WO2019099788A1 - Dosage de lymphocytes t régulateurs spécifiques d'un antigène - Google Patents

Dosage de lymphocytes t régulateurs spécifiques d'un antigène Download PDF

Info

Publication number
WO2019099788A1
WO2019099788A1 PCT/US2018/061462 US2018061462W WO2019099788A1 WO 2019099788 A1 WO2019099788 A1 WO 2019099788A1 US 2018061462 W US2018061462 W US 2018061462W WO 2019099788 A1 WO2019099788 A1 WO 2019099788A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
antigen
molecule
treg
detection molecule
Prior art date
Application number
PCT/US2018/061462
Other languages
English (en)
Inventor
Eric WAMBRE
Blake RUST
Original Assignee
Benaroya Sesearch Institute At Virginia Mason
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 Benaroya Sesearch Institute At Virginia Mason filed Critical Benaroya Sesearch Institute At Virginia Mason
Publication of WO2019099788A1 publication Critical patent/WO2019099788A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464839Allergens
    • A61K39/46484Allergens from pollen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70507CD2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56972White blood cells

Definitions

  • T cells with the primary purpose of suppressing other T cells through both contact-mediated and soluble mechanisms are referred to as regulatory' T cells (Tregs).
  • Tregs regulatory' T cells
  • Their primary' function serves to prevent autoimmune reactivity' to self-antigens by conferring peripheral tolerance after education in the thymus (thymic Tregs)
  • thymic Tregs thymic Tregs
  • Regulatory T cells can also he induced in the periphery (peripheral Tregs).
  • peripheral Tregs are not specific to autoantigens, but instead are specific to exogenous antigens found in allergens and pathogens.
  • Regulatory T cells are also implicated m the suppression of anti tumor responses intended to be directed against self-antigens carried on proliferating host cancer cells.
  • Tregs Absence or dysregulation of Tregs can conversely fail to protect against autoimmune responses by failing to suppress autoreactive effector T cell responses (Teffs). Regulatory T cells exert their suppression by both inhibiting the proliferation of Teffs as well as suppressing cytokine production in their target population.
  • Teffs autoreactive effector T cell responses
  • CD25 Regulatory T cells were first identified as CD4+ T ceils carrying abundant amounts of the alpha chain of the high-affinity IL-2 receptor (CD25). While increased expression of CD25 can be used to identify Tregs, CD25 is also expressed on large numbers of effector ( 1)4 T cells because IL-2 is essential for T cell survival and proliferation. Tims, CD25 can also be upregulated on activated CD4+ T cells, making the separation of regulatory T cells and activated T effector cells difficult based on the presence of CD25. High purity is the aim for therapeutic Tregs with the potential to treat autoimmune diseases like rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes (T1D), and psoriasis.
  • RA rheumatoid arthritis
  • MS multiple sclerosis
  • T1D type 1 diabetes
  • psoriasis psoriasis.
  • Tregs include graft versus host disease (GvHD) and allergic diseases, where effector T cells responses are implicated in the rejection of grafts or the induction of allergy.
  • GvHD graft versus host disease
  • allergic diseases where effector T cells responses are implicated in the rejection of grafts or the induction of allergy.
  • regulatory T cells need to be isolated without any contaminating effector T cells that would exacerbate the disease response.
  • Tregs thymic Tregs
  • pTregs peripheral Tregs
  • Tregs can be identified on the basis of high levels of CD25, with the limitations discussed above.
  • CD 137 (4-1 BB) is a molecule that has been noted to discriminate antigen-specific Tregs and CD8+ T cells.
  • CD137 expression is no more effective at discriminating antigen-specific Tregs than CD25.
  • CD154 (CD40L) expressing effector T cells must be removed prior to CD137 enrichment, because CD154+ effector T ceils begin to express CD137 over extended periods of activation and serve as contamination to the CD137+ Tregs.
  • antigen-specific Tregs express CD137 at shorter intervals (4 hours) whereas antigen-specific effector T cells only begin to express CD137 at (12-16 hours).
  • it has been shown that the expression of CD137 on CD 154+ Teffs occurs at a shorter interval (6-8 hours) when no artificial costimulation, such as anti-CD28, is added.
  • Teffs are defined as all T cells that do not belong to the Treg category', which is indicated by stable expression of the transcription factor Foxp3, along with high levels of CD25 and little to no CD127. These Teffs typically exhibit their effector function through the secretion of cytokines such as IL-2, IL-4, IL-9, IL-17, IL-22, and IFNy. The Teffs may or may not be antigen-specific.
  • the secretion of IL-10 alone without subsequent Foxp3 expression does not exclude a T cell from the Teff distinction, as this type of suppressive capability can arise independently of Foxp3 expression but still serves as an essential suppressive T cell subset.
  • the disclosure provides a method of detecting an activated regulatory' T (Treg) cell.
  • the method comprises:
  • Treg regulatory' T
  • the disclosure provides a method of producing an enriched population of activated T regulator ⁇ ' (Treg) cells.
  • the method comprises:
  • Treg regulatory T
  • the disclosure provides a cell in the enriched population produced from the method described herein.
  • the disclosure provides a method of treating a condition treatable by the presence of an activated Treg.
  • the method comprises administering the cell described herein to a subject in need thereof.
  • the disclosure provides a method for monitoring a T regulator ⁇ ' (Treg) cell response to potential exposure of a subject to an antigen.
  • the method comprises:
  • the disclosure provides a method for monitoring the sensitivity of a subject to an antigen of interest.
  • the method comprises:
  • Treg T regulatory
  • the disclosure provides a method of screening for T regulatory (Treg) cell stimulatory epitopes from an allergen or antigen of interest.
  • the method composes:
  • Treg cells exposing the Treg cells to an epitope derived from an antigen of interest in an MHC context
  • Treg cells contacting the Treg cells with a first detection molecule that specifically binds to a latent TGFfi complex protein and a second detection molecule that specifically binds to a co-stimulatory marker;
  • the disclosure pro vides a method of identifying the MHC Class II molecule that binds to a Treg stimulatory epitope.
  • the method comprises:
  • Treg cells exposing the Treg cells to an epitope derived from an antigen of interest in an MHC context
  • Treg cells contacting the Treg cells with a first detection molecule that specifically binds to a latent TGFfi complex protein and a second detection molecule that specifically binds to a co-stimulatory marker;
  • the disclosure provides a method of testing the effect of a putative therapeutic compound on an activated T regulatory (Treg) cell.
  • the method comprises exposing a ceil described herein to the therapeutic compound.
  • FIGURES 1A and IB illustrate that de novo expression of GARP/LAP within CD137 distinguishes highly activated Tregs. Shown are flow cytometric output following polyclonal T cell stimulation of PBMCs for 18 hours. It is shown that antigen- specific Tregs express GARP/LAP and 0X40 in CD137hi, but not CDI37mid compartments. GARP and 0X40 expression, and thus antigen specificity is limited to only the CD137hi, CD154neg compartment. (FIGURE 1 A illustrates the lack of GARP, LAP, and 0X40 in the CDl37mid compartment, while FIGURE IB illustrates the presence of these molecules in the CD137hi compartment).
  • FIGURE 2 illustrates that antigen-specific Tregs express GARP/LAP and 0X40 in CD137hi, but not CD137mid compartments. Shown are flow cytometric output following polyclonal T cell stimulation of PBMCs for 18 hours. It is shown that antigen- specific Tregs express GARP/LAP and 0X40 in CD137hi, but not CD137mid compartments. GARP and 0X40 expression, and thus antigen specificity, is limited to only the CD137hi, CD154neg compartment. (Inset box “A” illustrates the lack of GARP, LAP, and 0X40 in the CDI 37mid compartment, while inset box “B” illustrates the presence of these molecules in the CD I37hi compartment).
  • FIGURE 3A and 3B graphically illustrate that CD137+ Tregs expressing GARP/LAP exhibit higher levels of activation in multiple disease models.
  • Patient PBMCs were isolated from various disease states and stimulated with relevant peptide antigen pools m cancer (tumor-associated antigens), autoimmunity (T1D pancreatic islet antigens), allerg ⁇ (Ain g 1 alder antigen), and vaccine (HA1 influenza antigen) for 16 hours. Cultures were then enriched for CD 137 and assessed for GARP/LAP expression, as well as other Treg-related activation markers (FIGURE 3A). Across all antigen models, GARP/LAP+ Tregs expressed increased activation as compared to CD137+ Tregs.
  • FIGURE 4 illustrates that optimal CD 137 and GARP/LAP expression occurs at 16 hours of ex vivo stimulation with peptide antigen.
  • PBMCs from a healthy patient were stimulated with influenza HA1 peptide antigen for 0, 2, 6, 8, 12, 16 and 18 hours to determine optimal expression.
  • Foxp3+ CD25+ Tregs begin to express slight levels of CD137 and GARP/LAP at 6 hours, expression plateaus at 12-16 hours. Similar trends can be seen when tracking percent expression within memory CD4+ T cells.
  • Ki67 expression over the same times and found there was no change, indicating that ex vivo stimulation did not induce cell division.
  • phenotypic markers of Tregs like CD39 we also saw no changes in expression over the same timeframe (data not shown).
  • FIGURES 5A-5D illustrate that GARP/LAP expression is necessary for detection of highly Foxp3-ennched antigen-specific Tregs.
  • GARP/LAP expression in conjunction with other activation-induced markers FIGURE 5 A-CD 137, FIGURE 5B-OX40, FIGURE 5C-CD27, FIGURE 5D-CD25
  • highly enriched antigen- specific Foxp3+ Tregs can be isolated by sorting or magnetic enrichment (Foxp3 purity can be seen in corresponding bottom panels).
  • FIGURE 6 graphically illustrates that polyclonal activation of human PBMCs induces enriched regulatory T cell activation GARP+ 0X40+ and CD137+ CD25+ expression.
  • HA1 influenza hemagglutinin peptide pools
  • the polyclonal stimulation When removing CD 154+ effectors prior to the CD137+ CD25+ analysis, the polyclonal stimulation reaches a lower percent of activation for regulatory T ceils, with a higher background. When using just GARP and 0X40 expression with no CD 154 removal, the polyclonal stimulation is higher than CD137+ CD25+ CD154- analysis and has less background in the DMSO control.
  • the GARP+ 0X40+ compartment is also enriched for Foxp3 expression (-95% Foxp3+) as compared to the CD137+ CD25+ CD 154- compartment (-90% Foxp3+) (data not shown).
  • FIGURES 7A and 7B illustrate that CD 154+ T effectors do not express GARP/LAP after ex vivo stimulation.
  • PBMCs were isolated from a grass allergic patient and stimulated for 6 or 18 hours with grass allergen Phlp peptide pools or tetanus toxoid peptide pools.
  • FIGURE 7A GARP/LAP expression is not observed during the 6 hour stimulation in effectors (CD 154+ CD137+) or Tregs (CD154- CD 137+) at 6 hours.
  • FIGURE 7B After 18 hours, GARP/LAP expression is induced by both model antigens, however it is only observed on Tregs (CD154- CD 137+) and not Teffs (CD154+ CD 137 i.
  • FIGURES 8A and 8B graphically illustrates that CD137+ regulatory T cells are distinct in their gene expression from CD154+ effector T cells, and GARP+ CD137+ regulatory T cells are distinct from GARP- CD 137 cells.
  • RNAseq and transcript profiles were clustered by principal component analysis (PCA) (FIGURE 8A).
  • PCA principal component analysis
  • Significantly upreguiated and differentially expressed regulator ⁇ ' T cell-associated genes m GARP+ CD137+ Tregs (green) and GARP- CD137+ Tregs (red) were plotted (FIGURE 8B).
  • FIGURES 9A-9D graphically illustrate that GARP/LAP + CD 137+ regulatory T cells have increased regulatory potential in peanut allergic patients.
  • Increased co-inhibitory expression of CTLA-4 (p ⁇ 0.01) (FIGURE 9B) and LAG-3 ip 0.05 ⁇ (FIGURE 9C) 'as seen in GARP+ populations, as 'ell as inhibitory cytokine IL-10 (p ⁇ 0.05) (FIGURE 9A) expression when measured by paired T test analysis.
  • increased expression of CCR8 p ⁇ 0.Gl
  • FIGURE 9D was observed wiien measured by paired T test analysis.
  • FIGURES 10A-10D illustrate that GARP/LAP+ Treg cell lines isolated from patients stain more effectively with cognate tetramer than CD137+ Treg cell lines.
  • GARP+ CD137+ and GARP- CD137+ regulatory T cells were sorted after ex vivo stimulation of alder allergic patients with Alngl p48 peptide. After two weeks m vitro culture, the lines were stained with cognate tetramer and analyzed by flow' cytometry.
  • GARP+ CD137+ cell lines stain more effectively with Alngl p48 tetramer than GARP- CD137+ cell lines (FIGURE 10B) from the same patient.
  • GARP/LAP+ Treg lines had significantly higher staining (FIGURE 10C).
  • the same result was demonstrated in influenza HA 1 -specific Treg lines sorted for GARP/LAP expression (FIGURE 10D). * represents p ⁇ 0.05 as measured by paired T test analysis.
  • FIGURE 11A and TIB graphically illustrate increased antigen-specificity in the GARP+ CD137+ population as compared to the GARP- CD137+ population when assessed by the number of ceils secreting the inhibitory cytokine IL-10.
  • FIGURE 11 A PBMCs isolated from a patient were stimulated with a tumor-associated antigen (TAA) peptide pool. Cells were magnetically enriched for GARP and LAP and CD 137 and were then sorted and cultured for 2 weeks in vitro. After culture, the cells were then restimulated with either the relevant TAA peptide pool (black bars) or an irrelevant influenza (HA1) peptide pool (gray bars) for 18 hours.
  • TAA tumor-associated antigen
  • FIGURE 11B PBMCs isolated from a patient were stimulated with an autoimmune peptide pool including antigens specific for type 1 diabetes (T1D). Cells were magnetically enriched for GARP and LAP and CD137 and were then sorted and cultured for 2 weeks in vitro.
  • the cells were then restimulated with either the relevant T1D peptide pool (black bars) or an irrelevant influenza (HA1) peptide pool (gray bars) for 18 hours. Ceils were then plated into an ELISpot specific for IL-10 to assess their antigen-specificity in response to relevant and irrelevant peptide restimulation.
  • GARP+ CD137+ fines had a significant increase (p ⁇ 0.001) in the number of cells with the ability' to secrete IL-10 with relevant peptide stimulation as compared to irrelevant stimulation using T test analysis, while GARP- CD137+ fines showed no significant difference m IL-10-secreting cells when comparing irrelevant and relevant peptide stimulation.
  • FIGURE 12 illustrates that sorted GARP/LAP+ cell lines respond to antigen- specific restimulation through the upregulation of costimulatory molecules.
  • GARP/LAP+ CD137+ cell lines respond to antigen-specific restimulation through the upregulation of Treg-associated costimulatory' molecules while GARP- CD137+ cell lines do not.
  • PBMCs isolated from a patient were stimulated with an autoimmune peptide pool including antigens specific for type 1 diabetes (T1D). Cells were magnetically enriched for GARP and LAP and CD 137 and were then sorted and cultured for 2 weeks in vitro. After culture, the cells were then restimulated with either the relevant T1D peptide pool (solid line) or an irrelevant influenza (HA! ) peptide pool (dashed line) for 18 hours. Upregulation of costimulatory molecules CD 137 and 0X40, and activation molecule CD69 were seen only in the relevant restimulation of the GARP/LAP+ CD137+ lines.
  • FIGURES 13A and 13B graphically illustrate that GARP/LAP+ Tregs proliferate in response to vaccination whereas CD137+ Tregs do not. Correlations were plotted between percent Ki67 expression and either percent CD 137 expression (FIGURE. 13A) or percent GARP/LAP expression (FIGURE 13B). Eight patients were assayed for regulatory ' T cell proliferation two weeks after flu vaccination by restimulation with HA1 influenza peptide. Percent expression of GARP/LAP expression has been correlated with either CD137 alone or CD137+ GARP/LAP+.
  • FIGURES 14A-14C graphically illustrate that allergen-specific GARP/LAP+ Tregs proliferate m response to seasonal allergen exposure more effectively than CD137+ Tregs alone.
  • Increased Ki67 expression in GARP+ Tregs indicated increased antigen- specificity' after natural allergen exposure.
  • Patient blood was collected before the alder allergen season and two weeks post-peak alder allergen exposure to assess changes in Ki67 expression in antigen-specific regulator ⁇ T cell populations (FIGURE 14A).
  • FIGURE 15 graphically illustrates that GARP and LAP are coexpressed within CD 137+ CD 154- Tregs.
  • PBMCs from a healthy patient were stimulated polyclonally for 18 hours and enriched for GARP/LAP/CD137.
  • GARP and LAP are reliably co-expressed as parts of the latent TGFfl complex.
  • FIGURE 16 graphically illustrates that increased Foxp3 MFI in GARP/LAP+ Tregs indicates increased activation status.
  • PBMCs were stimulated for 18 hours ex vivo with HA1 peptides and enriched for CD137/GARP/LAP. Ceils were then permeabilized and stained forFoxp3 expression. Significance was determined by paired T test with pO.OOOl.
  • FIGURES 17A-17C illustrate that single cell RNAseq shows that GARP/LAP+ Tregs have a mutually exclusive TCR repertoire as compared to CD 154+ Teffs.
  • 48 CD 154+ Teffs and 48 CD 137+ GARP/LAP+ Tregs were sorted from a grass-allergic patient during season after 18 hour stimulation with Phi p 5a and Phi p 5b peptides.
  • scRNAseq we were able to effectively cluster the Teffs (orange) and Tregs (blue) using PCA analysis (FIGURE 17A).
  • FIGURE 18 graphically illustrates that increased proliferation in GARP/LAP+ Tregs m response to influenza vaccination only observed in peptide treatment groups. Correlations were plotted between percent Ki67 expression and either percent CD137 expression or percent G ARP/LAP expression. Eight patients were assayed for regulatory T cell proliferation two weeks after flu vaccination by restimulation with HA1 influenza peptide. Percent expression of GARP/LAP expression has been correlated with either CD137 alone or CDI37+ GARP/LAP+. The correlation is extremely significant when using GARP/LAP, but nonexistent m the CD 137 alone group when compared to % of ceils in these groups expressing Ki67, a marker we use here as a surrogate for recent antigen exposure from the vaccination. These correlations are not observed in the control groups.
  • FIGURES 19A and 19B graphically illustrate the use of a novel sequential CD154/GARP/CD137 assay to monitor the ex vivo frequency, surface markers and phenotypes of antigen-specific T cells.
  • Sequential CD154/GARP/CD137 assays were carried out with peptide pools derived from TAA (FIGURE 19A) or self-antigens (FIGURE 19B).
  • the self-antigen pool included only peptides from islet antigens, GAD65 and IGRP.
  • Subjects tested include an HC individual (top row); and a renal cell carcinoma patient sampled before ICI therapy with nivolumab (middle row) and after two cycles of ICI (bottom row). The patient was assessed as having stable disease, but was taken off therapy because of complications due to a preexisting condition. Asterisks indicate massive increases in CD8+ Teff cells reactive with tumor- and auto- antigens.
  • Tregs antigen-specific regulatory T cells
  • the latent TGFft complex includes the pro-TGF receptor GARP (LRRC32) as well as the latency associated peptide (LAP) and is exclusively expressed by antigen-specific Tregs and not antigen-specific Teffs designated by CD 154 expression GARP expression is directly controlled by Foxp3 transcriptional programming, providing a direct link between the prevalence of this complex exclusively on activated Tregs While TGFfl is known to be a critical cytokine in the induction of Tregs, as well as an important inhibitory molecule in Treg suppressive capability, the latent complex (GARP and LAP) also selectively identifies antigen- specific Tregs m humans.
  • GARP pro-TGF receptor GARP
  • LAP latency associated peptide
  • GARP and LAP can be used in conjunction with costimulatory molecules like CD137, 0X40, CD27, ICOS, and CD25 to selectively identify and isolate highly pure populations of Tregs for immune monitoring or therapeutic applications.
  • costimulatory molecules like CD137, 0X40, CD27, ICOS, and CD25.
  • the present disclosure provides methods and compositions for detecting, monitoring, enriching for, isolating, and using activated T regulator ⁇ ' (Treg) cells.
  • Treg activated T regulator ⁇ '
  • Tregs regulatory T cells
  • Tregs refer to a subpopulation of T cells of the immune system.
  • suppressor T cells Tregs work to modulate immune responses to maintain tolerance to self-antigens and to prevent autoimmune disease. They are also known to have an effect to suppress or prevent inappropriate responses to allergens. They generally work by affecting a downregulation in the induction and proliferation of effector T cells Teffs.
  • the disclosure provides a method of detecting an activated regulator ⁇ ' T (Treg) cell.
  • the method comprises obtaining a sample comprising regulator ⁇ ' T (Treg) cells that have been exposed to an antigen and/or activator in a manner such that the sample may contain activated Treg cells; contacting the sample with a first detection molecule that specifically binds to a latent TORb complex protein and a second detection molecule that specifically binds to a co-stimulatory marker; and detecting a cell in the sample that is bound by the first detection molecule and the second detection molecule, thereby detecting an activated Treg cell.
  • the exposure of the Tregs to an antigen and/or acti vator allows any Treg cells in the sample to progress to activated Tregs if the conditions are appropriate.
  • the Tregs have been exposed to an antigen of interest, which facilitates detection of activated Tregs that are specific for the antigen of interest.
  • the antigen of interest is typically a peptide or proteinaceous antigen.
  • the exposure of the antigen of interest to the Tregs occurs "in a manner such that the sample may contain activated Treg ceils", which refers to the appropriate presentation of a peptide epitope to the Treg within an peptide/MHC complex.
  • the peptide/MHC complex can be expressed by an antigen presenting cell (APC) where the APC has process the antigen and loaded the peptide into its own MHC, according to processes well-understood in the art.
  • APCs that have been exposed to the antigen of interest, have been exposed to the Tregs under conditions sufficient for the ceils to come into contact.
  • the peptide MHC complex if properly matched to the Treg, can stimulate activation of those Tregs that are specific for the peptide loaded onto the MHC.
  • the peptides/MHC complexes are not expressed on APCs, but rather are loaded onto and complexed with MHC monomer or multimers, according to approaches known in the art, to produce soluble peptide/MHC complexes that can also stimulate activation in Tregs that are matched to the MHC type and specifically recognize the epitope in the MHC context.
  • the Treg cells have been exposed to an activator that, with sufficient quantities and conditions, can produce non-specific activation of Treg cells.
  • the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected from CD 137, 0X40 (CD 134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • the activator comprises costimulatory molecules that bind to and activate one or more receptors selected from CDS, CD2, and CD28.
  • the co-stimulator molecules can be ligands or antibodies that bind to and activate signalin cascades associated with the above receptor molecules. Ligands and antibodies (or functional fragments thereof) are well-known in the art.
  • the one or more co-stimulatory molecules can be administered as a cocktail that, in sufficient concentration, can stimulate non-antigen specific activation of Tregs the sample.
  • the activator is contacted to the sample with the antigen of interest (as described above), where the activator is formulated to merely enhance the activation of antigen-specific Tregs and not the general population of the Tregs (i.e., not in an antigen-independent fashion).
  • an exemplary activator cocktail for enhanced antigen-specific activation is described in more detail below.
  • the term "latent TOTb complex protein” includes any one of GARP (LRRC32), LAP (latent-associated peptide), and TGFfll-4.
  • This disclosure encompasses embodiments where the first detection molecule can specifically bind to any one of the above latent TORb complex proteins. Considering that these protein associate in a complex, in other embodiments, the first detection molecule can bind to a complex or association of any two or more of the above proteins. In some embodiments, additional detection molecules are used that bind to additional latent TORb complex proteins (or complexes thereof) that are different than the latent TORb complex proteins (or complexes thereof) that are bound by the first detection molecule.
  • the second detection molecule specifically binds to a co-stimulatory marker on a Treg ceil that can indicate progression to an activated state.
  • the co-stimulatory marker can he, for example, receptors specific or substantially unique to activated Treg cells, which typically are receptors that would bind to cognate ligands on, e.g., APCs.
  • the co-stimulatory marker can be selected from CD 137, 0X40 (CD 134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • the method further comprises contacting the sample with a third detection molecule that specifically binds to CD4, and wherein detecting an activated T regulatory cell comprises detecting a cell in the sample that is bound by the first detection molecule, the second detection molecule, and the third detection molecule. It is the combination of the detectable presence of the latent T ⁇ Rb complex protein, the co-stimulatory molecule, and in some cases, CD4, that provides an accurate indication of activation of the Treg cells. When the cells are produces as the result of exposure to a particular antigen, the activated cells are specific for the antigen.
  • detection molecule and “enrichment molecule” (see below) refer to any molecule having an ability to bind to a specific target molecule (e.g., a marker of interest such as a latent TORb complex protein or a eo-stimulatory molecule) with a specific affinity' (i.e., detectable over background).
  • a specific target molecule e.g., a marker of interest such as a latent TORb complex protein or a eo-stimulatory molecule
  • affinity' i.e., detectable over background
  • Exemplary categories of detection/enrichment molecules that can be used in the context of the present disclosure include antibodies, antibody derivatives (also referred to as "antibody-like molecules"), functional target molecule-binding portions of antibodies or antibody-like molecules, peptides that specifically interact with a particular target antigen/marker (e.g., peptibodies), receptor molecules that specifically interact with a particular target antigen/marker, functional target/marker-binding portions of proteins that comprise a ligand-binding portion of a receptor that specifically binds a particular target/marker, ligands themselves (or moieties that incorporate a ligand), antigen/marker-binding scaffolds (e.g., DARPms, HEAT repeat proteins, ARM repeat proteins, tetratricopeptide repeal proteins, and other scaffolds based on naturally occurring repeat proteins, etc., (see, e.g., Boersma and Pluckthun, Curr. Opin. Biotechnol. 22: 849-8
  • the detection/enrichment molecule is an antibody.
  • antibody encompasses antibodies and antibody fragments thereof, derived from any antibody-producing mammal (e.g., mouse, rat, rabbit, and primate including human), that specifically bind to a target molecule of interest (i.e., latent TGF complex protein, co-stimulatory marker, etc.)
  • target molecule of interest i.e., latent TGF complex protein, co-stimulatory marker, etc.
  • Exemplary antibodies include polyclonal, monoclonal and recombinant antibodies; multispecific antibodies (e.g., bi specific antibodies); humanized antibodies; murine antibodies; chimeric, mouse-human, mouse- primate, primate-human monoclonal antibodies; and anti-idiotype antibodies.
  • the antigen-binding molecule can be any intact antibody molecule or fragment thereof (e.g., with a functional antigen-binding domain).
  • An antibody fragment is a portion derived from or related to a full-length antibody, preferably including the complementarity-determining regions (CDRs), antigen binding regions, or variable regions thereof.
  • Illustrative examples of antibody fragments useful in the present disclosure include Fab, Fab', F(ab)2, F(ab')2 and Fv fragments, scFv fragments, diabodies, linear antibodies, single-chain antibody molecules, multispecific antibodies formed from antibody fragments, and the like.
  • a "single-chain Fv” or "scFv” antibody fragment comprises the VJJ and Vp domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide can further comprise a polypeptide linker between the VJJ and V domains, which enables the scFv to form the desired structure for antigen binding.
  • Antibody fragments can be produced recombinanily, or through enzymatic digestion.
  • Antibodies can be further modified to suit various uses. For example, if the detection markers are intended for administration to a subject, a chimeric antibody can be used to minimize antigenicity of the binding molecule itself.
  • a chimeric antibody is a recombinant protein that contains the variable domains and complementarity-determining regions (CDRs) derived from a non-human species (e.g., rodent) antibody, while the remainder of the antibody molecule is derived from a human antibody.
  • CDRs complementarity-determining regions
  • a "humanized antibody” is a chimeric antibody that comprises a minimal sequence that conforms to specific complementanty-determimng regions derived from non-human immunoglobulin that is transplanted into a human antibody framework. Humanized antibodies are typically recombinant proteins in which only the antibody complementarity-determining regions (CDRs) are of non-human origin.
  • the production of a polyclonal antibody can be accomplished by administering an immunogen containing the target molecule of interest to an antibody-producing animal.
  • the target molecule of interest can be administered to a mammal (e.g., a rat, a mouse, a rabbit, a chicken, cattle, a monkey, a pig, a horse, a sheep, a goat, a dog, a cat, a guinea pig, a hamster) or a bird (e.g., a chicken) so as to induce production of a serum containing an target molecule-specific polyclonal antibody.
  • a mammal e.g., a rat, a mouse, a rabbit, a chicken, cattle, a monkey, a pig, a horse, a sheep, a goat, a dog, a cat, a guinea pig, a hamster
  • a bird e.g., a chicken
  • the target molecule can be administered in combination with other components known to facilitate induction of a B-cell response, such as any appropriate adjuvant known in the art.
  • the polyclonal antibody reagent can be further processed to remove or subtract any antibody members that have unacceptable affinity for antigens that are not the antigen of interest. Tire resulting polyclonal antibody reagent will exhibit enhanced specificity for the target molecule and are useful for detection and quantification purposes.
  • Many approaches for adsorption of polyclonal antibody reagents to reduce cross-reactivity exist, are familiar to persons of ordinary skill in the art, and are encompassed by the present disclosure.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow' et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory' Press, 2nd ed. 1988); Hammerling et ah, in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N.Y., 1981), incorporated herein by reference in their entireties.
  • the term "monoclonal antibody” refers to an antibody that is derived from a single clone. including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. Methods for producing and screening for specific antibodies using hybridoma technology are routine and well-known in the art.
  • Antibody fragments that recognize specific epitopes can be generated by any technique known to those of skill in the art.
  • Fab and F(ab')2 fragments of the invention can be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • F(ab3 ⁇ 4 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
  • the antibodies of the present invention can also be generated using various phage display methods known in the art.
  • nucleic acid aptamers refers to oligonucleic or peptide molecules that can hind to specific target molecules.
  • Nucleic acid aptamers usually are short strands of oligonucleotides that exhibit specific binding properties. They are typically produced through several rounds of in vitro selection or systematic evolution by exponential enrichment protocols to select for the best binding properties, including avidity and selectivity.
  • One type of useful nucleic acid aptamers are thioaptamers, in which some or all of the non-bridging oxygen atoms of phosphodiester bonds have been replaced with sulfur atoms, which increases binding energies with proteins and slows degradation caused by nuclease enzymes.
  • nucleic acid aptamers contain modified bases that possess altered side-chains that can facilitate the aptamer/target binding.
  • Peptide aptamers are protein molecules that often contain a peptide loop attached at both ends to a protamersem scaffold.
  • the loop typically has between 10 and 20 amino acids long, and the scaffold is typically any protein that is soluble and compact.
  • One example of the protein scaffold is Thioredoxin-A, wherein the loop structure can be inserted within the reducing active site.
  • Peptide aptamers can be generated/selected from various types of libraries, such as phage display, mRNA display, ribosome display, bacterial display and yeast display libraries.
  • the antigen-binding molecule is a receptor molecule or comprises a binding domain of a receptor molecule.
  • the receptor molecule can be any receptor known that can specifically bind the target molecule as the ligand.
  • the antigen- binding molecule is or contains a protein binding domain that enables the detection of the target molecule.
  • the antigen-binding molecule can be or comprise a ligand or portion of a ligand that is specific for a receptor or a binding domain of a protein, which receptor or a binding domain of a protein would then serve as the target molecule.
  • the term "selectively binds" refers to the ability of the antigen binding molecule to bind to the target molecule, without significant binding to other unrelated molecules, under standard conditions known m the art.
  • Tire antigen-binding molecule can bind to other peptides, polypeptides, or proteins, but with lower affinity as determined by, e.g., immunoassays, BIAcore, or other assays known in the art.
  • antigen-binding molecule preferably does not cross-react with other antigens.
  • the detection reagents are typically delectably labelled. Conventional labels that can be used are well-known in the art. Typically, the first and second (and third, etc.) detection molecules are each labeled with a different label such that they can be distinguishable from each other. The detectable labels facilitate efficient detection of binding of the first and second (and third, etc.) binding molecules to their intended targets on the Treg ceils.
  • FACS fluorescence-activated cell sorting
  • CyTOF mass cytometry
  • the binding molecules are labeled with isotopicaliy pure elements produce signature signals in a time-of-flight mass spectrometer.
  • Treg cells that have the first and second detection molecules bound thereto will produce a distinct signature as compared to Treg cells that have only one, or ceils that have neither, of the detection molecules bound thereto.
  • FACS and CyTOF can also be used to collect (e.g., enrich, isolate, etc.) the cells.
  • the method comprises enriching for activated Tregs based on the binding status detected in the method. This can be accomplished, for example, using FACS that sorts and collects cells with a particular fluorescence pattern based on the binding of the detection molecules.
  • enriching means increasing the relative proportion of the activated Tregs as compared to the sample.
  • the activated Tregs are substantially isolated from other cells, such as quiescent/nafve Tregs. In this context, “substantially” implies near complete but does not require complete isolation.
  • Some amount of cells that are not activated Tregs are permitted so long as the proportion remains small (e.g., 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2 %, 1%, or less) of the batch.
  • the sample can be obtained from or derived from a subject.
  • the subject can be any vertebrate animal that produces Treg cells, e.g., human, mouse, rat, rabbit, dog, cat, horse, cow, goat, and the like.
  • Tire sample can be, or be derived from, whole blood, tissue, lavage, tumor biopsy, and the like.
  • the method further comprises an initial step of obtaining or deri ving the sample of Treg cells from a subject.
  • the method comprises the active step of contacting the initial sample comprising Treg cells with sufficient antigen or activator in a manner and amount such that at least a portion of the Treg ceils are activated. Exposure to antigen of interest and activator compositions are described above. Furthermore, a description of an illustrative approach to contacting the sample comprising Tregs with antigen of interest and activator to activated Tregs is provided below.
  • the method further comprises contacting the cells with a co-stimulatory molecule, as described above.
  • the disclosure provides a method of producing an enriched population of activated T regulatory (Treg) cells.
  • the method comprises incubating a sample comprising Treg cells with an antigen of interest and/or an activator in a manner and amount such that at least a portion of the Treg cells are activated; contacting the sample with a first enrichment molecule that specifically binds to a latent TGFp complex protein and a second enrichment molecule that specifically binds to a co-stimuiatory marker; and enriching for cells that are bound to the first enrichment molecule arid the second enrichment molecule, thereby producing an enriched population of activated Treg cells.
  • the method further comprises an initial step of obtaining the sample of sample of Treg cells from a subject.
  • a sample can be, or be derived from, whole blood, tissue, lavage, tumor biopsy, and the like, which is obtained from a subject.
  • the sample is incubated with an antigen of interest, as described above, which results in antigen-specificity of the activated Tregs.
  • the sample is incubated with an activator, as described above, in a manner that induced antigen-independent activation of the Tregs.
  • the sample is incubated with an antigen of interest and sufficient activator, in a manner that further promotes the activation of antigen -sped fie Tregs but does not result in substantial activation of Tregs that are not specific for the antigen of interest.
  • the enriched Tregs can be administered to a subject in need thereof, for example, to treat or ameliorate a condition characterized by inappropriate or excessive Teff response to an antigen.
  • a condition characterized by inappropriate or excessive Teff response to an antigen include allergies, autoimmune disease, graft vs host reactivity, and the like.
  • the enriched Tregs are MHC (HLA) matched to the subject.
  • the subject from whom the sample of Tregs was originally obtained is the same subject receiving administration of the enriched activated Tregs.
  • the disclosure also provides the activated Treg cell produced from the above methods.
  • the activated Treg ceil can be specific for an antigen of interest used to produce the activated Treg. Additionally, the Treg can be in an enriched or isolated population of activated Tregs with the same antigen specificity.
  • the disclosure also encompasses methods of using the disclosed activated Treg cells.
  • the disclosure provides a method of treating a condition treatable by administration of an activated Treg.
  • Tire method comprises administering the disclosed Tregs.
  • the condition is an allergy, autoimmune disease, graft vs host reactivity, and the like.
  • the Tregs can be rationally produced to be specific for an antigen of interest that is determined to be a causal component of the condition to be treated.
  • the terms “treat” or “treatment” refer to therapeutic interventions to stop, ameliorate, or manage a disease, disorder, or condition.
  • Therapeutic or prophylactic/preventive benefits include improved clinical outcome; lessening or alleviation of symptoms associated with a condition; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay of disease progression; remission; survival; prolonged survival; or any combination thereof.
  • the disclosure provides a method for monitoring a T regulatory (Treg) cell response to potential antigens.
  • the method comprises contacting a sample with a first detection molecule that specifically binds to a latent TGFj] complex protein and a second detection molecule that specifically binds to a co stimulatory marker.
  • the sample comprises T cells and was obtained from a subject suspected of being exposed to an antigen.
  • the method also comprises detecting or quantifying cells in the sample that are bound by both the first detection molecule and the second detection molecule. Cells that are bound by the first detection molecule and the second detection molecule are established as being activated Tregs.
  • the absence, presence, or relative abundance of cells bound by both the first detection molecule and the second detection molecule is indicative of a state of the Treg cell response.
  • This method can be applied, for example, on a sample obtained or derived from a subject after the subject has been administered or has otherwise been exposed to an antigen of interest.
  • the method can determine whether the subject generates Tregs in response to the administration or exposure.
  • the sample could comprise naive T cells and the method further comprises incubating the sample with an antigen of interest, as described above.
  • this can be applied to determine the responsivity of Treg development and activation to allergens, autoantigens, and/or cancer antigens, and the like.
  • the presence, absence, or level of activated, antigen-specific Tregs is indicative of the subject's capacity to generate Tregs specific for the antigen of interest.
  • the disclosure provides a method for monitoring the sensitivity of a subject to an antigen of interest.
  • the method comprises contacting a sample comprising Treg cells with a first detection molecule that specifically binds to a latent TGFp complex protein and a second detection molecule that specifically binds to a co-stimulatory marker, wherein the Treg cells have been exposed to an antigen of interest and the sample was obtained from the subject; detecting or quantifying the cells in the sample that are bound by both the first detection molecule and the second detection molecule; comparing the number of cells in the sample that are bound by both the fust detection molecule and the second detection molecule to an established threshold to determine the sensitivity of the subject to the antigen of interest.
  • the Treg cells can be been exposed to an antigen of interest, as described above, in vivo, or ex vivo after the same was obtained.
  • a relative increase in the number of cells in the sample that are bound by both the first detection molecule and the second detection molecules compared to the established threshold indicates a low sensitivity of the subject to the antigen of interest.
  • the threshold can be established at an earlier time point from the subject using the same method.
  • a relative increase in cells that are bound by both the first detection molecule and the second detection molecule over the established threshold indicates a decreasing sensitivity to the antigen of interest.
  • a relative decrease in cells that are bound by both the first detection molecule and the second detection molecule over the established threshold indicates an increasing sensitivity to the antigen of interest.
  • the threshold is established during or prior to administration of the therapeutic treatment and the sensitivity of the subject to the antigen of interest at a later time point is indicative of the efficacy of the therapeutic treatment.
  • the disclosure provides a method of screening for T regulatory (Treg) cell stimulatory epitopes from an allergen or antigen of interest.
  • the method comprises steps of: obtaining a sample comprising Treg cells; exposing the Treg cells to an epitope derived from an antigen of interest in an MHC context; contacting the Treg cells with a first detection molecule that specifically binds to a latent TGFfi complex protein and a second detection molecule that specifically binds to a co-stimulatory marker; and quantifying the relative abundance of cells in the sample that are bound by both the first detection molecule and the second detection molecule.
  • a high relative abundance indicates that the epitope stimulates development of activated Treg cells.
  • Treg cells to an epitope Exposing Treg cells to an epitope is generally described above, and can be mediated by antigen presenting cells or can be accomplished with soluble epitope-loaded MHC complexes.
  • the method can further comprise exposing the Tregs to an appropriate amount of activator to facilitate activation of antigen-specific Tregs, but not all Tregs generally. Elements, such as the activator are described above.
  • the method can be repeated multiple times against a panel of different epitopes. For example, multiple epitopes derived from the same antigen can be tested to determine the most (or least) stimulating epitope from the antigen for purposes of producing Tregs.
  • the antigens can be selected from allergen, autoantigen, cancer antigen, and the like.
  • the disclosure provides a method of identifying an MHC molecule/complex that binds to a Treg-stimulatory epitope.
  • the method comprises: obtaining a sample comprising Treg cells; exposing the Treg cells to an epitope derived from an antigen of interest in an MHC context; contacting the Treg cells with a first detection molecule that specifically binds to a latent TORb complex protein and a second detection molecule that specifically binds to a co stimulatory marker; quantifying the relative abundance of cells in the sample that are bound by both the first detection molecule and the second detection molecule, wherein a high relative abundance indicates that the epitope stimulates development of activated Treg cells; and characterizing the MHC molecule complexed with the epitope that resulted in a high abundance of cells bound by both the first detection molecule and the second detection molecule.
  • the Tregs can be exposed separately to a panel of different APCs that have been allowed to process and display an epitope. After quantification of the resulting activated Tregs, the MHC from the APCs resulting in the best activation can be characterized.
  • the disclosure provides for methods of testing effect of a putative therapeutic compound on a Treg ceil, including on the development of a Treg ceil into an activated state.
  • the method comprises exposing the disclosed Treg ceil (before or after activation) and monitoring the effect of the therapeutic compound.
  • the latent TGFf> complex protein and co-stimuiatory marker, activator, or any other elements not explicitly described are defined previously in other contexts, and are understood to be equally applicable here.
  • This Example describes an exemplary protocol for performing an antigen-specific antibody
  • T regulatory ceil assay of the present disclosure also referred to as ASTRA.
  • ASTRA T regulatory ceil assay of the present disclosure
  • Use of the ASTRA approach in combination with multicolor flow cytometry can be a valuable tool for detecting and monitoring of CD4+ regulatory ' T cell response to a particular antigen, determining which epitopes or antigen fragments that bind to MHC class II molecule to elicit a regulatory T cell response, and for the development of specific immunotherapies.
  • the ASTRA method allows simultaneous assessment of other ceil phenotypes or functions, and is compatible with downstream RNA-based assays and preserves cell viability. Accordingly, the ASTRA method allows for tracking antigen- specific T regulatory cells and can be applied for diverse applications, such as:
  • PBMCs were isolated from whole blood using density gradient centrifugation.
  • PBMCs (l-2x l0 7 cells) were then stimulated for 18 hr in RPMI 1640 supplemented with 5% AB serum, with the antigen (peptide, protein, vaccine, crude extract, or stimulatory cocktail) at 37°C and 5% C0 2 .
  • Enriching antibodies (a-GARP, a-LAP) were added to remaining cells at a volume of 3 mE per 50 million cells. 5. Cells were vortexed and resuspended and allowed to incubate for 15-20 minutes at room temperature in the dark.
  • step 6 1 mL of PBS was added to the appropriate number of columns or tubes loaded onto a magnet. Flow through was collected in a new- tube.
  • Ceils were resuspended m 1 mL. of PBS per 50 million cells. Resuspended cells were added to a magnetic column or tube and allow to completely pass through column or settle while the flow through was collected in the same tube as step 7.
  • Surface staining antibody cocktail (including at minimum GARP and/or LAP) w3 ⁇ 4s then added to remaining volume of cells and allowed to incubate for 15-20 minutes at room temperature in the dark.
  • Surface panel addressed by the cocktail can optionally include CD154, CD69, ICOS for live-cell analysis of highly -enriched antigen- specific regulatory T cells. For fixation with Foxp3, Ki67 and Helios staining, proceed to step 15. Otherwise, analyze live ceils after step 15.
  • Nuclear/intracellular staining antibody cocktail was added to the remaining volume of cells. Cells were vortexed and incubated at 4°C for 18-24 hours. Shorter incubations times can be used with discretion.
  • Ceils were resuspended in 200 pL of PBS and analyzed via flow cytometr'.
  • the cells detected and sorted by flow cytometry were confirmed to be highly enriched antigen-specific Foxp3-t- Treg cells (see, e.g., disclosure of Example 2, below), demonstrating that the ASTRA approach results in enriched, live Tregs specific for a stimulating antigen that can then be isolated by cell sorting or alternatively using magnetic enrichment.
  • GARP/LAP expression in conjunction with other activation-induced markers (e.g., CD137, 0X40, CD27, CD25) can be used to detect highly enriched antigen-specific Foxp3+ Tregs.
  • This Example describes the further development of the ASTRA approach that utilizes combinations of CD 137 and TORb complex components GARP and LAP as markers to isolate and characterize antigen specific Tregs.
  • Tregs regulatory' T cells
  • Tregs can be effectively characterized as expressing higher levels Foxp3, along with other Treg-associated markers, and can be sorted, cultured, and confirmed with epitope-specific tetramer staining.
  • antigen-specific Treg populations as opposed to bulk Treg populations, nuanced characteristics of regulator ' subsets can be ascertained which would otherwise be occluded by non-specific cellular contamination
  • Tregs Regulatory CD4+ T cells
  • MHCII major histocompatibility class II
  • CD 154 is unable to track Tregs m humans and should be used to remove Teffs from the Treg isolation. Instead these groups use CD 137 (4- IBB) as a marker for highly stable Foxp3+ Tregs.
  • CD137 4- IBB
  • tetramer can be used to validate the epitope-specific nature of the cells, as well as antigen-specific induction of TL-10 and re expression of relevant costimulatory molecules after resting.
  • mRNA sequencing also provides insight into the phenotype of these cells in comparison to other antigen-specific effector T cell subsets.
  • latent TGFf latent TGFf components GARP and LAP as markers of antigen specificity', as w'ell as costimulatory molecule CD 137 as a marker of demethylated TSDR in regulatory T cells, it is possible to enumerate and isolate highly enriched, Foxp3-stable, antigen-specific regulatory T cells from human patients.
  • These antigen-specific Tregs can be used as an immune-monitoring technique or as a therapeutic target for disease treatment.
  • Activation-induced regulatory T cells have high levels of de novo GARP/LAP expression
  • TNSFRSF protein family members of which CD137 and 0X40 are members of, were also expressed in highest abundance on these Tregs.
  • GARP and LAP both components of the latent TGFfi complex, GARP and LAP, were reliably coexpressed on CD 137+ CD 154- regulatory T cells (FIGURE 15). This allowed us to co-stain for them to increase fluorescence intensity during flow cytometry as well as enrichment efficiency.
  • CD137 and GARP/LAP+ Tregs In a polyclonal aCD3 aCD28 stimulation, the majority of the cells expressed G ARP/LAP and no significant differences were seen in the activation states of the CD137+ and GARP/LAP+ Tregs. We then determined that optimal CD137 and GARP/LAP expression occurs beginning at 16 hours of ex vivo stimulation with peptide antigen. PBMCs from a healthy patient were stimulated with influenza HAf peptide antigen for 0, 2, 6, 8, 12, 16 and 18 hours to determine optimal expression. Although Foxp3+ CD25+ Tregs begin to express slight levels of CD137 and GARP/LAP at 6 hours, expression plateaus at 12-16 hours. Similar trends can be seen when tracking percent expression within memory CD4+ T cells.
  • GARP/LAP+ Tregs after TCR-nonspecific polyclonal stimulation for 18 hours, we found that within the memory CD4 T cells (CD4+ CD45RA- ) GARP/LAP could be used in conjunction with multiple costimulatory molecules to enrich an extremely pure population of Foxp3+ regulatory T ceils. Using either CD137 (FIGURE 5A) or 0X40 (FIGURE 5B) in tandem with the latent TGFfl complex achieves a purity of close to 95% Foxp3+ CD25+ Tregs. GARP/LAP could also be faithfully used w th either CD25 or CD27 (FIGURES 5C and 5D) although these markers can be nonspecifically upregulated during stimulation.
  • CD137+ CD25+ regulator had a Foxp3 MFI significantly less than that seen in GARP/LAP+ CD137+ regulator )' T cells, indicating increased activation and functionality as Tregs (FIGURE 16).
  • the CD137+ CD154- compartment contains regulatory T cells with highly demethylated TSDR regions, but we contest that antigen-specificity is conferred by the expression of the latent TORb complex components GARP and LAP.
  • G ARP/LAP + regulatory T cells do not upregulate CD 154
  • Antigen-specific Tregs have been shown to be identifiable by a lack of CD154 expression, distinct from the effector subset, as well as expression of the costimulatory molecule CD137.
  • CD137 expression is markedly slower in upregulation after antigen stimulation than CD154, a phenomenon which has not been fully explored. This is effect is most likely due to the decreased proliferation of Tregs ex vivo as compared to Teffs, requiring different costimulatory molecules to maintain interaction of the TCR with the pMHC.
  • G ARP/LAP expression was limited to just regulator ⁇ ' T cells, as its expression has been tied to Foxp3 activation.
  • CD154- CD137- populations are shown m both antigen models to display background expression in non-activated memory T cells. Lack of GARP/LAP expression in the effector T cells allow' us to faithfully use it as a marker of antigen-specific regulatory T cells without the removal of effector T ceils that could potential] )' be contaminating the population through shared molecule expression.
  • GARP/LAP r regulatory T cells are transcriptionally distinct from regulatory T cells lacking surface-expressed GARP/LAP As CD 137, 0X40, and other TNFRSF family members also serve as activation- induced costimulatory molecules on effector T cells, we focused our efforts on determining the role GARP/LAP expression may have on driving Treg development and homeostasis.
  • TOEb has been well described as an essential cytokine for the induction and maintenance of regulatory T cell populations and is not a canonical costimulatory molecule like other TNFSRF family members.
  • GARP /LAP + upregulation denotes antigen-specificity among regulatory T cells
  • Tregs into cell lines based on GARP/LAP expression and assess their tetramer binding after two weeks of in vitro expansion.
  • Using a singular immunodominant epitope from the alder pollen antigen Ain g 1 we stimulated PBMCs from an HLA DBR1*1501 alder allergic patient and sorted CD 137+ Tregs for GARP/LAP expression.
  • GARP/LAP regulatory T cells are activated by natural exposure to antigen
  • Tregs While these markers serve well as indicators of Foxp3+ Tregs, they make no distinction between the entirety of Tregs observed in a patient and just those specific for an antigen of interest. Foxp3 is similar in that it detects all regulatory T cells within a patient, but not those specific to certain antigens. It also requires fixation and nuclear staining, dashing all chances of further analysis of the detected Tregs. By identifying the expression of activation-induced molecules that correspond only to antigen-specific regulator ⁇ T cells, we are able to effectively isolate only Tregs specific for an antigen of interest. We demonstrate the universality of the approach by detecting and characterizing these cells across a broad cohort of patients, including allergic, autoimmune, cancer, infectious disease, and healthy patients.
  • the assay requires no intracellular staining or Golgi -blocking treatments, as earlier versions of CD 154 assays required. There is no artificial stimulation required in the form of anti-CD28, as seen m recent CD137 assays. There is no need to remove effector T cells prior to regulatory T cell detection, as GARP/LAP is not expressed on effector T ceils, unlike CD137. Cells can be sorted or sequenced following detection because there is no necessity to stain for intracellular antigens. Tire single, positive- enrichment step allows for a simple assay that imposes no undue stress upon the cells by subjecting them to multiple enrichment steps.
  • GARP/LAP serves as an indicator of antigen-specificity in Tregs and denotes stable Foxp3 expression
  • CD137 and other TNFSRF costimulatory molecules like 0X40 are upregulated by highly TSDR demethyiated Tregs after activation in preparation for interaction with APCs, presumably to propagate the regulatory response as well as dampen signals to APCs in order to suppress reactivity in the environment.
  • Deregulation of certain costimulatory molecules during antigen stimulation can be affected by bystander effects due to cytokine signaling (IL-2, IL-7, !L-33) and other growth factors present in the stimulation milieu.
  • TNFR tumor necrosis factor receptor family
  • the advantageousness of the technique derives from the ability to target antigen-specific, rather than bulk populations. This decreases the possibility for nonspecific, off-target effects as seen in checkpoint blockade therapies, while increasing the potency of the response, ultimately requiring less cells due to the increased specificity.
  • vriro-induced Tregs iTregs
  • iTregs possibly fail to correct autoimmune reactions, not because they lack the density to occupy the compartment, but most likely because they fail to properly engage with presented autoantigens and induce contact-mediated suppression, as well as become properly activated via TCR to cany out suppressive tasks.
  • There are also risks with iTregs converting back into more pathogenic phenotypes due to decreased Foxp3 stability.
  • PBMCs Blood draws from patients were arranged by the Benaroya Research Institute clinical core in conjunction with the Virginia Mason Hospital and Medical Center. Blood arrived within 24 hours of draw and w3 ⁇ 4s stored in the dark at room temperature. PBMCs were then isolated from whole blood using ficoll separation. The ficoll preparation was centrifuged at 1800 RPM for 20 minutes and the bully coat was gently removed using a transfer pipet. Red blood cells were lysed using an in-house hemolytic buffer after 10 minutes of incubation at room temperature. Cells were then washed and counted. PBMCs were first incubated with anti CD40 at a concentration of 1 uL per 10 6 cells.
  • Flow cytometry experiments consisted of twelve-color to sixteen-color panels. We acquired data on an LSRFortessa flow cytometer for analysis (BD Biosciences), and analyzed it in FlowJo version 10.0.7 (Tree Star). We performed cell-sorting experiments on a FACSAria Fusion cytometer (BD Biosciences). We collected at least 150,000 events for antigen-specific assays.
  • samples of 100 CD154+, CD137+, or GARP/LAP+ CD4+ memory T-cells were sorted. 10 samples from 10 unique donors were prepared (both pre and post influenza vaccination). Samples were sorted into SMARTer v4 lysis reagents (Clontech). Cells were lysed and cDNA was synthesized. After amplification, sequencing libraries w3 ⁇ 4re prepared using the Nextera XT DNA Library Preparation Kit (Illumina).
  • Single-read sequencing of libraries was performed on either on a HiScanSQ with 100-base pair long reads (5 Cl preparations) or a HiSeq2500 sequencer (Illumina, 12 Cl preparations and all bulk RNA-seq data with 58-base pair long reads, using TruSeq v3 Cluster and SBS kits (Illumina) with a target depth of >2.5M reads.
  • Reads were aligned to the UCSC Human genome assembly version 19 in Galaxy using bowtie and TopHat (Tophat for Illumina tool, v.1.5.) (Trapnell, C., et al, TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1 105-1111 (2009).). Read counts per Ensemhl gene ID were estimated in Galaxy using htseq-count (htseq-count tool, n.0.4.1 ) (Anders, S., et al., HTSeq-a Python framework to work with high-throughput sequencing data. J Bioinformatics 31, 166- 169 (2015)).
  • Capture antibodies include TL-10, ITNg, and TI.,-4 from ELISpot kits (U-CyTecb Biosciences). Stimulated cells were incubated at 37 °C for another 24 h and then washed and developed.
  • This Example describes application of the disclosed ASTRA approach to profile and characterize T cells that are specific for tumor or auto-antigens in an attempt to better study T cell signatures in a relevant context, i.e., during cancer treatment.
  • a relevant application of the ASTRA method disclosed herein is to elucidate the relationship between tumor- and auto-antigen-specific T cells in response to immune checkpoint inhibitor (ICI) therapy.
  • ICI immune checkpoint inhibitor
  • irAE immune-related adverse events
  • Measuring antigen-specific T cell responses, which are proximal pharmacodynamics markers of immune responses, will likely clarify the relationship between anti-tumor and autoimmune responses, and will enable better management of these events clinically during ICI.
  • the ASTRA assay disclosed herein is applied to identify, enumerate and characterize tumor- and auto- antigen-specific T cells m parallel. In a subset of subjects, these cells are isolated using the same approach and then perform low input bulk RNA- seq to identify antigen -sped lie T cell signatures and determine how they change in response to ICI therapy.
  • samples are obtained using our "Immune Checkpoint Inhibitor (ICI) Cancer Research Protocol" (ICICR).
  • ICI Immuno Checkpoint Inhibitor
  • the samples tested are from visits before and 1-3 months after initiation of CPI therapy. Testing both pre-and post-treatment visits enables better understanding of the relationship of antigen-specific T cell signatures to therapy.
  • 30 Million PBMCs are collected from each subject and each time point to run all experiments. Frozen PBMC are used for all assays, allowing performance of assays for each subject's samples on the same day. It is proposed to study patients with and without irAE in order to determine if the responses m individuals with irAE differ from those without, or by type of irAE. Prior therapy of cancer patients also has the potential to confound investigations, therefore this variable is considered when selecting subjects and during analyses.
  • the ASTRA assay as described herein is used to monitor the ex vivo frequency, surface markers and phenotypes of tumor-specific T cells in samples from patients prior to and after ICI therapy.
  • a major advantage of this assay is that multiple possible peptide epitopes can be pooled, and then analyzed in side-by-side assays. For example, we have successfully experimented with peptide pools comprising up to -1,000 different peptides.
  • TAA tumor associated antigens
  • neoantigens i.e., created by tumor-specific mutations
  • enriched cells are characterized for a variety of markers, for example, using markers of maturation/differentiation (CD4, CD27, CD45RB, CD45RA), exhaustion (TIM-3, LAG-3, PD-1, CD49b), activation (CD38, CD69, ICOS), and horning (CCR4, CXCR3, CCR6).
  • markers of maturation/differentiation CD4, CD27, CD45RB, CD45RA
  • exhaustion TIM-3, LAG-3, PD-1, CD49b
  • activation CD38, CD69, ICOS
  • Co-enriched antigen-specific CD4+ Treg cells and CD8+ Teff cells can be specifically characterized using markers of maturation/differentiation (CD4, CD27, CD45RA), exhaustion (TIM-3, LAG-3, PD-1, CD49b, KLRGI , CD 160) and activation (CD38, CD25, CD69, OX-40).
  • Anti-CD 14, CD 19 and Via-Probe reagent can be used as a dump gate.
  • This multi-parameter immune profiling can be carried out with a FACS Fusion flow cytometer, such that cells of interest can be simultaneously sorted for subsequent use in transcript profiling.
  • the assay was performed with tumor antigens and T cells from a healthy control individual and a cancer patient before and after ICI therapy using CD137 and GARP as markers for antigen-specific activation of Treg cells. See FIGURE 19A.
  • CD137 and GARP markers for antigen-specific activation of Treg cells.
  • FIGURE 19A is the massive tumor-antigen specific CD8+ T cell response after ICI therapy.
  • the antigen-specific results were pronounced of the global increase in CD8+ T cells observed by others following ICI therapy.
  • the self-antigen pool therefore includes overlapping peptides derived from insulin, IGRJP, GAD65, thyroxin, thyroglobulin, vimentin, alpha enolase, desmoglein 3, and tyrosinase.
  • An example of the assay with self- antigens and T cells from a healthy control individual and a cancer patient before and after ICI therapy is shown in FIGURE 19B. Strikingly, there was a massive increase in CD8+ Teff cells reactive with self-antigens (islet antigens only, in this case) m cells in the cancer patient after ICI-therapy, in parallel with the tumor-antigen specific CD8+ T ceils. Together, these results suggest that this patient had both anti-tumor and anti-islet CDS T cell responses triggered by ICI therapy. It is noteworthy that autoimmune diabetes can be triggered by anti -PD- 1 ICI therapy.
  • the preliminary results disclosed herein demonstrate the utility of the disclosed ASTRA approach to assaying activated, antigen-specific Treg cells in a wide variety of contexts. Specifically, the results show that antigen-specific Treg cells can be detected and monitored in the context of a cancer treatment to determine the effects on immune- related adverse events connected with the cancer treatment.
  • Embodiment 1 A method of detecting an activated regulator' T (Treg) cell, comprising:
  • Treg regulatory T
  • Embodiment 2 The method of embodiment 1, wherein the Treg cells have been exposed to antigen presenting cells that display a peptide antigen in a peptide/MHC complex.
  • Embodiment 3 The method of embodiment 1, wherein the Treg cells have been exposed to a soluble peptide-MHC monomer or mul timer complex.
  • Embodiment 4 The method of embodiment 1 , 2, or 3, wherein the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected from CD137, 0X40 (CD 134), CD27, CD69, ICOS (CD278), CDS, CD49d, CD2, and CD25.
  • Embodiment 5. The method of one of embodiments 1-4, wherein the latent TOEb complex protein is GARP (LRRC32), LAP (latent-associated peptide), TGF[> I -4. or a complex of any thereof.
  • Embodiment 6 The method of one of embodiments 1-5, wherein the eo-stimulatory marker is selected from CD 137, 0X40 (CD 134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • the eo-stimulatory marker is selected from CD 137, 0X40 (CD 134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • Embodiment ? The method of one of embodiments 1-6, wherein the method further comprises contacting the sample with a third detection molecule that specifically binds to CD4, and wherein detecting an activated T regulatory cell comprises detecting a cell in the sample that is bound by the first detection molecule, the second detection molecule, and the third detection molecule.
  • Embodiment s The method of one of embodiments 1-7, wherein one or more of the first detection molecule, the second detection molecule, and the third detection molecule is an antibody, antibody-like molecule, aptamer, or a functional antigen-binding fragment or domain thereof.
  • Embodiment 9 The method of embodiment 8, wherein the antibody-like molecule is a single-chain antibody, a bispecific antibody, a Fab fragment, or a F(ah) 2 fragment.
  • Embodiment 10 The method of embodiment 9, wherein the single-chain antibody is a single chain variable fragment (scFv), single-chain Fab fragment (scFab), VJJH fragment, V N AR, or nanobody.
  • scFv single chain variable fragment
  • scFab single-chain Fab fragment
  • VJJH fragment V N AR, or nanobody.
  • Embodiment 11 The method of one of embodiments 1 -10, wherein each of the first detection molecule and the second detection molecule is detectably labeled with mutually distinguishable labels.
  • Embodiment 12 The method of one of embodiments 1-11, wherein each of the first detection molecule, the second detection molecule, and the third detection molecule is detectably labeled with mutually distinguishable labels.
  • Embodiment 13 The method of one of embodiments 1-12, wherein detecting binding of the first detection molecule to the cell, binding of the second detection molecule to the cell, and/or binding of the third detection molecule to the cell comprises use of fluorescence-activated cell sorting (FACS) or mass cytometr ' (CyTOF).
  • FACS fluorescence-activated cell sorting
  • CDTOF mass cytometr '
  • Embodiment 14 The method of one of embodiments 1-13, further comprising enriching for the activated Treg cell.
  • Embodiment 15 The method of one of embodiments 1 -14, further comprising isolating the activated Treg cell.
  • Embodiment 16 The method of one of embodiments 1 -15, wherein the sample is a biological sample from a subject, such as blood, tissue, lavage, tumor, or is derived therefrom.
  • Embodiment 17 The method of one of embodiments 1 -16, further comprising an initial step of obtaining the sample of Treg cells from a subject.
  • Embodiment 18 The method of one of embodiments 1-17, wherein obtaining a sample comprises a step of contacting an initial sample comprising Treg cells with sufficient antigen or activator in a manner and amount such that at least a portion of the Treg cells are activated.
  • Embodiment 19 The method of embodiment 18, comprising contacting the cells with the activator.
  • Embodiment 20 The method one of embodiments 18-19, wherein the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected CD137, 0X40 (CD 134), CD27, CD69, !CQS (CD278), CDS, CD49d, CD2, and CD25.
  • the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected CD137, 0X40 (CD 134), CD27, CD69, !CQS (CD278), CDS, CD49d, CD2, and CD25.
  • Embodiment 21 A method of producing an enriched population of activated T regulatory' (Treg) cells, comprising:
  • Treg regulatory T
  • Embodiment 22 The method of embodiment 21 , wherein the antigen of interest is a peptide that is complexed with MHC on an antigen presenting cell.
  • Embodiment 23 The method of embodiment 21, wherein the antigen of interest is a peptide complexed with a MHC monomer or multimer to form a soluble complex.
  • Embodiment 24 The method of one of embodiments 21-23, wherein the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected from CD137, 0X40 (CD 134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and (1)25
  • Embodiment 25 The method of one of embodiments 21-24, wherein the latent TORb complex protein is GARP (LRRC32), LAP (latent-associated peptide), TGFf)I-4, or a complex of any thereof.
  • GARP LRRC32
  • LAP latent-associated peptide
  • TGFfI-4 TGFfI-4
  • Embodiment 26 The method of one of embodiments 21-25, wherein the costimulatory marker is selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • CD137, 0X40 CD134
  • CD27, CD69, ICOS CD278, CD3, CD49d, CD2, and CD25.
  • Embodiment 27 The method of one of embodiments 21-26, wherein the method further comprises contacting the sample with a third enrichment molecule that specifically binds to CD4, and enriching for cells that are bound by the first enrichment molecule, the second enrichment molecule, and the third enrichment molecule.
  • Embodiment 28 The method of one of embodiments 21-27, wherein one or more of the first enrichment molecule, the second enrichment molecule, and the third enrichment molecule is an antibody, antibody-like molecule, aptamer, or a functional antigen-binding fragment or domain thereof.
  • Embodiment 29 The method of embodiment 28, wherein the antibody -like molecule is a single-chain antibody, a bispecific antibody, a Fab fragment, or a Fiabjy fragment.
  • Embodiment 30 The method of embodiment 29, wherein the single-chain antibody is a single chain variable fragment (scFv), single-chain Fab fragment (scFab), V H H fragment, V ⁇ AR, or nanobody.
  • scFv single chain variable fragment
  • scFab single-chain Fab fragment
  • V H H fragment V ⁇ AR, or nanobody.
  • Embodiment 31 The method of one of embodiments 21 -30, wherein the first enrichment molecule and the second enrichment molecule are each detectably labeled with mutually distinguishable labels.
  • Embodiment 32 The method of one of embodiments 27-31 , wherein the first enrichment molecule, the second enrichment molecule, and the third enrichment molecule are each detectably labeled with mutually distinguishable labels.
  • Embodiment 33 The method of one of embodiments 21 -32, wherein the enriching step comprises detecting binding of the first enrichment molecule to the cell and binding of the second enrichment molecule to the cell, or detecting binding of the first enrichment molecule to the cell, binding of the second enrichment molecule to the cell, and binding of the third enrichment molecule to the cell, using fluorescence- activated cell sorting (FACS).
  • FACS fluorescence- activated cell sorting
  • Embodiment 34 The method of one of embodiments 21 -33, further comprising an initial step of obtaining the sample of Treg cells from a subject.
  • Embodiment 35 The method of one of embodiments 21-34, wherein the sample is a biological sample from a subject, such as blood, tissue, lavage, tumor, or is derived therefrom.
  • Embodiment 36 The method of one of embodiments 21-35, comprising incubating the sample with the activator.
  • Embodiment 37 The method of embodiment 36, wherein the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected from CD137, 0X40 (CD134), CD27, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • the activator is a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected from CD137, 0X40 (CD134), CD27, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • Embodiment 38 The method of one of embodiments 21-37, further comprising administering one or more activated Treg cells of the enriched population to a subject in need thereof.
  • Embodiment 39 The method of embodiment 38, wherein the subject is the same subject from whom the sample was obtained or derived, or is HLA matched to the subject from whom the sample was obtained or derived.
  • Embodiment 40 A cell m the enriched population produced from the method of any one of embodiments 21-37.
  • Embodiment 41 A method of treating a condition treatable by the presence of an activated Treg, comprising administering the cell of embodiment 40 to a subject in need thereof.
  • Embodiment 42 The method of embodiment 41, wherein the condition is an autoimmune disease or an allergy.
  • Embodiment 43 A method for monitoring a T regulator' (Treg) cell response to potential exposure of a subject to an antigen, comprising:
  • Embodiment 44 The method of embodiment 43, wherein the latent TORb complex protein is GARP (LRRC32), LAP (latent-associated peptide), TGFpl-4, or a complex of any thereof.
  • GARP LRRC32
  • LAP latent-associated peptide
  • TGFpl-4 TGFpl-4
  • Embodiment 45 The method of embodiment 43 or 44, wherein the costimulatory marker is selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • CD137, 0X40 CD134
  • CD27, CD69, ICOS CD278, CD3, CD49d, CD2, and CD25.
  • Embodiment 46 A method for monitoring the sensitivity of a subject to an antigen of interest, comprising:
  • Treg T regulatory
  • Embodiment 47 The method of embodiment 46, wherein the latent TOEb complex protein is GARP (LRRC32), LAP (latent-associated peptide), TORb I -4. or a complex of any thereof
  • Embodiment 48 The method of embodiment 46 or 47, wherein the costimulatory marker is selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • CD137, 0X40 CD134
  • CD27, CD69, ICOS CD278, CD3, CD49d, CD2, and CD25.
  • Embodiment 49 The method of one of embodiments 46-48, wherein a relative increase in the number of cells m the sample that are bound by both the first detection molecule and the second detection molecules compared to the established threshold indicates a low sensitivity of the subject to the antigen of interest.
  • Embodiment 50 The method of one of embodiments 46-49, wherein the threshold is established at an earlier time point from the subject using the same method, and wherein a relative increase in cells that are bound by both the first detection molecule and the second detection molecule over the established threshold indicates a decreasing sensitivity to the antigen of interest, and wherein a relative decrease in cells that are bound by both the first detection molecule and the second detection molecule over the established threshold indicates an increasing sensitivity to the antigen of interest.
  • Embodiment S l The method of embodiment 50, wherein the threshold is established during or prior to administration of the therapeutic treatment and the sensitivity of the subject to the antigen of interest at a later time point is indicative of the efficacy of the therapeutic treatment.
  • Embodiment 52 A method of screening for T regulator) ' (Treg) cell stimulatory epitopes from an allergen or antigen of interest, comprising:
  • Treg cells exposing the Treg cells to an epitope derived from an antigen of interest in an MHC context
  • Treg cells contacting the Treg cells with a first detection molecule that specifically binds to a latent TGF[3 complex protein and a second detection molecule that specifically binds to a eo-stimulatory marker;
  • Embodiment 53 The method of embodiment 52, wherein exposing the Treg cells to an epitope comprises exposing the Treg cells to antigen presenting cells that display the epitope m a peptide/MHC complex.
  • Embodiment 54 The method of embodiment 52, wherein exposing the Treg cells to an epitope comprises exposing the Treg cells to soluble peptide-MHC monomer or multimer complexes that comprise the epitope loaded onto the MHC monomer or multimer in a complex.
  • Embodiment 55 The method of one of embodiments 52-54, further comprising exposing the Treg ceils to a composition comprising one or more costimulatory molecules that bind to and activate one or more co-stimulatory receptors selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • Embodiment 56 The method of one of embodiments 52-55, wherein the latent TGFB complex protein is GARP (LRRC32), LAP (latent-associated peptide), TGFjll -4, or a complex of any thereof.
  • Embodiment 57 The method of one of embodiments 52-56, wherein the costimulatory marker is selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • Embodiment 58 The method of one of embodiments 52-57, further comprising repeating the method for one or more different epitopes derived from the same antigen of interest to determine the epitopes that stimulate the relatively increased levels of activated Treg cells.
  • Embodiment 59 A method of identifying the MHC Class 11 molecule that binds to a Treg stimulatory epitope, comprising:
  • Treg cells exposing the Treg cells to an epitope derived from an antigen of interest in an MHC context
  • Treg cells contacting the Treg cells with a first detection molecule that specifically binds to a latent TORb complex protein and a second detection molecule that specifically binds to a co-stimulatory marker;
  • Embodiment 60 The method of embodiment 59, wherein exposing the Treg cells to an epitope comprises exposing the Treg cells to an antigen presenting cell that displays the epitope in a peptide/MHC complex.
  • Embodiment 61 The method of embodiment 60 or 61, wherein the method further comprises isolating the antigen presenting cell that produced a high abundance of ceils bound by both the first detection molecule and the second detection molecule;
  • Embodiment 62 The method of one of embodiments 59-61, wherein exposing the Treg cells to an epitope comprises exposing the Treg cells to soluble peptide-MHC monomer or mul timer complexes that comprise the epitope loaded onto the MHC monomer or multimer in a complex.
  • Embodiment 63 The method of one of embodiments 59-62, further comprising exposing the Treg cells to a composition comprising one or more costimulatory molecules that bind to and activate one or more eo-stimulatory receptors selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CDS, CD49d, CD2, and CD25
  • a composition comprising one or more costimulatory molecules that bind to and activate one or more eo-stimulatory receptors selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CDS, CD49d, CD2, and CD25
  • Embodiment 64 The method of one of embodiments 59-63, wherein the latent T(3Rb complex protein is GARP (LRRC32), LAP (latent-associated peptide), TGFfd -4, or a complex of any thereof.
  • the latent T(3Rb complex protein is GARP (LRRC32), LAP (latent-associated peptide), TGFfd -4, or a complex of any thereof.
  • Embodiment 65 The method of one of embodiments 59-64, wherein the costimulatory molecule is selected from CD137, 0X40 (CD134), CD27, CD69, ICOS (CD278), CD3, CD49d, CD2, and CD25.
  • Embodiment 66 A method of testing the effect of a putative therapeutic compound on an activated T regulatory (Treg) cell, comprising exposing the cell of embodiment 40 to the therapeutic compound.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des procédés, des réactifs et des kits de détection, d'enrichissement et d'utilisation de lymphocytes T régulateurs (Treg) spécifiques d'un antigène. Les procédés mettent en contact des lymphocytes T exposés à un antigène d'intérêt (par exemple, par l'intermédiaire du CMH sur une cellule présentatrice d'antigènes ou le complexe CMH préchargé), avec des molécules de détection/d'enrichissement qui se lient spécifiquement à une protéine complexe ΤGFβ latente et à une molécule co-stimulatrice. Dans certains modes de réalisation, la protéine complexe ΤGFβ latente peut être GARP (LRRC32), LAP (peptide associé latent), TGFβ1-4, ou un complexe de l'un quelconque de ceux-ci. Dans certains modes de réalisation, la molécule co-stimulatrice peut être CD137 (4 1-BB), OX40, CD69, ICOS, CD25, ou CD27. Les procédés selon l'invention sont applicables pour surveiller la production de Treg activés pour des antigènes d'intérêt, pour surveiller la sensibilité d'un sujet à un antigène d'intérêt, pour cribler des antigènes stimulateurs de Treg et/ou le CMH qui favorisent le développement de Treg, et enrichir et potentiellement administrer des Treg activés pour le traitement d'une maladie.
PCT/US2018/061462 2017-11-16 2018-11-16 Dosage de lymphocytes t régulateurs spécifiques d'un antigène WO2019099788A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762587346P 2017-11-16 2017-11-16
US62/587,346 2017-11-16

Publications (1)

Publication Number Publication Date
WO2019099788A1 true WO2019099788A1 (fr) 2019-05-23

Family

ID=66539140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/061462 WO2019099788A1 (fr) 2017-11-16 2018-11-16 Dosage de lymphocytes t régulateurs spécifiques d'un antigène

Country Status (1)

Country Link
WO (1) WO2019099788A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117607223A (zh) * 2024-01-22 2024-02-27 南昌航空大学 一种基于整体柱富集分离的自驱动微流控系统

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120141537A1 (en) * 2010-11-18 2012-06-07 Pease Larry R Enhancing t cell activation using altered mhc-peptide ligands
US20150004176A1 (en) * 2011-07-01 2015-01-01 Beckman Coulter, Inc. Regulatory t cells and methods of identifying, obtaining and using to treat immuno-based disorders
US20160045594A1 (en) * 2013-03-27 2016-02-18 Fred Hutchinson Cancer Research Center Directed immune stimulation
US20170014474A1 (en) * 2007-02-01 2017-01-19 Tcf Gmbh Screening method for the identification of agents capable of activating cd4+cd25+ regulatory t-cells through interactions with the hiv-1 gp120 binding site on cd4
US20170021001A1 (en) * 2014-04-01 2017-01-26 INSERM (Institut National de la Santé et de la Recherche Médicale) An isolated donor mhc-derived peptide and uses thereof
US20170102386A1 (en) * 2009-09-10 2017-04-13 Miltenyi Biotech Gmbh Method for identifying and obtaining activated T-cells that have a demethylated Foxp3 TSDR region

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170014474A1 (en) * 2007-02-01 2017-01-19 Tcf Gmbh Screening method for the identification of agents capable of activating cd4+cd25+ regulatory t-cells through interactions with the hiv-1 gp120 binding site on cd4
US20170102386A1 (en) * 2009-09-10 2017-04-13 Miltenyi Biotech Gmbh Method for identifying and obtaining activated T-cells that have a demethylated Foxp3 TSDR region
US20120141537A1 (en) * 2010-11-18 2012-06-07 Pease Larry R Enhancing t cell activation using altered mhc-peptide ligands
US20150004176A1 (en) * 2011-07-01 2015-01-01 Beckman Coulter, Inc. Regulatory t cells and methods of identifying, obtaining and using to treat immuno-based disorders
US20160045594A1 (en) * 2013-03-27 2016-02-18 Fred Hutchinson Cancer Research Center Directed immune stimulation
US20170021001A1 (en) * 2014-04-01 2017-01-26 INSERM (Institut National de la Santé et de la Recherche Médicale) An isolated donor mhc-derived peptide and uses thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117607223A (zh) * 2024-01-22 2024-02-27 南昌航空大学 一种基于整体柱富集分离的自驱动微流控系统
CN117607223B (zh) * 2024-01-22 2024-04-09 南昌航空大学 一种基于整体柱富集分离的自驱动微流控系统

Similar Documents

Publication Publication Date Title
US20230280341A1 (en) Cd127 expression inversely correlates with foxp3 and suppressive function of cd4+ tregs
JP4714767B2 (ja) ヒト血液由来のcd4+cd25+調節t細胞
US8975069B2 (en) Method for identifying antigen-specific regulatory T cells
Picarda et al. Transient antibody targeting of CD45RC induces transplant tolerance and potent antigen-specific regulatory T cells
US11022615B2 (en) Regulatory T-cells, method for their isolation and uses
Germar et al. Generation and characterization of anti–citrullinated protein antibody–producing B cell clones from rheumatoid arthritis patients
US20220155321A1 (en) Selection of t cell receptors
Hisada et al. Circulating plasmablasts contribute to antiphospholipid antibody production, associated with type I interferon upregulation
Chow et al. Assessment of CD4+ T cell responses to glutamic acid decarboxylase 65 using DQ8 tetramers reveals a pathogenic role of GAD65 121–140 and GAD65 250–266 in T1D development
Karahan et al. Polyclonal B cell activation for accurate analysis of pre-existing antigen-specific memory B cells
IL291859A (en) A high-throughput method for screening t-cell cognate and epitope reactivity in primary human cells
Wilfahrt et al. Histone deacetylase 3 represses cholesterol efflux during CD4+ T-cell activation
US20130052642A1 (en) Method for the identification and purification of human naturally occurring regulatory t cells (ntregs)
WO2019099788A1 (fr) Dosage de lymphocytes t régulateurs spécifiques d'un antigène
Dufaud et al. PD-1 checkpoint blockade disrupts CD4 T cell regulated adaptive B cell tolerance to foreign antigens
Reimer et al. Isolation of transcriptionally active umbilical cord blood‐derived basophils expressing FcɛRI, HLA‐DR and CD203c
Clarkson et al. Preservation of antigen-specific responses in cryopreserved CD4+ and CD8+ T cells expanded with IL-2 and IL-7
Sérazin et al. Single cell analysis reveals CD45RClow/-TNFR2+ CD29lowCD8+ Tregs with superior activity
US20090075296A1 (en) Methods and compositions for assaying regulatory t cells
Valma Antigen Specific CD4+ T cells and their role in viral infection and autoimmune disease
Smith et al. Identification of antigen-specific TCR sequences using a strategy based on biological and statistical enrichment in unselected subjects
Janowska Regulation of human early B lymphopoiesis
Rezk Interactions between B cells and CD8+ T cells in multiple sclerosis
Tuttle TCR signaling affects the development of thymic NKT cell subsets
Rathbone Investigation of the adaptive immune response in multiple sclerosis

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: 18879944

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18879944

Country of ref document: EP

Kind code of ref document: A1