooth cells and macrophages).
  • atherosclerosis e.g., smooth cells and macrophages
  • read counts were transformed into a relative scale by applying Z scoring, then converted to a binary scale by assigning any Z score value greater than 0 as 1 , and anything below as 0.
  • a value of 1 indicates that a cell had higher than average expression for a given cytokine.
  • cells were grouped together by their corresponding donor IDs and clonality to calculate the percentage of cells that show positive expression for a given marker gene.
  • RNA from PBMCs was purified using All Prep DNA/RNA kit (Qiagen). Using 30% of total RNA as input, cDNA was synthesized using Superscript III (Life Technologies) with a TCRp constant regionspecific primer containing a 12N randomized molecular identifier (MID). Free primers were removed using Exonuclease I digestion (New England Biolabs). Two successive PGR steps were performed to amplify the cDNA and ligate sequencing adaptors, where the temperature for the transition between annealing and elongation was slowly ramped up at 0.1 °C/s.
  • PGR products were purified by 2% E-gel EX (Invitrogen, Cat# G401002) and quantified by qPCR Library Quantification Kit (KAPA Biosystems, catalog number KK4824). Samples were multiplexed, pooled and sequenced on the Illumina HiSeq-4000 machines. Analysis for clonality was performed using MIDCI RS as previously described.
  • GLIPH2 Grouping of Lymphocyte Interactions by Paratope Hotspots (GLIPH2) analysis GLIPH2 clusters. TCRs based on two similarity indexes: (1 ) global similarity, that is, the CDR3 sequences differ by up to one amino acid; and (2) local similarity, that is, two TCRs contain a common CDR3 motif of four or more amino acids (enriched over random sub- sampling of unselected repertoires).
  • the first cluster labeled “Naive”, was primarily composed of conventional CD4 and CD8 T cells with high expression of SELL, CCR7 and LEF1 .
  • the second cluster annotated as CD4 memory-like T regulatory cells (e.g., “Tern reg”) given its higher expression of IL7R, but lower, more variable expression of SELL and CCR7 as well as expression of CCR6.
  • T regulatory genes IL2RA, CTLA4, FOXP3, and TGFB1 e.g., IL2RA, CTLA4, FOXP3, and TGFB1 .
  • the cluster annotated as “NK-like T cells”, was distinct in its expression of natural killer cell triggering receptor (NKTR) but no other natural killer (NK) markers. It also had little to no expression of cytokines or cytolytic molecules.
  • CD8 T effector memory clusters CD8 CTL Tem1 and CD8CTL Tem2 clusters that displayed cytolytic features characterized by high expression of NKG7 (e.g., a regulator of exocytosis of cytotoxic granules) and granzymes (e.g., GZMA, GZMM, GZMH, and/or GZMB) (FIG. 1 B-E, FIG. 11 C).
  • NKG7 e.g., a regulator of exocytosis of cytotoxic granules
  • granzymes e.g., GZMA, GZMM, GZMH, and/or GZMB
  • FIG. 11 C CD56, CD57, or killer Ig-like receptors (KIRs)
  • KIRs killer Ig-like receptors
  • FCGR3A natural killerlike receptors
  • CD8 T cells express high levels of the chemokines CCL4 and CCL5, which recruit natural killer cells, myeloid cells, and activated T cells to inflammatory sites.
  • CCL4 and CCL5 chemokines that recruit natural killer cells, myeloid cells, and activated T cells to inflammatory sites.
  • highly cytotoxic CD8 subsets similar to these have been associated with specificities for hepatitis C, HIV, EBV, CMV, and influenza (Flu).
  • CD8 clusters lacked IL7R, CD28, and CD27 markers, suggesting they are terminally differentiated.
  • CD8 clusters differ in their expression of granzyme K, a recently identified marker of “inflammaging”, and granulysin, a protein causing osmotic lysis and has been traditionally associated with microbial infections.
  • the CD4 Tern cluster is a unique CD4 subset with high expression of KLRB1 and IL7R.
  • the last cluster labeled “T-myeloid like”, expresses myeloid specific markers including LYZ, S100A8 and S100A9 as well as T cell specific markers including CD3, GZMA, and CCL5. While it can be argued that these could still be doublet cells, despitecomputational verification that these are, in fact, singlets (FIG. 11 B), this unique cell subset has been described previously in patients with cancer, and even in those with atherosclerosis.
  • GLIPH2 an improved version of GLIPH
  • plaque-motifs plaque-motifs
  • blood motifs blood motifs
  • Plaque and blood T cell transcriptome differ in their activation and differentiation states.
  • Musher DM Abers MS, Corrales-Medina VF. Acute infection and myocardial infarction. New England Journal of Medicine. 2019;380(2) :171-176.
  • Grifoni A Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS- CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. Jun 25 2020;181 (7) :1489-1501 ,e15. doi : 10.1016/j.cell .2020.05.015
  • Gardner SE Humphry M, Bennett MR, Clarke MC. Senescent vascular smooth muscle cells drive inflammation through an interleukin-1 a-dependent senescence- associated secretory phenotype. Arterioscler Thromb Vase Biol. Sep 2015;35(9):1963- 74.
  • Leistner DM Krankel N, Meteva D, et al. Differential immunological signature at the culprit site distinguishes acute coronary syndrome with intact from acute coronary syndrome with ruptured fibrous cap: results from the prospective translational OPTICO- ACS study. European Heart Journal. 2020;41 (37):3549-3560.
  • Luetteke NC Qiu TH, Fenton SE, et al. Targeted inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor ligands in mouse mammary gland development. Development. Jun 1999;126(12):2739-50.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Cardiology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Une maladie coronarienne est traitée par modulation de l'activité de l'amphiréguline, ou par inhibition d'une protéine cytolytique des lymphocytes T, lesdites protéines cytolytiques comprenant, par exemple, la granulysine et la perforine. Des lymphocytes T cytolytiques, pro-inflammatoires, sont associés à la plaque d'athérosclérose. Il s'avère qu'un sous-ensemble des lymphocytes T qui exprime des niveaux élevés de perforine et de granulysine suit la progression de la plaque, augmentant à mesure que les plaques mûrissent de lésions riches en lipides en lésions plus complexes, y compris en fibroathérome, puis déclinant lorsque les plaques deviennent plus stables et calcifiées après rupture. Ceci permet une intervention ciblée pour réduire les effets indésirables de protéines cytolytiques, par blocage de l'activité pendant des stades vulnérables de progression de plaque.
PCT/US2023/014924 2022-03-10 2023-03-09 Traitement d'une maladie coronarienne WO2023172699A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263318590P 2022-03-10 2022-03-10
US202263318606P 2022-03-10 2022-03-10
US63/318,606 2022-03-10
US63/318,590 2022-03-10

Publications (1)

Publication Number Publication Date
WO2023172699A1 true WO2023172699A1 (fr) 2023-09-14

Family

ID=87935755

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/014924 WO2023172699A1 (fr) 2022-03-10 2023-03-09 Traitement d'une maladie coronarienne

Country Status (1)

Country Link
WO (1) WO2023172699A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012018260A1 (fr) * 2010-08-05 2012-02-09 Universiteit Utrecht Holding B.V. Thérapie immunitaire ciblant le récepteur du facteur de croissance épidermique
US11078284B2 (en) * 2014-10-01 2021-08-03 Medimmune Limited Antibodies specific for LOX1 and use in treatment of cardiovascular disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012018260A1 (fr) * 2010-08-05 2012-02-09 Universiteit Utrecht Holding B.V. Thérapie immunitaire ciblant le récepteur du facteur de croissance épidermique
US11078284B2 (en) * 2014-10-01 2021-08-03 Medimmune Limited Antibodies specific for LOX1 and use in treatment of cardiovascular disorders

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BU JIN, HUANG SHIYUAN, WANG JUE, XIA TONG, LIU HUI, YOU YA, WANG ZHAOHUI, LIU KUN: "The GABAA Receptor Influences Pressure Overload-Induced Heart Failure by Modulating Macrophages in Mice", FRONTIERS IN IMMUNOLOGY, vol. 12, XP093092298, DOI: 10.3389/fimmu.2021.670153 *
DIAL CATHERINE F., TUNE MIRIYA K., DOERSCHUK CLAIRE M., MOCK JASON R.: "Foxp3 + Regulatory T Cell Expression of Keratinocyte Growth Factor Enhances Lung Epithelial Proliferation", AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY., AMERICAN LUNG ASSOCIATION, NEW YORK, NY, US, vol. 57, no. 2, 1 August 2017 (2017-08-01), NEW YORK, NY, US , pages 162 - 173, XP093092300, ISSN: 1044-1549, DOI: 10.1165/rcmb.2017-0019OC *
LOFGREN KRISTOPHER A, SREEKUMAR SREEJA, JENKINS JR E CHARLES, ERNZEN KYLE J, KENNY PARAIC A: "Anti-tumor efficacy of an MMAE-conjugated antibody targeting cell surface TACE/ADAM17-cleaved Amphiregulin in breast cancer", DEPARTMENT OF MEDICINE, UNIVERSITY OF WISCONSIN SCHOOL OF MEDICINE AND PUBLIC HEALTH , MADISON, WISCONSIN, USA, vol. 4, no. 4, 8 October 2021 (2021-10-08), pages 252 - 261, XP093092299, DOI: 10.1093/abt/tbab026 *

Similar Documents

Publication Publication Date Title
JP6905163B2 (ja) サイトカイン放出症候群を予測するバイオマーカー
JP7430136B2 (ja) 疲弊したt細胞に関連する疾患を治療するための方法および組成物
Spivey et al. Gene expression profiling in acute allograft rejection: challenging the immunologic constant of rejection hypothesis
Smith et al. Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E–deficient mice
JP7038353B2 (ja) エクスビボのbite活性化t細胞
EP3695408A1 (fr) Procédés et compositions pour détecter et moduler une signature génétique de résistance à l'immunothérapie dans un cancer
EP3606518A1 (fr) Méthodes et compositions de détection et de modulation d'une signature génique de résistance à l'immunothérapie d'un cancer
Ayano et al. Increased CD226 expression on CD8+ T cells is associated with upregulated cytokine production and endothelial cell injury in patients with systemic sclerosis
KR20200010179A (ko) 세포 요법에 있어서 투약과 관련된 조성물, 제조 물품 및 방법
CA2963935A1 (fr) Biomarqueurs predictifs de la reactivite therapeutique a une therapie par recepteurs antigeniques chimeres et leurs utilisations
JP2016525873A (ja) T細胞バランス遺伝子発現、組成物およびその使用方法
KR20200116081A (ko) 유전자 조작된 세포의 투약 및 조절 방법
Bednar et al. Exploiting CD22 to selectively tolerize autoantibody producing B-cells in rheumatoid arthritis
US20190178888A1 (en) Methods of determining prognosis of sepsis and treating same
US11262358B2 (en) Infiltrating immune cell proportions predict anti-TNF response in colon biopsies
Sharma et al. Mucosal-associated invariant T cells develop an innate-like transcriptomic program in anti-mycobacterial responses
Hesse et al. Profound inhibition of CD73-dependent formation of anti-inflammatory adenosine in B cells of SLE patients
US20230203157A1 (en) Bispecific molecules for selectively modulating t cells
Fatica et al. How has molecular biology enhanced our undertaking of axSpA and its management
JP2008502368A (ja) Siglec−6関連疾患の診断および処置
Schett The role of ACPAs in at-risk individuals: Early targeting of the bone and joints
WO2023172699A1 (fr) Traitement d'une maladie coronarienne
WO2023173024A2 (fr) Traitement d'une coronaropathie par réduction de l'activité de lymphocytes t inter-réactifs
WO2020160285A1 (fr) EXPRESSION DE SIRPα SUR LYMPHOCYTES T COMME BIOMARQUEUR DE LYMPHOCYTES T FONCTIONNELS DURANT L'ÉPUISEMENT
Byng-Maddick Differential effects of anti-TNF therapies for inflammatory arthritides on immune responses to Mycobacterium tuberculosis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23767486

Country of ref document: EP

Kind code of ref document: A1