WO2018024895A1 - Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells - Google Patents

Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells Download PDF

Info

Publication number
WO2018024895A1
WO2018024895A1 PCT/EP2017/069824 EP2017069824W WO2018024895A1 WO 2018024895 A1 WO2018024895 A1 WO 2018024895A1 EP 2017069824 W EP2017069824 W EP 2017069824W WO 2018024895 A1 WO2018024895 A1 WO 2018024895A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
foxp3
regulatory
population
invariant
Prior art date
Application number
PCT/EP2017/069824
Other languages
French (fr)
Inventor
Hélène LE BUANEC
Armand Bensussan
Valérie SCHIAVON
Daniel Zagury
Sophie Duchez
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Medecine Et Innovation
Université Paris Diderot - Paris 7
Centre National De La Recherche Scientifique (Cnrs)
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Medecine Et Innovation, Université Paris Diderot - Paris 7, Centre National De La Recherche Scientifique (Cnrs) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Priority to EP17752074.9A priority Critical patent/EP3493833A1/en
Priority to US16/322,188 priority patent/US20190167791A1/en
Publication of WO2018024895A1 publication Critical patent/WO2018024895A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • 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
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • 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
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0637Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/49Breast
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/02Compounds of the arachidonic acid pathway, e.g. prostaglandins, leukotrienes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/04Immunosuppressors, e.g. cyclosporin, tacrolimus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2315Interleukin-15 (IL-15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/999Small molecules not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/99Coculture with; Conditioned medium produced by genetically modified cells

Definitions

  • the present invention relates to therapeutic uses of ex vivo generated Foxp3 + regulatory T cells.
  • the focal point for cancer treatment is using a combination of conventional therapies: chemotherapy, radiotherapy and surgery. In most cases, they are effective in treating primary tumors; however, they inefficiently prevent metastasis through disseminated tumor cells.
  • immunotherapy has emerged as an alternative therapeutic strategy for the treatment of cancer, largely due to the clinical development of novel agents including cytokines, monoclonal antibodies (mAbs), genetically engineered chimeric antigen receptor and immune checkpoint blockade inhibitors.
  • Immune checkpoint therapy based on the passive administration of mAbs blocking negative regulators of the activation of effector T cells such as cytotoxic T lymphocyte- associated antigen 4 (CTLA-4) (Ipilimumab) and programmed death- 1 (PD1) (Pembrolizumab and Nivolumab), have indeed achieved durable responses in some patients, including long-term remissions with no clinical signs of cancer.
  • CTL-4 cytotoxic T lymphocyte- associated antigen 4
  • PD1 programmed death- 1
  • interruption of immune checkpoints with mAbs is commonly associated with immune mediated toxicities such as auto-immune sequelae and inflammatory damage to normal parenchyma.
  • Ipilimumab The most common damages related to Ipilimumab, include rash/pruritus, enterocolitis, uveitis, pancreatitis, hypophysisitis and leucopenia. These adverse events could be anticipated because of the systemic activity of the administered mAbs, targeting thus all tissues instead of acting locally.
  • Anti-cancer vaccines are designed to elicit an immune response against tumor antigens or cells allowing a protection against tumor recurrence or metastatic disease.
  • TAA tumour-associated
  • TSA tumour-specific
  • TAA expression is a function of inappropriate production of embryonic gene-encoded oncofetal antigens or overexpression of normal protein
  • TSA are either new antigens induced mostly by chemical carcinogens and sometimes by virus or viral antigens.
  • whole tumor cell-based vaccines have been developed.
  • the advantage of using whole tumor cells as immunogen is that the cells provide a source of several TAAs or TSAs, some identified and some as yet undefined.
  • These anti-cancer vaccines used as active principle 1) whole tumor cells or genetically modified tumor cell lines to express cytokines, chemokines or costimulatory molecules to enhance their immunogenicity, 2) cellular lysates or 3) immunogenic apoptotic bodies derived from tumor cells.
  • immunization with cancer cell- based vaccines failed to induce long-lasting anti-tumor responses and has thus not resulted in significant long-term therapeutic benefits.
  • anti-CTLA-4 or anti-PD-1 mAbs also eliminate regulatory T cells (Tregs) present in tumor microenvironment.
  • Tregs regulatory T cells
  • Treg express surface molecules that can be specifically targeted by antibodies (Abs) or pharmacologic inhibitors.
  • Abs for example, CD25, TGF ⁇ pathway, CTLA-4, PD-1, CD73, CD390... are targeted for Treg silencing either by Treg removal or impairment of Treg suppressor functions.
  • Abs doclizumab: anti-CD25 Ab
  • IL-2 fusion toxins such as denileukin diftitox (Ontak)
  • drugs such as cyclophosphamide or tyrosine kinase inhibitors (sunitinib)
  • unitinib tyrosine kinase inhibitors
  • the present invention concerns the development of a vaccine directed against the immune regulatory T cells specifically activated by cancer cells.
  • the stroma of malignant tumors is indeed comprised of TILs (Tumor-infiltrating lymphocytes) known 1) to be highly enriched on regulatory T cells and 2) to exert an immune suppressive activity, which likely accounts for the local cancer immune escape.
  • the inventors thus suggest using a vaccine composition comprising inactivated Foxp3 + regulatory T cells as active principle.
  • the inactivated Foxp3 + regulatory T cells may represent an antigenic target to induce an immune response directed against the Foxp3 + regulatory T cells present in the TILs, thereby preventing their immune suppressive activity and allowing the cytotoxic activity of effector cells such as NK cells against the tumor cells.
  • the TILs may comprise one or more different Foxp3 + regulatory T cells populations
  • the inventors suggest determining the one or more Foxp3 + regulatory T cells population overrepresented in the TILs, to ex vivo generate and expand the corresponding Foxp3 + regulatory T cells populations and thus prepare a treatment adapted to the tumor to be treated.
  • the present invention relates to an immunogenic product comprising at least one inactivated ex vivo generated Foxp3 + regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population.
  • Another object of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising at least one inactivated ex vivo generated regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient.
  • a further object of the present invention relates to a vaccine composition comprising at least one inactivated ex vivo generated regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population and at least one adjuvant.
  • the present invention also relates to a pharmaceutical composition comprising at least one ex vivo generated regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated regulatory T cells population remains stable when placed in inflammatory condition.
  • the immunogenic product, pharmaceutical composition or vaccine composition according to the invention is for use in treating cancer.
  • the present invention further relates to a method for preparing the immunogenic product, pharmaceutical composition or vaccine composition according to the invention, comprising: identifying from a tumor sample obtained from the subject the at least one regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population, that is overrepresented, ex vivo generating the at least one overrepresented regulatory T cells population, inactivating the at least one ex vivo generated regulatory T cell population.
  • Another object of the present invention is a pharmaceutical composition according to the invention for use in cell therapy.
  • a further object of the present invention is a pharmaceutical composition according to the invention for use in treating inflammatory or autoimmune diseases or for preventing transplant rejection or graft versus host disease (GVHD).
  • GVHD transplant rejection or graft versus host disease
  • the at least one ex vivo generated regulatory T cells population of the immunogenic product, pharmaceutical composition or vaccine composition according to the invention is obtained by a method comprising: for the MHCII restricted CD4 + Foxp3 + regulatory T cells population: culturing CD3 + CD4 + CD25 " T cells in the presence of a TCRa- ⁇ cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the ⁇ Foxp3 + regulatory T cells population: culturing CD3 + TCR ⁇ + T cells in the presence of a ⁇ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphat
  • the TCRa- ⁇ cell activator is a polyclonal TCRa- ⁇ cell activator, preferably an anti-CD3 antibody or an anti-TCRaP antibody;
  • the ⁇ T cell activator is a polyclonal ⁇ T cell activator, preferably an anti-TCR ⁇ antibody or a non peptide phosphoantigen;
  • the invariant T cell activator is a polyclonal invariant T cell activator, preferably a Va24 activator.
  • the TCRa- ⁇ cell activator is an antigen-specific TCRa- ⁇ cell activator, preferably tolerogenic dendritic cells (DCs) and pulsed with at least one self- peptide antigen;
  • the ⁇ T cell activator is an antigen-specific ⁇ T cell activator, preferably tolerogenic dendritic cells (DCs) and pulsed with at least one bisphosphonate, preferably at least one aminobiphosphonate and
  • the invariant T cell activator is an antigen-specific invariant T cell activator, preferably tolerogenic dendritic cells (DCs) expressing CDl and pulsed with at least one non peptide lipid antigen.
  • the cAMP activator is selected from the group comprising prostaglandin E2 (PGE2), an EP2 or EP4 agonist, a membrane adenine cyclase activator or a metabotropic glutamate receptors agonist.
  • PGE2 prostaglandin E2
  • EP2 or EP4 agonist an EP2 or EP4 agonist
  • membrane adenine cyclase activator or a metabotropic glutamate receptors agonist.
  • the TGF pathway activator is selected from the group comprising TGF , bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin and nodal .
  • BMPs bone morphogenetic proteins
  • GDFs growth and differentiation factors
  • AH anti-mullerian hormone
  • the mTOR inhibitor is selected from the group comprising rapamycin, rapamycin analogs, wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG), curcumin, resveratrol; genistein, 3, 3- diindolylmethane (DIM), LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l-benzopyran-4- one), PP242, PP30, Torinl, Ku-0063794, WAY-600, WYE-687, WYE-354, GNE477, NVP-BEZ235, PI- 103, XL765 and WJD008.
  • the at least one ex vivo generated Foxp3 + regulatory T cells population are expanded by a method comprising: - for the MHCII restricted CD4 + Foxp3 + regulatory T cells population: culturing
  • invariant Foxp3 + regulatory T cells population culturing CD3 + Va24 + T cells in the presence of an invariant T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days.
  • an cAMP Cyclic adenosine monophosphate
  • TGF Transforming growth factor beta pathway activator
  • mTOR inhibitor optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days.
  • regulatory T cells refers to cells capable of suppressive activity (i.e. inhibiting proliferation of conventional T cells), either by cell-cell contact or by MLR suppression (Mixed Lymphocytes Reaction). These cells include different subpopulations including but not limited to, peripheral regulatory T cells, ⁇ regulatory T cells and invariant regulatory T cells.
  • invariant Foxp3 + regulatory T cells refers to cells having the following phenotype: CD3 + Va24 + Foxp3 + .
  • the term "invariant” as used herein includes the term “semi-invariant", where the semi-invariant T cells are T cells not expressing ⁇ 1.
  • the isolated population of the invention is a population of semi-invariant Foxp3 + T cells having the following phenotype: CD3 + Va24 + Foxp3 + Vpi ⁇ . These cells recognize non peptide lipid antigens under CD1 restriction.
  • y5Foxp3 + regulatory T cells refers to cells having the following phenotype: y5TCR + Foxp3 + . These cells recognize non peptide phospho antigens with no MHC (major histocompatibility complex) restriction.
  • MHCII restricted CD4 + Foxp3 + regulatory T cells refers to cells having the following phenotype: CD4 + CD25 + Foxp3 + . These cells are thymic derived or peripherally induced. These cells can be identified by their a TCR (T cell receptor) and recognize peptides (including foreign or self peptides) presented by restricted MHC class II (major histocompatibility complex class II) molecules.
  • treatment refers to therapeutic treatment and prophylactic and preventive measures, wherein the object is to prevent or slow down (lessen, diminish) the targeted pathological disorder or condition.
  • Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented.
  • a subject or mammal is successfully "treated” for a disease if, after receiving a therapeutic amount of Foxp3 + regulatory T cells or a therapeutically amount of inactivated Foxp3 + regulatory T cells according to the present invention, the patient shows observable and/or measurable reduction in or absence of one or more of the following: reduction in the number of pathogenic cells; reduction in the percent of total cells that are pathogenic; and/or relief to some extent, of one or more of the symptoms associated with the specific disease or condition; reduced morbidity and mortality, and improvement in quality of life issues.
  • the above parameters for assessing successful treatment and improvement in the disease are readily measurable by routine procedures familiar to a physician.
  • therapeutically effective amount refers to the number of Foxp3 + regulatory T cells or of inactivated Foxp3 + regulatory T cells that is aimed at inducing a therapeutic response, without causing significant negative or adverse side effects to the target.
  • a therapeutically effective amount may be administered prior to the onset of the disease to be treated, for a prophylactic or preventive action. Alternatively or additionally, the therapeutically effective amount may be administered after initiation of the disease to be treated, for a therapeutic action.
  • therapeutic response refers to a therapeutic benefit induced by the Foxp3 + regulatory T cell therapy or the Foxp3 + regulatory T cell vaccination in a subject.
  • a therapeutic response may include the fact of (1) delaying or preventing the onset of the disease to be treated; (2) slowing down or stopping the progression, aggravation, or deterioration of one or more symptoms of the disease to be treated; (3) bringing about ameliorations of the symptoms of the disease to be treated; (4) reducing the severity or incidence of the disease to be treated; or (5) curing the disease to be treated.
  • subject or patient refers to a mammal, preferably a human.
  • the terms subject and patient may be used with the same meaning.
  • non-human mammal include a pet such as a dog, a cat, a domesticated pig, a rabbit, a ferret, a hamster, a mouse, a rat and the like; a primate such as a chimp, a monkey, and the like; an economically important animal such as cattle, a pig, a rabbit, a horse, a sheep, a goat.
  • the subject is awaiting the receipt of, or is receiving medical care or was/is/will be the object of a medical procedure, or is monitored for the development of a disease.
  • the subject is an adult (for example a subject above the age of 18).
  • the subject is a child (for example a subject below the age of 18).
  • the subject is a male.
  • the subject is a female.
  • allogeneic cells refers to cells isolated from one subject (the donor) and infused in another (the recipient or host).
  • autologous cells refers to cells that are isolated and infused back into the same subject (recipient or host).
  • the present invention relates to a method for generating ex vivo invariant Foxp3 + regulatory T cells as defined here above.
  • the method for generating ex vivo invariant Foxp3 + regulatory T cells comprises:
  • CD3 + Va24 + T cells preferably CD3 + Va24 + CD45RA+ T cells, in the presence of an invariant T cell activator and the following agents: i) an cAMP
  • cytokine selected from the group of IL-2, IL-7, IL-15 and TSLP (Thymic stromal lymphopoietin), for at least 5 days, thereby obtaining a population of invariant Foxp3 + regulatory T cells ex vivo generated, preferably from invariant naive (CD45RA + ) T cells.
  • the CD3 + Va24 + T cells are obtained by any technic well known in the art from a blood sample.
  • the CD3 + Va24 + T cells preferably CD3 + Va24 + CD45RA+ T cells
  • the CD3 + Va24 + T cells preferably CD3 + Va24 + CD45RA+ T cells, may be isolated from frozen PBMCs.
  • the obtainment of isolated CD3 + Va24 + T cells may be improved by an optional first to a purification step.
  • the CD3 + Va24 + T cells preferably CD3 + Va24 + CD45RA + T cells, are stimulated with antigen pulsed tolerogenic DCs (for example ovalbumin pulsed tolerogenic DCs) in the presence of soluble anti-CD28 and anti-CD40 antibodies.
  • the time of stimulation ranges between 1 hour and 24 hours, preferably between 10 hours and 20 hours, more preferably during about 16 hours. After stimulation, cells are washed, for example with PBS, and stained with anti-CD 154 and anti-CD4 antibodies for sorting.
  • the purified CD3 + Va24 + CD154 + T cells are enriched and may be used for the following activation step.
  • the CD3 + Va24 + T cells are activated in the presence of an invariant T cell activator.
  • Said invariant T cell activator can be a polyclonal invariant T cell activator or an antigen-specific invariant T cell activator.
  • the polyclonal invariant T cell activator is a Va24 activator.
  • Va24 activator include, but are not limited to, anti-Va24 antibody such as 6B11 antibody (Montoya CJ et al. Immunology.
  • CD1 ligands including CD la ligands, CD lb ligands, CDlc ligands and CD Id ligands, preferably CD Id ligands such as a-galactosylceramide (a-GalCer) and analogs such as for example HS44 (a synthetic amino cyclitolic ceramide analogue in which the sugar head group is a carba cyclitol ring that mimics glucose instead of galactose, and which has the O- glycosidic linkage replaced with an amide group), cc-GalCer analogs of the table 1 herein below:
  • a-GalCer a-galactosylceramide
  • analogs such as for example HS44 (a synthetic amino cyclitolic ceramide analogue in which the sugar head group is a carba cyclitol ring that mimics glucose instead of galactose, and which has the O- glycosidic linkage replaced with an amide
  • R is C0 2 Me, C0 2 H or CH 2 OH; a-glucuronyl- and a-galacturonyl-ceramides and analogs thereof; iGb3 (Isoglobotriosylceramide); N-glycolyl (NGc) gangliosides such as for example NGcGM3; glycosphingo lipids or phosphoglycero lipids such as phosphatidylinositol, phosphatidylethanolamine, and phosphatidylglyerol presented preferably by CD Id.
  • NGc N-glycolyl
  • the polyclonal invariant T cell activator is an anti-Va24 antibody, preferably a m6Bl 1 antibody.
  • the polyclonal invariant T cell activator is soluble in the culture medium. In another embodiment, the polyclonal invariant T cell activator is coated to the culture plate. In one embodiment, the polyclonal invariant T cell activator is used in the presence of feeder cells, preferably autologous feeder cells.
  • Feeder cells include, but are not limited to, ACD3 cells (T cell-depleted accessory cells), irradiated PBMCs, irradiated DCs, artificial APCs (antigen presenting cells), Sf9 cells, insect cells, a pool of PBMCs or a pool of B cells from different subjects, KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
  • ACD3 cells T cell-depleted accessory cells
  • irradiated PBMCs irradiated DCs
  • artificial APCs antigen presenting cells
  • Sf9 cells insect cells
  • a pool of PBMCs or a pool of B cells from different subjects KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
  • the feeder cells used in the invention are ACD3 cells that are isolated by negative selection from PBMCs by incubation with anti-CD3 coated beads and then irradiated at 3000 rad.
  • the ratio T cells / feeder cells ranges from 1:100 to 1:10000, preferably from 1:1000 to 1:5000.
  • the expression “from 1:100 to 1:10000” includes, without limitation 1:100, 1:200, 1:300, 1:400, 1:500, 1:600, 1:700, 1:800, 1:900, 1:1000, 1:1250, 1:1500, 1: 1750, 1:2000, 1:2250, 1:2500, 1:2750, 1:3000, 1:3250, 1:3500, 1:4000, 1:4250, 1:4500, 1:4750, 1:5000, 1:5250, 1:5500, 1: 5750, 1:6000, 1:6250, 1:6500, 1: 6750, 1:7000, 1:7250, 1:7500, 1: 7750, 1:8000, 1:8250, 1:8500, 1: 8750, 1:9000, 1:9250, 1:9500, 1:9750 and 1:10000.
  • the antigen-specific invariant T cell activator is tolerogenic dendritic cells (DCs) expressing CD1, i.e. CD la, CD lb, CDlc and/or CD Id, and pulsed with at least one non peptide lipid antigen.
  • DCs dendritic cells
  • the tolerogenic DCs express CD Id.
  • tolerogenic DCs express on their surface the major histocompatibility (MHC) class la and/or MHC class lb.
  • MHC class la presentation refers to the "classical” presentation through HLA-A, HLA-B and/or HLA-C molecules whereas the MHC class lb presentation refers to the "non-classical" antigen presentation through HLA-E, HLA-F, HLA-G and/or HLA-H molecules.
  • tolerogenic DCs express 50% of MHC class la molecules and 50% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 45% of MHC class la molecules and 55% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 40%> of MHC class la molecules and 60% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 35% of MHC class la molecules and 65% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 30% of MHC class la molecules and 70% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 25% of MHC class la molecules and 75% of MHC class lb molecules on their surface.
  • tolerogenic DCs express 20% of MHC class la molecules and 80% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 15% of MHC class la molecules and 85% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 10% of MHC class la molecules and 90% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 5% of MHC class la molecules and 95% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express only MHC class lb molecules on their surface.
  • tolerogenic DCs express 50% of HLA-A, HLA-B and/or HLA-C molecules and 50% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 45% of HLA-A, HLA-B and/or HLA-C molecules and 55% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 40% of HLA-A, HLA-B and/or HLA-C molecules and 60% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 35% of HLA-A, HLA-B and/or HLA-C molecules and 65% of HLA-E molecules on their surface.
  • tolerogenic DCs express 30% of HLA-A, HLA-B and/or HLA-C molecules and 70% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 25% of HLA-A, HLA-B and/or HLA-C molecules and 75% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 20% of HLA-A, HLA-B and/or HLA-C molecules and 80% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 15% of HLA-A, HLA-B and/or HLA-C molecules and 85% of HLA-E molecules on their surface.
  • tolerogenic DCs express 10% of HLA-A, HLA-B and/or HLA-C molecules and 90% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 5% of HLA-A, HLA-B and/or HLA-C molecules and 95% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express only HLA-E molecules on their surface.
  • the non peptide lipid antigen is a recombinant antigen a- galactosylceramide and analogs such as for example HS44 (a synthetic amino cyclitolic ceramide analogue in which the sugar head group is a carba cyclitol ring that mimics glucose instead of galactose, and which has the O-glycosidic linkage replaced with an amide group), a-GalCer analogs of the table 1 herein above and homodimeric a- galactosylceramide analogs including the following:
  • R is C0 2 Me, C0 2 H or CH 2 OH; a-glucuronyl- and a-galacturonyl-ceramides and analogs; iGb3 (Isoglobotriosylceramide); N-glycolyl (NGc) gangliosides such as for example NGcGM3; glycosphingo lipids or phosphoglycerolipids such as phosphatidylinositol, phosphatidylethanolamine, and phosphatidylglyerol presented preferably by CD Id.
  • NGc N-glycolyl
  • the non peptide lipid antigen is derived from immunogenic apoptotic bodies from cancer cells or derived from tissue lysate. Cancer cells may derive from tumor biopsy or from expansion of circulatory cancer cells.
  • Immunogenic apoptotic bodies from cancer cells may be obtained for example with anthracyclines including, without limitation, doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin, UVC or ⁇ -radiation treated cancer cells releasing apoptotic bodies or can be directly isolated from anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin; UVC or ⁇ -radiation treated cancer.
  • anthracyclines including, without limitation, doxorubicin, daunorubicin, idarubicin and mitoxanthrone
  • oxaliplatin UVC or ⁇ -radiation treated cancer.
  • tissue lysate examples include, but are not limited to, synovial liquid or inflammatory tissue lysate.
  • tolerogenic DCs refers to DCs capable to induce tolerance.
  • tolerogenic DCs are capable of secreting more suppressive cytokines such as IL-10 and TGF than proinflammatory cytokines such as IL-12, IL-23 or TNFa.
  • DCs are defined as tolerogenic when they secrete IL-10 and IL-12 in a ratio IL-10 : IL-12 > 1.
  • Methods for obtaining tolerogenic DCs are well-known in the art.
  • An exemplary method is the generation of tolerogenic DCs from CD14 + monocytes.
  • CD14 + monocytes are cultured in the presence of GM-CSF and IL-4, or in the presence of GM-CSF and IFNa, for the generation of immature DCs.
  • TAP transporter transporter associated with antigen processing
  • Exemplary methods to inhibit the TAP transporter in the endoplasmic reticulum include, but are not limited to, CRISPR-CAS-9 technology, silencing RNA, transfected DCs with the UL-10 viral protein from the CMV (cytomegalovirus) or the use of viral proteins.
  • viral proteins able to inhibit the TAP transporter include, but are not limited to, HSV-1 ICP47 protein, varicella- virus UL49.5 protein, cytomegalovirus US6 protein or gammaherpesvirus EBV BNLF2a protein.
  • Another method is the use of a chemical product to inhibit the expression of MHC class la molecules without changing HLA-E expression on the surface of tolerogenic DCs.
  • chemical products include, but are not limited to, 5'- methyl-5 '- thioadenosine or leptomycin B.
  • Methods for inducing the expression of CDl, i.e. CD la, CD lb, CDlc and/or CD Id on the surface of the tolerogenic DCs are well-known in the art.
  • sulfatide can be used for the expression of CD la; rifampin and a number of its derivative (e.g., rifabutin) for the expression of CD lb; cholesteryl esters and acylated steryl glycosides for the expression of CDlc and rosiglitazone; retinoic acid; RARa agonist such as AM580, CD437, AM80, BMS961, NRX195183, All-trans-retinoic acid, 9-cis-Retinoic acid, Ch55, TTNPB (4-[(E)-2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)- l-propenyl]benzoic acid), tamibarotene; PPARy agonist such as ciglitazone, darglitazone, edaglitazone, genistein, indomethacin, GW 1929, LG
  • AM580 can be used at a concentration ranging from 1 nM to 10 ⁇ .
  • the cAMP activator added in the culture allows the activation of the cAMP pathway.
  • cAMP activator include, but are not limited to PGE2 (prostaglandin E2), an EP2 or EP4 agonist, a membrane adenine cyclase activator such as forskolin, or metabotropic glutamate receptors agonists.
  • PGE2 examples include, but are not limited to, PGE2 of ref P5640 or P0409 (Sigma- Aldrich), PGE2 of ref 2296 (R&D Systems), PGE2 of ref 2268 (Bio Vision), PGE2 of ref 72192 (Stemcell), PGE2 of ref abl44539 (Abeam), and PGE2 of ref 14010 (Cayman Chemical).
  • the cAMP activator preferably PGE2 is used at a concentration ranging from 0.01 ⁇ to 10 ⁇ .
  • the expression "from 0.01 ⁇ to 10 ⁇ " includes, without limitation 0.02 ⁇ , 0.03 ⁇ , 0.04 ⁇ , 0.05 ⁇ , 0.06 ⁇ , 0.07 ⁇ , 0.08 ⁇ , 0.09 ⁇ , 0.1 ⁇ , 0.2 ⁇ , 0.3 ⁇ , 0.4 ⁇ , 0.5 ⁇ , 0.6 ⁇ , 0.7 ⁇ , 0.8 ⁇ , 0.9 ⁇ , 1 ⁇ , 1.5 ⁇ , 2 ⁇ , 2.5 ⁇ , 3 ⁇ , 3.5 ⁇ , 4 ⁇ , 4.5 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ .
  • PGE2 is at a concentration ranging from 0.03 ⁇ to 1.5 ⁇ .
  • the TGF pathway activator added in the culture allows the activation of the TGF pathway.
  • TGF pathway activators include, but are not limited to, TGFP family (TGFpi, TGFP2, TGFP3), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, and nodal.
  • TGFP examples include, but are not limited to, TGFpi of ref T7039 (Sigma- Aldrich), TGFp2 of ref T2815 (Sigma- Aldrich), TGFp3 of ref T5425 (Sigma- Aldrich), human TGFpl of ref P01137 (R&D system), human TGFpl of ref 580702 (Biolegend), TGFpl of ref HZ-1011 (HumanZyme), human TGFpl of ref 14- 8348-62 (Affymetrix eBioscience).
  • the pathway activator is used at a concentration ranging from 1 ng/ml to 20 ng/ml.
  • the expression "from 1 ng/ml to 20 ng/ml” includes, without limitation 2 ng/ml, 2.5 ng/ml, 3 ng/ml, 3.5 ng/ml, 4 ng/ml, 4.5 ng/ml, 5 ng/ml, 5.5 ng/ml, 6 ng/ml, 6.5 ng/ml, 7 ng/ml, 7.5 ng/ml, 8 ng/ml, 8.5 ng/ml, 9 ng/ml, 9.5 ng/ml, 10 ng/ml, 11 ng/ml, 12 ng/ml, 13 ng/ml, 14 ng/ml, 15 ng/ml, 16 ng/ml, 17 ng/ml, 18 ng/ml, 19 ng/ml.
  • TGFP is at a concentration ranging from 2.5 ng/ml to 7.5 ng/ml.
  • the mTOR inhibitor added in the culture allows the inhibition of the mTOR pathway.
  • mTOR inhibitor include, but are not limited to, rapamycin (also named sirolimus) and its analogs (termed rapalogs); wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG); curcumin; resveratrol; genistein; 3, 3-diindolylmethane (DIM); LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l- benzopyran-4-one); PP242; PP30; Torinl ; Ku-0063794; WAY-600; WYE-687; WYE- 354; and mTOR and PI3K dual-specificity inhibitors such as GNE477, NVP-BEZ235, PI-103, XL765 and WJD008.
  • rapamycin examples include, but are not limited to, rapamycin of ref R0395 (Sigma- Aldrich), rapamycin of ref SI 039 (Selleckchem), rapamycin of ref 1292 (Tocris), rapamycin of ref R-5000 (LC Laboratories), rapamycin of ref tlrl-rap (InvivoGen), rapamycin of ref abl20224 (Abeam), rapamycin of ref R0395 (Sigma-Aldrich).
  • Examples of compounds of the same chemical class than rapamycin used clinically include, but are not limited to, Everolimus (code name RADOOl), Temsirolimus (code name CCI-779, NSC 683864), Zotarolimus (code name ABT-578).
  • the mTOR inhibitor preferably rapamycin
  • the expression "from 0.1 nM to 50 nM” includes, without limitation 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 26 nM, 27 nM, 28 nM, 29 nM
  • IL-2 is used at a concentration ranging from 10 IU/ml to 1000 IU/ml.
  • the expression "from 10 IU/ml to 1000 IU/ml” includes, without limitation 15 IU/ml, 20 IU/ml, 25 IU/ml, 30 IU/ml, 35 IU/ml, 40 IU/ml, 45 IU/ml, 50 IU/ml, 55 IU/ml, 60 IU/ml, 65 IU/ml, 70 IU/ml, 75 IU/ml, 80 IU/ml, 85 IU/ml, 90 IU/ml, 95 IU/ml, 100 IU/ml, 150 IU/ml, 200 IU/ml, 250 IU/ml, 300 IU/ml, 350 IU/ml, 400 IU/ml, 450 IU/ml, 500 IU/ml, 550 IU/ml, 600 IU/m
  • IL-7 is used at a concentration ranging from 1 ng/ml to 100 ng/ml.
  • the expression "from 1 ng/ml to 100 ng/ml” includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
  • IL-15 is used at a concentration ranging from 1 ng/ml to 50 ng/ml.
  • the expression "from 1 ng/ml to 50 ng/ml” includes, without limitation 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 9 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml.
  • IL-15 is used at a concentration ranging from 10 ng/ml to 30 ng/ml.
  • TSLP is used at a concentration ranging from 1 ng/ml to 100 ng/ml.
  • the expression "from 1 ng/ml to 100 ng/ml” includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
  • neutralizing antibodies can be added to the culture to prevent the generation of other populations of regulatory T cells.
  • neutralizing antibodies include, but are not limited to, anti-IFNy, anti-IL-4, and/or anti-IL12 antibodies.
  • anti-IFNy antibodies examples include, but are not limited to, Affymetrix eBioscience (Ref 14-7318), R&D systems (Ref MAB285), Novus Biologicals (Ref AF- 485-NA).
  • anti-IL-4 antibodies include, but are not limited to, R&D Systems (Ref MAB304, MAB204, or MAB204), Affymetrix eBioscience (Ref 14-7048), GeneTex (Ref GTX10755).
  • anti-IL-12 antibodies include, but are not limited to, Affymetrix eBioscience (Ref 16-7129 or 16-8126), Biolegend (Ref 508803), R&D systems (Ref MAB219, AF-219, or AB-219).
  • the culture medium used in the culture of the invention comprises (i) one or more pH buffering system(s); (ii) inorganic salt(s); (iii) trace element(s); (iv) free amino acid(s); (v) vitamin(s); (vi) hormone(s); (vii) carbon/energy source(s).
  • inorganic salts include, but are not limited to, calcium bromide, calcium chloride, calcium phosphate, calcium nitrate, calcium nitrite, calcium sulphate, magnesium bromide, magnesium chloride, magnesium sulphate, potassium bicarbonate, potassium bromide, potassium chloride, potassium dihydrogen phosphate, potassium disulphate, di- potassium hydrogen phosphate, potassium nitrate, potassium nitrite, potassium sulphite, potassium sulphate, sodium bicarbonate, sodium bromide, sodium chloride, sodium disulphate, sodium hydrogen carbonate, sodium dihydrogen phosphate, di-sodium hydrogen phosphate, sodium sulphate and a mix thereof.
  • trace elements include, but are not limited to cobalt (Co), copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), selenium (Se), zinc (Zn) and the salts thereof.
  • free amino acids include, but are not limited to L-alanine, L-arginine, L- asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, taurine, L-threonine, L-tryptophan, L-tyrosine, L-valine and a mix thereof
  • vitamins include, but are not limited to biotin (vitamin H); D-calcium- pantothenate; choline chloride; folic acid (vitamin B9); myo-inositol; nicotinamide; pyridoxal (vitamin B6); riboflavin (vitamin B2); thiamine (vitamin Bl); cobalamin (vitamin B 12); acid ascorbic; a-tocopherol (vitamin E) and a mix thereof.
  • biotin vitamin H
  • D-calcium- pantothenate choline chloride
  • folic acid vitamin B9
  • myo-inositol nicotinamide
  • pyridoxal vitamin B6
  • riboflavin vitamin B2
  • thiamine vitamin Bl
  • cobalamin vitamin B 12
  • acid ascorbic a-tocopherol (vitamin E) and a mix thereof.
  • the culture medium is a commercially available cell culture medium, in particular selected in a group comprising the IMDM (Iscove's Modified Dulbecco's Medium) from GIBCO® or the RPMI 1640 medium from GIBCO®.
  • IMDM Iscove's Modified Dulbecco's Medium
  • the culture medium is a serum-free culture medium such as the AIM-V medium from GIBCO®, the X-VIVO 10, 15 and 20 media from LONZA.
  • the culture medium can be further supplemented with additional compound(s), in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
  • the culture medium is IMDM.
  • the culture medium comprises IMDM cell culture medium; from 1% (w/w) to 5% (w/w) of foetal bovine serum; from 10 IU/ml to 200 IU/ml of penicillin; from 10 IU/ml to 200 IU/ml of streptomycin; from 0.1 mM to 10 mM of a mixture of non-essential amino acids, in particular amino acids selected in a group comprising alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine; from 0.5 mM to 10 mM of glutamine from 10 mM
  • the medium is a nTreg polarizing medium.
  • nTreg polarizing medium as a medium such as RPMI medium comprising at least one cAMP activator as described hereabove, at least one TGF pathway activator as described here above and at least one mTor inhibitor as described hereabove.
  • the "nTreg polarizing medium” refers to a RPMI medium comprising TGF , rapamycin and PGE2.
  • the medium is an inflammatory medium.
  • the inventors define an "inflammatory medium" as a medium such as IMDM comprising inflammatory cytokines such as for example IL- ⁇ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml).
  • the culture for generating the invariant Foxp3 + regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days.
  • the expression "at least 5 days” includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days.
  • a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium.
  • portion is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium.
  • fresh culture medium refers to a culture medium that has not been in contact with any CD3+ T cells.
  • the method for generating ex vivo invariant Foxp3 + regulatory T cells comprises: - culturing CD3 + Va24 + T cells, preferably CD3 + Va24 + CD45RA+ T cells, in the presence of ACD3 feeder cells and coated 6B11 mAb and the following agents: i) PGE2, ii) TGFp, iii) rapamycin, and optionally iv) IL-2 and/or IL-15, for at least 5 days, thereby obtaining a population of invariant Foxp3 + regulatory T cells ex vivo generated, preferably from invariant naive (CD45RA + ) T cells.
  • the method for generating ex vivo invariant Foxp3 + regulatory T cells comprises:
  • CD3 + Va24 + T cells preferably CD3 + Va24 + CD45RA+ T cells
  • tolerogenic DCs expressing CDld incubation with AM580 during about 24h
  • aGalactosyleramide in the presence of ACD3 feeder cells and the following agents: i) PGE2, ii) TGFp, iii) rapamycin, and optionally iv) IL-2 and/or IL-15, for at least 5 days, thereby obtaining a population of invariant Foxp3 + regulatory T cells ex vivo generated, preferably from invariant naive (CD45RA + ) T cells.
  • the present invention also relates to an ex vivo method of generation and expansion of invariant Foxp3 + regulatory T cells, comprising: generating the invariant Foxp3 + regulatory T cells as described here above, expanding the invariant Foxp3 + regulatory T cells generated by contacting them with an invariant T cell activator (preferably either ACD3 feeder cells and coated 6B11 mAb or tolerogenic DCs expressing CDld (incubation with AM580 during about 24h) and pulsed with aGalactosyleramide in the presence of ACD3 feeder cells) in the presence of and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator (preferably PGE2), ii) a TGF (Transforming growth factor beta) pathway activator (preferably TGFP), iii) a mTOR inhibitor (preferably rapamycin), and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TS
  • the invariant Foxp3 + regulatory T cell population generated ex vivo is isolated by flow cytometry based on the following phenotype: CD3 + Va24 + CD45RO + Foxp3 + .
  • the isolated invariant Foxp3 + regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of a polyclonal T cell activator.
  • polyclonal T cell activator examples include, but are not limited to, mitogen such as PMA/ionomycin, super-antigen, anti-CD3 antibody...
  • the anti-CD3 monoclonal antibody is coated.
  • the polyclonal T cell activator can be used in the presence of feeder cells as described here above.
  • the isolated invariant Foxp3 + regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of antigen- specific invariant T cell activator as described here above.
  • the antigen-specific T cell activator can be used in the presence of feeder cells as described here above.
  • the culture for expanding the ex vivo generated invariant Foxp3 + regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days.
  • the expression "at least 5 days” includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days.
  • a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium.
  • portion is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium.
  • 40%) (v/v) to 60%) (v/v) of the volume of the culture medium of the first step is discarded.
  • the volume that is discarded is replaced with an identical volume of fresh culture medium.
  • fresh culture medium refers to a culture medium that has not been in contact with any CD3+ T cells.
  • invariant Foxp3 + regulatory T cells are generated ex vivo by culturing CD3 + Va24 + CD45RA + T cells obtained from PBMCs by flow cytometry (5.10 3 cells/ml) in the presence of autologous ACD3 feeder cells (125 10 5 cells/ml) and coated 6B11 mAb (2 ⁇ g/ml) in the presence of PGE2 (1 ⁇ ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1 , IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture.
  • invariant Foxp3 + regulatory T cells are generated ex vivo by culturing CD3 + Va24 + CD45RA + T cells (5.10 3 cells/ml) obtained from PBMCs by flow cytometry (5.10 3 cells/ml) in the presence of tolerogenic DCs expressing CD Id (incubation with AM580 during about 24h) and pulsed with aGalactosyleramide and in the presence of ACD3 feeder cells (125 10 5 cells/ml), PGE2 (1 ⁇ ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5.
  • CD3 + Va24 + CD45RA + T cells 5.10 3 cells/ml obtained from PBMCs by flow cytometry (5.10 3 cells/ml) in the presence of tolerogenic DCs expressing CD Id (incubation with AM580 during about 24h) and pulsed with aGalactosyleramide and in the presence of ACD3
  • IL-2 100 Ul/ml
  • IL- 15 10 ng/ml
  • TGF 5 ng/ml
  • PGE2 50 nM
  • TGF 5 ng/ml
  • Rapamycin 1 nM
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • tolerogenic DCs were obtained by culturing CD14 + monocytes isolated from PBMCs in the presence of AIMV supplemented with GMCSF (lOO ng/ml), IL-4 (10 ng/ml) and AM580 (100 nM). At day 3 and 6, the medium is discarded and replaced by fresh medium comprising GM-CSF and IL-4. On day 6, the tolerogenic DCs are pulsed for 24 hours in the presence of aGalactosylceramide (100 ng/ml) and AM580 (100 nM). yd Foxp3 + regulatory T cells
  • the present invention also relates to a method for generating ex vivo ⁇ Foxp3 + regulatory T cells.
  • the method for generating ex vivo ⁇ Foxp3 + regulatory T cells comprises:
  • CD3 + TCRy5 + T cells preferably CD3 + TCRy5 + CD45RA+ T cells, in the presence of an ⁇ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (Thymic stromal lymphopoietin), for at least 5 days, thereby obtaining a population of ⁇ Foxp3 + regulatory T cells ex vivo generated, preferably from ⁇ naive (CD45RA + ) T cells.
  • an cAMP Cyclic adenosine monophosphate
  • TGF Transforming growth factor beta pathway activator
  • iii) a mTOR inhibitor optionally iv) at least one cytokine selected
  • the CD3 + TCRy5 + T cells are obtained by any technic well known in the art from a blood sample.
  • the CD3 + TCRy5 + T cells preferably CD3 + TCRy5 + CD45RA+ T cells
  • the CD3 + TCRy5 + T cells preferably CD3 + TCRy5 + CD45RA+ T cells, may be isolated from frozen PBMCs.
  • the obtainment of isolated CD3 + TCRy5 + T cells may be improved by an optional first to a purification step.
  • the CD3 + TCRy5 + T cells preferably CD3 + TCRy5 + CD45RA + T cells, are stimulated with antigen pulsed tolerogenic DCs (for example ovalbumin pulsed tolerogenic DCs) in the presence of soluble anti-CD28 and anti-CD40 antibodies.
  • the time of stimulation ranges between 1 hour and 24 hours, preferably between 10 hours and 20 hours, more preferably during about 16 hours. After stimulation, cells are washed, for example with PBS, and stained with anti-CD 154 and anti-CD4 antibodies for sorting.
  • the purified CD3 + TCRy5 + CD154 + T cells are enriched and may be used for the following activation step.
  • the CD3 + TCRy5 + T cells are activated in the presence of an ⁇ T cell activator.
  • Said ⁇ T cell activator can be a polyclonal ⁇ T cell activator or an antigen-specific ⁇ T cell activator.
  • the polyclonal ⁇ T cell activator is a TCRy0 activator.
  • TCRy0 activator include, but are not limited to, anti-TCR ⁇ antibody such as purified mouse anti-human TCR ⁇ Clone B l (ref 555715, BD Biosciences), anti- human TCR ⁇ Antibody (ref 331209, Biolegend), monoclonal TCR ⁇ Antibody (ref NBP2-22489 or NBP2-22510, Novus Biologicals), anti-mouse ⁇ TCR (ref 12-571 1-81 , eBioscience), TCR ⁇ Antibody (ref MAB7297, R&D Systems), anti-T-Cell Receptor ⁇ antibody (ref ABIN2372990, antibodies-online), anti-TCR gamma + TCR delta antibody (ref ab25663, Abeam), anti- ⁇ TCR antibody clone IMMU510 (Beckman Coulter); non peptide phosphoantigens (Beckman Co
  • the polyclonal ⁇ T cell activator is MHC Class-I related A (MICA). In another embodiment, the polyclonal ⁇ T cell activator is immunogenic apoptotic bodies from cancer cells or derived from tissue lysate.
  • MICA MHC Class-I related A
  • Cancer cells may derive from tumor biopsy or from expansion of circulatory cancer cells.
  • Immunogenic apoptotic bodies from cancer cells may be obtained for example with anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin, UVC or ⁇ -radiation treated cancer cells releasing apoptotic bodies or can be directly isolated from anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin; UVC or ⁇ -radiation treated cancer.
  • the polyclonal ⁇ T cell activator is an anti-TCRy5 antibody or a non peptide phosphoantigen such as isoprenyl pyrophosphate (IPP).
  • the polyclonal ⁇ T cell activator preferably the anti-TCR ⁇ antibody
  • the polyclonal ⁇ T cell activator is soluble in the culture medium.
  • the polyclonal ⁇ T cell activator is coated to the culture plate.
  • the polyclonal ⁇ T cell activator, preferably the anti-TCR ⁇ antibody is used in the presence of feeder cells, preferably autologous feeder cells.
  • Feeder cells include, but are not limited to, ACD3 cells (T cell-depleted accessory cells), irradiated PBMCs, irradiated DCs, artificial APCs (antigen presenting cells), Sf9 cells, insect cells, a pool of PBMCs or a pool of B cells from different subjects, KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
  • ACD3 cells T cell-depleted accessory cells
  • irradiated PBMCs irradiated DCs
  • artificial APCs antigen presenting cells
  • Sf9 cells insect cells
  • a pool of PBMCs or a pool of B cells from different subjects KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
  • the feeder cells used in the invention are ACD3 cells that are isolated by negative selection from PBMCs by incubation with anti-CD3 coated beads and then irradiated at 3000 rad.
  • the ratio T cells / feeder cells ranges from about 1 : 100 to about 1 : 10 000, preferably from 1 : 1 000 to 1 :5 000.
  • the expression "from 1 : 100 to 1 : 10 000” includes, without limitation 1 : 100, 1 :200, 1 :300, 1 :400, 1 :500, 1 :600, 1 :700, 1 :800, 1 :900, 1 : 1 000, 1 : 1 250, 1 :1 500, 1 : 1 750, 1 :2 000, 1 :2 250, 1 :2 500, 1 :2 750, 1 :3 000, 1 :3 250, 1 :3 500, 1 : 3 750, 1 :4 000, 1 :4 250, 1 :4 500, 1 : 4 750, 1 :5 000, 1 :5 250, 1 :5 500, 1 :5 750, 1 :6 000, 1 :6 250, 1 :6 000, 1 :6 250,
  • tolerogenic DCs refers to DCs capable to induce tolerance.
  • tolerogenic DCs are capable of secreting more suppressive cytokines such as IL-10 and TGF than proinflammatory cytokines such as IL-12, IL-23 or TNFa.
  • DCs are defined as tolerogenic when they secrete IL-10 and IL-12 in a ratio IL-10: IL-12 > 1.
  • tolerogenic DCs express on their surface the major histocompatibility (MHC) class la and/or MHC class lb.
  • MHC class la presentation refers to the "classical” presentation through HLA-A, HLA-B and/or HLA-C molecules whereas the MHC class lb presentation refers to the "non-classical" antigen presentation through HLA-E, HLA-F, HLA-G and/or HLA-H molecules.
  • tolerogenic DCs express 50% of MHC class la molecules and 50%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 45%> of MHC class la molecules and 55%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 40%> of MHC class la molecules and 60% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 35%> of MHC class la molecules and 65%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 30% of MHC class la molecules and 70% of MHC class lb molecules on their surface.
  • tolerogenic DCs express 25%> of MHC class la molecules and 75%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 20% of MHC class la molecules and 80% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 15%> of MHC class la molecules and 85%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 10% of MHC class la molecules and 90% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 5% of MHC class la molecules and 95% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express only MHC class lb molecules on their surface.
  • tolerogenic DCs express 50% of HLA-A, HLA-B and/or HLA-C molecules and 50% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 45% of HLA-A, HLA-B and/or HLA-C molecules and 55% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 40% of HLA-A, HLA-B and/or HLA-C molecules and 60% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 35% of HLA-A, HLA-B and/or HLA-C molecules and 65% of HLA-E molecules on their surface.
  • tolerogenic DCs express 30% of HLA-A, HLA-B and/or HLA-C molecules and 70% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 25% of HLA-A, HLA-B and/or HLA-C molecules and 75% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 20% of HLA-A, HLA-B and/or HLA-C molecules and 80% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 15% of HLA-A, HLA-B and/or HLA-C molecules and 85% of HLA-E molecules on their surface.
  • tolerogenic DCs express 10% of HLA-A, HLA-B and/or HLA-C molecules and 90%> of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 5% of HLA-A, HLA-B and/or HLA-C molecules and 95% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express only HLA-E molecules on their surface.
  • An exemplary method is the generation of tolerogenic DCs from CD14 + monocytes.
  • CD14 + monocytes are cultured in the presence of GM-CSF and IL-4, or in the presence of GM-CSF and IFNa, for the generation of immature DCs.
  • TAP transporter transporter associated with antigen processing
  • Exemplary methods to inhibit the TAP transporter in the endoplasmic reticulum include, but are not limited to, CRISPR-CAS-9 technology, silencing RNA, transfected DCs with the UL-10 viral protein from the CMV (cytomegalovirus) or the use of viral proteins.
  • viral proteins able to inhibit the TAP transporter include, but are not limited to, HSV-1 ICP47 protein, varicella- virus UL49.5 protein, cytomegalovirus US6 protein or gammaherpesvirus EBV BNLF2a protein.
  • Another method is the use of a chemical product to inhibit the expression of MHC class la molecules without changing HLA-E expression on the surface of tolerogenic DCs.
  • chemical products include, but are not limited to, 5'- methyl-5 '- thioadenosine or leptomycin B.
  • the tolerogenic DCs are pulsed in the presence of at least one bisphosphonate, preferably aminobiphosphonate, during about 24h.
  • biphosphonate examples include, but are not limited to, zoledronic acid (or zoledronate), pamidronic acid, alendronic acid, risedronic acid, ibandronic acid, incadronic acid, etidronic acid, tiludronic acid, a combination thereof, a salt thereof and a hydrate thereof.
  • the biphosphanate is zoledronic acid or zoledronate.
  • biphosphonate in particular zoledronic acid
  • a concentration from 10 nM to 50 ⁇ is used at a concentration from 10 nM to 50 ⁇ .
  • the expression "from 10 nM to 50 ⁇ " includes, without limitation 50 nM, 100 nM, 250 nM, 500 nM, 750 nM, 1 ⁇ , 10 ⁇ , 20 ⁇ , 30 ⁇ , 40 ⁇ , 50 ⁇ .
  • the cAMP activator added in the culture allows the activation of the cAMP pathway.
  • Examples of cAMP activator include, but are not limited to, PGE2 (prostaglandin E2), an EP2 or EP4 agonist, a membrane adenine cyclase activator such as forskolin, or metabotropic glutamate receptors agonists.
  • PGE2 include, but are not limited to, PGE2 of ref P5640 or P0409 (Sigma- Aldrich), PGE2 of ref 2296 (R&D Systems), PGE2 of ref 2268 (Bio Vision), PGE2 of ref 72192 (Stemcell), PGE2 of ref abl44539 (Abeam), and PGE2 of ref 14010 (Cayman Chemical).
  • the cAMP activator preferably PGE2 is used at a concentration ranging from 0.01 ⁇ to 10 ⁇ .
  • the expression "from 0.01 ⁇ to 10 ⁇ " includes, without limitation 0.02 ⁇ , 0.03 ⁇ , 0.04 ⁇ , 0.05 ⁇ , 0.06 ⁇ , 0.07 ⁇ , 0.08 ⁇ , 0.09 ⁇ , 0.1 ⁇ , 0.2 ⁇ , 0.3 ⁇ , 0.4 ⁇ , 0.5 ⁇ , 0.6 ⁇ , 0.7 ⁇ , 0.8 ⁇ , 0.9 ⁇ , 1 ⁇ , 1.5 ⁇ , 2 ⁇ , 2.5 ⁇ , 3 ⁇ , 3.5 ⁇ , 4 ⁇ , 4.5 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ .
  • PGE2 is at a concentration ranging from 0.03 ⁇ to 1.5 ⁇ .
  • the TGFP pathway activator added in the culture allows the activation of the TGFP pathway.
  • TGFP pathway activators include, but are not limited to, TGFP family (TGFpi, TGFP2, TGFP3), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, and nodal.
  • TGFP examples include, but are not limited to, TGFpi of ref T7039 (Sigma- Aldrich), TGFp2 of ref T2815 (Sigma- Aldrich), TGFp3 of ref T5425 (Sigma- Aldrich), human TGFpl of ref P01137 (R&D system), human TGFpl of ref 580702 (Biolegend), TGFpl of ref HZ-1011 (HumanZyme), human TGFpl of ref 14- 8348-62 (Affymetrix eBioscience).
  • the pathway activator is used at a concentration ranging from 1 ng/ml to 20 ng/ml.
  • the expression "from 1 ng/ml to 20 ng/ml” includes, without limitation 2 ng/ml, 2.5 ng/ml, 3 ng/ml, 3.5 ng/ml, 4 ng/ml, 4.5 ng/ml, 5 ng/ml, 5.5 ng/ml, 6 ng/ml, 6.5 ng/ml, 7 ng/ml, 7.5 ng/ml, 8 ng/ml, 8.5 ng/ml, 9 ng/ml, 9.5 ng/ml, 10 ng/ml, 11 ng/ml, 12 ng/ml, 13 ng/ml, 14 ng/ml, 15 ng/ml, 16 ng/ml, 17 ng/ml, 18 ng/ml, 19 ng/ml.
  • TGF is at a concentration ranging from 2.5 ng/ml to 7.5 ng/ml.
  • the mTOR inhibitor added in the culture allows the inhibition of the mTOR pathway.
  • mTOR inhibitor include, but are not limited to, rapamycin (also named sirolimus) and its analogs (termed rapalogs); wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG); curcumin; resveratrol; genistein; 3, 3-diindolylmethane (DIM); LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l- benzopyran-4-one); PP242; PP30; Torinl ; Ku-0063794; WAY-600; WYE-687; WYE- 354; and mTOR and PI3K dual-specificity inhibitors such as GNE477, NVP-BEZ235, PI-103,
  • rapamycin examples include, but are not limited to, rapamycin of ref R0395 (Sigma- Aldrich), rapamycin of ref SI 039 (Selleckchem), rapamycin of ref 1292 (Tocris), rapamycin of ref R-5000 (LC Laboratories), rapamycin of ref tlrl-rap (InvivoGen), rapamycin of ref abl20224 (Abeam), rapamycin of ref R0395 (Sigma-Aldrich).
  • Examples of compounds of the same chemical class than rapamycin used clinically include, but are not limited to, Everolimus (code name RADOOl), Temsirolimus (code name CCI-779, NSC 683864), Zotarolimus (code name ABT-578).
  • the mTOR inhibitor preferably rapamycin
  • the expression "from 0.1 nM to 50 nM” includes, without limitation 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 26 nM, 27 nM, 28 nM, 29 nM
  • At least one cytokine selected from IL-2, IL-7, IL-15 and TSLP can be added in the culture.
  • IL-2 is used at a concentration ranging from 10 IU/ml to 1000 IU/ml.
  • the expression "from 10 IU/ml to 1000 IU/ml” includes, without limitation 15 IU/ml, 20 IU/ml, 25 IU/ml, 30 IU/ml, 35 IU/ml, 40 IU/ml, 45 IU/ml, 50 IU/ml, 55 IU/ml, 60 IU/ml, 65 IU/ml, 70 IU/ml, 75 IU/ml, 80 IU/ml, 85 IU/ml, 90 IU/ml, 95 IU/ml, 100 IU/ml, 150 IU/ml, 200 IU/ml, 250 IU/ml, 300 IU/ml, 350 IU/ml, 400 IU/ml, 450 IU/ml, 500 IU/ml, 550 IU/ml, 600 IU/m
  • IL-7 is used at a concentration from 1 ng/ml to 100 ng/ml.
  • the expression "from 1 ng/ml to 100 ng/ml” includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
  • IL-15 is used at a concentration ranging from 1 ng/ml to 50 ng/ml.
  • the expression "from 1 ng/ml to 50 ng/ml” includes, without limitation 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 9 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml.
  • IL-15 is used at a concentration ranging from 10 ng/ml to 30 ng/ml.
  • TSLP is used at a concentration ranging from 1 ng/ml to 100 ng/ml.
  • the expression "from 1 ng/ml to 100 ng/ml” includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
  • neutralizing antibodies can be added to the culture to prevent the generation of other populations of regulatory T cells.
  • neutralizing antibodies include, but are not limited to, anti-IFNy, anti-IL-4, and/or anti-IL12 antibodies.
  • anti-IFNy antibodies include, but are not limited to, Affymetrix eBioscience (Ref 14-7318), R&D systems (Ref MAB285), Novus Biologicals (Ref AF- 485-NA).
  • anti-IL-4 antibodies include, but are not limited to, R&D Systems (Ref MAB304, MAB204, or MAB204), Affymetrix eBioscience (Ref 14-7048), GeneTex (Ref GTX10755).
  • anti-IL-12 antibodies include, but are not limited to, Affymetrix eBioscience (Ref 16-7129 or 16-8126), Biolegend (Ref 508803), R&D systems (Ref MAB219, AF-219, or AB-219).
  • the culture medium used in the culture of the invention comprises (i) one or more pH buffering system(s); (ii) inorganic salt(s); (iii) trace element(s); (iv) free amino acid(s); (v) vitamin(s); (vi) hormone(s); (vii) carbon/energy source(s).
  • inorganic salts include, but are not limited to, calcium bromide, calcium chloride, calcium phosphate, calcium nitrate, calcium nitrite, calcium sulphate, magnesium bromide, magnesium chloride, magnesium sulphate, potassium bicarbonate, potassium bromide, potassium chloride, potassium dihydrogen phosphate, potassium disulphate, di- potassium hydrogen phosphate, potassium nitrate, potassium nitrite, potassium sulphite, potassium sulphate, sodium bicarbonate, sodium bromide, sodium chloride, sodium disulphate, sodium hydrogen carbonate, sodium dihydrogen phosphate, di-sodium hydrogen phosphate, sodium sulphate and a mix thereof.
  • trace elements include, but are not limited to, cobalt (Co), copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), selenium (Se), zinc (Zn) and the salts thereof.
  • free amino acids include, but are not limited to, L-alanine, L-arginine, L- asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, taurine, L-threonine, L-tryptophan, L-tyrosine, L-valine and a mix thereof.
  • vitamins include, but are not limited to, biotin (vitamin H); D-calcium- pantothenate; choline chloride; folic acid (vitamin B9); myo-inositol; nicotinamide; pyridoxal (vitamin B6); riboflavin (vitamin B2); thiamine (vitamin Bl); cobalamin (vitamin B 12); acid ascorbic; a-tocopherol (vitamin E) and a mix thereof.
  • biotin vitamin H
  • D-calcium- pantothenate choline chloride
  • folic acid vitamin B9
  • myo-inositol nicotinamide
  • pyridoxal vitamin B6
  • riboflavin vitamin B2
  • thiamine vitamin Bl
  • cobalamin vitamin B 12
  • acid ascorbic a-tocopherol (vitamin E) and a mix thereof.
  • the culture medium is a commercially available cell culture medium, in particular selected in a group comprising the IMDM (Iscove's Modified Dulbecco's Medium) from GIBCO® or the RPMI 1640 medium from GIBCO®.
  • IMDM Iscove's Modified Dulbecco's Medium
  • the culture medium is a serum-free culture medium such as the AIM-V medium from GIBCO®, the X-VIVO 10, 15 and 20 media from LONZA.
  • the culture medium can be further supplemented with additional compound(s), in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
  • the culture medium is IMDM.
  • the culture medium comprises IMDM cell culture medium; from 1% (w/w) to 5% (w/w) of foetal bovine serum; from 10 IU/ml to 200 IU/ml of penicillin; from 10 IU/ml to 200 IU/ml of streptomycin; from 0.1 mM to 10 mM of a mixture of non-essential amino acids, in particular amino acids selected in a group comprising alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine; from 0.5 mM to 10 mM of glutamine from 10 mM to 25 mM of HEPES pH 7.6-7.8.
  • the medium is a nTreg polarizing medium.
  • the inventors define a "nTreg polarizing medium" as a medium such as RPMI medium comprising at least one cAMP activator as described hereabove, at least one TGF pathway activator as described here above and at least one mTor inhibitor as described hereabove.
  • the "nTreg polarizing medium” refers to a RPMI medium comprising TGF , rapamycin and PGE2.
  • the medium is an inflammatory medium.
  • the inventors define an "inflammatory medium" as a medium such as IMDM comprising inflammatory cytokines such as for example IL- ⁇ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml).
  • the culture for generating the ⁇ Foxp3 + regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days.
  • the expression "at least 5 days” includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days.
  • a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium.
  • portion is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium.
  • fresh culture medium refers to a culture medium that has not been in contact with any CD3+ T cells.
  • the method for generating ex vivo ⁇ Foxp3 + regulatory T cells comprises:
  • CD3 + TCR ⁇ + T cells preferably CD3 + TCR ⁇ + CD45RA + T cells, in the presence of autologous ACD3 feeder cells and coated anti-TCR ⁇ antibody and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, thereby obtaining a population of ⁇ Foxp3 + regulatory T cells ex vivo generated, preferably from ⁇ naive (CD45RA + ) T cells.
  • agents i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, thereby obtaining a population of ⁇ Foxp3 + regulatory T cells ex vivo generated, preferably from ⁇ naive (CD45RA + ) T
  • the method for generating ex vivo ⁇ Foxp3 + regulatory T cells comprises: - culturing CD3 + TCR ⁇ + T cells, preferably CD3 + TCR ⁇ + CD45RA + T cells, in the presence of tolerogenic DC that have been pulsed with zoledronate during about 24h and in the presence of ACD3 feeder cells and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, - thereby obtaining a population of ⁇ Foxp3 + regulatory T cells ex vivo generated, preferably from ⁇ naive (CD45RA + ) T cells.
  • the present invention also relates to an ex vivo method of generation and expansion of ⁇ Foxp3 + regulatory T cells, comprising: generating the ⁇ Foxp3 + regulatory T cells as described here above, - expanding the ⁇ Foxp3 + regulatory T cells generated by contacting them in the presence of an ⁇ T cell activator (preferably either autologous ACD3 feeder cells and coated anti-TCR ⁇ antibody or tolerogenic DC that have been pulsed with zoledronate during about 24h and in the presence of ACD3 feeder cells) and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator (preferably PGE2), ii) a TGF (Transforming growth factor beta) pathway activator (preferably TGFP), iii) a mTOR inhibitor (preferably rapamycin), and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (preferably IL-2 and/or IL-15), for at least
  • the ⁇ Foxp3 + regulatory T cell population generated ex vivo is isolated by flow cytometry based on the following phenotype: CD3 + TCR ⁇ + CD45RO + Foxp3 + .
  • the isolated ⁇ Foxp3 + regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of a polyclonal ⁇ T cell activator.
  • polyclonal ⁇ T cell activator examples include, but are not limited to, mitogen such as PMA/ionomycin, super- antigen, anti-CD3 antibody...
  • the anti-CD3 monoclonal antibody is coated.
  • the polyclonal ⁇ T cell activator can be used in the presence of feeder cells as described here above.
  • the isolated ⁇ Foxp3 + regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of an antigen- specific ⁇ T cell activator as described here above.
  • the antigen- specific T cell activator can be used in the presence of feeder cells as described here above.
  • the culture for expanded the ex vivo generated ⁇ Foxp3 + regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days.
  • the expression "at least 5 days” includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days or more.
  • a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium.
  • portion is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium.
  • fresh culture medium refers to a culture medium that has not been in contact with any CD3+ T cells.
  • ⁇ Foxp3 + regulatory T cells are generated ex vivo by culturing CD3 + TCRy5 + CD45RA + T cells obtained from PBMCs by flow cytometry (5.10 3 cells/ml) in the presence of autologous ACD3 feeder cells (125 10 5 cells/ml) and coated anti-TCRy5 antibody (2 ⁇ g/ml) in the presence of PGE2 (1 ⁇ ), TGFP (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture.
  • ⁇ Foxp3 + regulatory T cells are generated ex vivo by culturing CD3 + TCRy5 + CD45RA + T cells (5.10 3 cells/ml) obtained from PBMCs by flow cytometry (5.10 3 cells/ml) in the presence of tolerogenic DCs, that have been pulsed with zoledronate during about 24h, and in the presence of ACD3 feeder cells (1.25 10 5 cells/ml), PGE2 (1 ⁇ ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5.
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • TGF 5 ng/ml
  • PGE2 50 nM
  • TGF 5 ng/ml
  • Rapamycin 1 nM
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • tolerogenic DCs were obtained by culturing CD14 + monocytes isolated from PBMCs in the presence of AIMV supplemented with GMCSF (100 ng/ml) and IL-4 (10 ng/ml). At day 3 and 6, the medium is discarded and replaced by fresh medium comprising GM-CSF and IL-4. On day 6, the tolerogenic DCs are pulsed for 24 hours in the presence of zoledronate (100 nM).
  • the present invention also relates to a method for generating ex vivo MHCII restricted CD4 + Foxp3 + regulatory T cells.
  • the method for generating ex vivo MHCII restricted CD4 + Foxp3 + regulatory T cells comprises:
  • CD3 + CD4 + CD25 T cells preferably CD3 + CD4 + CD25 CD45RA + T cells
  • a TCRa activator and the following agents i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (Thymic stromal lymphopoietin), for at least 5 days, thereby obtaining a population of MHCII restricted CD4 + Foxp3 + regulatory T cells ex vivo generated, preferably from naive (CD45RA + ) T cells.
  • cAMP Cyclic adenosine monophosphate
  • TGF Transforming growth factor beta pathway activator
  • a mTOR inhibitor optionally iv) at least one cytokine selected in the group of IL
  • the CD3 + CD4 + CD25 T cells are obtained by any technic well known in the art from a blood sample.
  • the CD3 + CD4 + CD25 T cells preferably CD3 + CD4 + CD25 " CD45RA + T cells, are isolated from PBMCs (peripheral blood mononuclear cells) by flow cytometry or by negative selection using a MACS system for example.
  • the CD3 + CD4 + CD25 T cells are CD62L + .
  • the CD3 + CD4 + CD25 T cells are CD127 + .
  • CD3 + CD4 + CD25 T cells are CD27 + .
  • the CD3 + CD4 + CD25 T cells are CD3 + CD4 + CD25 CD45RA + T cells, are CD62L + CD127 + .
  • the CD3 + CD4 + CD25 T cells are CD62L + CD27 + .
  • the CD3 + CD4 + CD25 T cells are CD3 + CD4 + CD25 CD45RA + T cells, are CD127 + CD27 + .
  • CD3 + CD4 + CD25 T cells are CD62L + CD127 + CD27 + .
  • CD3 + CD4 + CD25 T cells preferably CD3 + CD4 + CD25 CD45RA + T cells, are TCRy5 ⁇
  • the CD3 + CD4 + CD25 T cells are Va24 ⁇
  • the CD3 + CD4 + CD25 T cells may be isolated from frozen PBMCs.
  • the obtainment of isolated CD3 + CD4 + CD25 " T cells, preferably CD3 + CD4 + CD25 CD45RA + T cells may be improved by an optional first to a purification step.
  • the CD3 + CD4 + CD25 T cells, preferably CD3 + CD4 + CD25 " CD45RA + T cells are stimulated with antigen pulsed tolerogenic DCs (for example ovalbumin pulsed tolerogenic DCs) in the presence of soluble anti-CD28 and anti-CD40 antibodies.
  • antigen pulsed tolerogenic DCs for example ovalbumin pulsed tolerogenic DCs
  • the time of stimulation ranges between 1 hour and 24 hours, preferably between 10 hours and 20 hours, more preferably during about 16 hours.
  • cells are washed, for example with PBS, and stained with anti- CD 154 and anti-CD4 antibodies for sorting.
  • the purified CD3 + CD4 + CD25 CD154 + T cells are enriched and may be used for the following activation step.
  • the CD3 + CD4 + CD25 " T cells are activated in the presence of an aPTCR cell activator.
  • Said a TCR cell activator can be a polyclonal a TCR cell activator or an antigen-specific a TCR cell activator.
  • the polyclonal a ⁇ TCR cell activator is a TCR ⁇ activator.
  • TCR ⁇ activator include, but are not limited to, anti-TCR ⁇ antibody such as purified anti-human TCR ⁇ / ⁇ antibody (ref 306702, Biolegend), Anti-Human alpha beta TCR antibody (ref 11-9986-41, eBioscience), anti- human TCR ⁇ (ref 563826, BD Biosciences), TCR alpha/beta antibody (ref GTX80083, GeneTex); anti- CD3 antibody such as purified anti-human CD3 antibody (ref 344801, BioLegend), anti-CD3 antibody (ab5690, Abeam), anti-human CD3 purified (ref 14-0038-80, eBioscience), CD3 antibody (ref MA5- 17043, Invitrogen antibodies), CD3 monoclonal antibody (ref ALX-804-822-C100, Enzo Life Sciences), human CD3 antibody (ref 130- 09
  • the polyclonal TCRa- ⁇ cell activator is an anti-TCRa ⁇ antibody or an anti-CD3 antibody.
  • the polyclonal TCRa- ⁇ cell activator preferably the anti-TCRa ⁇ or anti-CD3 antibody, is soluble in the culture medium. In another embodiment, the polyclonal TCR ⁇ cell activator is coated to the culture plate.
  • the polyclonal TCRa- ⁇ cell activator is used in the presence of feeder cells, preferably autologous feeder cells.
  • Feeder cells include, but are not limited to, ACD3 cells (T cell-depleted accessory cells), irradiated PBMCs, irradiated DCs, artificial APCs (antigen presenting cells), Sf9 cells, insect cells, a pool of PBMCs or a pool of B cells from different subjects, KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
  • the feeder cells used in the invention are ACD3 cells that are isolated by negative selection from PBMCs by incubation with anti-CD3 coated beads and then irradiated at 3000 rad.
  • the ratio T cells / feeder cells ranges from about 1:100 to about 1:10000, preferably from 1:1000 to 1:5000.
  • the expression "from 1:100 to 1:10000” includes, without limitation 1:100, 1:200, 1:300, 1:400, 1:500, 1:600, 1:700, 1:800, 1:900, 1:1000, 1:1250, 1:1500, 1: 1750, 1:2000, 1:2250, 1:2500, 1:2750, 1:3000, 1:3250, 1:3500, 1: 3750, 1:4000, 1:4250, 1:4500, 1: 4750, 1:5000, 1:5250, 1:5500, 1:5750, 1:6000, 1:6250, 1:6500, 1:6750, 1:7000, 1:7250, 1 :7500, 1 :7750, 1 :8000, 1 :8250, 1 :8500, 1 : 8750, 1 :9000, 1 :9250, 1 :9500, 1
  • the antigen-specific TCRa cell activator is tolerogenic dendritic cells (DCs).
  • tolerogenic DCs refers to DCs capable to induce tolerance.
  • tolerogenic DCs are capable of secreting more suppressive cytokines such as IL-10 and TGF than proinflammatory cytokines such as IL-12, IL-23 or TNFa.
  • DCs are defined as tolerogenic when they secrete IL-10 and IL-12 in a ratio IL-10: IL-12 > 1.
  • tolerogenic DCs express on their surface the major histocompatibility (MHC) class la and/or MHC class lb.
  • MHC class la presentation refers to the "classical” presentation through HLA-A, HLA-B and/or HLA-C molecules whereas the MHC class lb presentation refers to the "non-classical" antigen presentation through HLA-E, HLA-F, HLA-G and/or HLA-H molecules.
  • tolerogenic DCs express 50% of MHC class la molecules and 50% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 45%> of MHC class la molecules and 55%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 40%> of MHC class la molecules and 60% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 35%> of MHC class la molecules and 65%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 30% of MHC class la molecules and 70% of MHC class lb molecules on their surface.
  • tolerogenic DCs express 25% of MHC class la molecules and 75% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 20% of MHC class la molecules and 80% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 15% of MHC class la molecules and 85% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 10% of MHC class la molecules and 90% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 5% of MHC class la molecules and 95% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express only MHC class lb molecules on their surface.
  • tolerogenic DCs express 50% of HLA-A, HLA-B and/or HLA-C molecules and 50% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 45% of HLA-A, HLA-B and/or HLA-C molecules and 55% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 40% of HLA-A, HLA-B and/or HLA-C molecules and 60% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 35% of HLA-A, HLA-B and/or HLA-C molecules and 65% of HLA-E molecules on their surface.
  • tolerogenic DCs express 30% of HLA-A, HLA-B and/or HLA-C molecules and 70% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 25% of HLA-A, HLA-B and/or HLA-C molecules and 75% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 20% of HLA-A, HLA-B and/or HLA-C molecules and 80% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 15% of HLA-A, HLA-B and/or HLA-C molecules and 85% of HLA-E molecules on their surface.
  • tolerogenic DCs express 10% of HLA-A, HLA-B and/or HLA-C molecules and 90% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 5% of HLA-A, HLA-B and/or HLA-C molecules and 95% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express only HLA-E molecules on their surface. Methods for obtaining tolerogenic DCs are well-known in the art. An exemplary method is the generation of tolerogenic DCs from CD14 + monocytes. For example, CD14 + monocytes are cultured in the presence of GM-CSF and IL-4, or in the presence of GM-CSF and IFNa, for the generation of immature DCs.
  • TAP transporter transporter associated with antigen processing
  • Exemplary methods to inhibit the TAP transporter in the endoplasmic reticulum include, but are not limited to, CRISPR-CAS-9 technology, silencing RNA, transfected DCs with the UL-10 viral protein from the CMV (cytomegalovirus) or the use of viral proteins.
  • viral proteins able to inhibit the TAP transporter include, but are not limited to, HSV-1 ICP47 protein, varicella- virus UL49.5 protein, cytomegalovirus US6 protein or gammaherpesvirus EBV BNLF2a protein.
  • Another method is the use of a chemical product to inhibit the expression of MHC class la molecules without changing HLA-E expression on the surface of tolerogenic DCs.
  • chemical products include, but are not limited to, 5'- methyl-5 '- thioadenosine or leptomycin B.
  • the tolerogenic DCs are pulsed in the presence of at least one self-peptide antigen, modified self-peptide antigen, over-expressed self-peptide antigen or foreign antigen.
  • self-peptide antigen is meant an antigen that is normally expressed in the body from which the regulatory T cells are derived.
  • self-antigen is comparable to one, or, in another embodiment, indistinct from one normally expressed in a body from which the regulatory T cells are derived, though may not directly correspond to the antigen.
  • self-antigen refers to an antigen, which when expressed in a body, may result in the education of self-reactive T cells.
  • self-antigen is expressed in an organ that is the target of an autoimmune disease.
  • the self-antigen is expressed in a pancreas, thyroid, connective tissue, kidney, lung, digestive system or nervous system.
  • self-antigen is expressed on pancreatic ⁇ cells.
  • self-peptide antigen, modified self-peptide antigen and over-expressed self-peptide antigen include, but are not limited to, antigenic peptides of insulin, insulin beta, glutamic acid decarboxylase 1 (GAD1), glutamic acid decarboxylase 65 (GAD 65), HSP, thyro globulin, nuclear proteins, acetylcholine receptor, collagen, thyroid stimulating hormone receptor (TSHR), ICA512(IA-2) and ⁇ -2 ⁇ (phogrin), carboxypeptidase H, ICA69, ICA12, thyroid peroxidase, native DNA, myelin basic protein, myelin proteolipid protein, acetylcholine receptor components, histocompatibility antigens, antigens involved in graft rejection and altered
  • the self-peptide antigen is derived from immunogenic apoptotic bodies from cancer cells or derived from tissue lysate.
  • Cancer cells may derive from tumor biopsy or from expansion of circulatory cancer cells.
  • Immunogenic apoptotic bodies from cancer cells may be obtained for example with anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin, UVC or ⁇ -radiation treated cancer cells releasing apoptotic bodies or can be directly isolated from anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin; UVC or ⁇ -radiation treated cancer.
  • anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone
  • oxaliplatin UVC or ⁇ -radiation treated cancer.
  • tissue lysate examples include, but are not limited to, synovial liquid or inflammatory tissue lysate.
  • foreign antigen is meant a molecule or molecules which is/are not endogenous or native to a mammal which is exposed to it.
  • the foreign antigen may elicit an immune response, e.g. a humoral and/or T cell mediated response in the mammal.
  • the foreign antigen will result in the production of antibodies there against.
  • foreign antigens include, but are not limited to, proteins (including a modified protein such as a glycoprotein, a mucoprotein, etc.), nucleic acids, carbohydrates, proteoglycans, lipids, mucin molecules, immunogenic therapeutic agents (including proteins such as antibodies, particularly antibodies comprising non-human amino acid residues, e.g.
  • rodent, chimeric/humanized, and primatized antibodies include toxins (optionally conjugated to a targeting molecule such as an antibody, wherein the targeting molecule may also be immunogenic), gene therapy viral vectors (such as retroviruses and adenoviruses), grafts (including antigenic components of the graft to be transplanted into the heart, lung, liver, pancreas, kidney of graft recipient and neural graft components), infectious agents (such as bacteria and virus or other organism, e.g., protists), alloantigens (i.e.
  • a targeting molecule such as an antibody, wherein the targeting molecule may also be immunogenic
  • gene therapy viral vectors such as retroviruses and adenoviruses
  • grafts including antigenic components of the graft to be transplanted into the heart, lung, liver, pancreas, kidney of graft recipient and neural graft components
  • infectious agents such as bacteria and virus or other organism, e.g., pro
  • an antigen that occurs in some, but not in other members of the same species) such as differences in blood types, human lymphocyte antigens (HLA), platelet antigens, antigens expressed on transplanted organs, blood components, pregnancy (Rh), and hemophilic factors (e.g. Factor VTfl and Factor IX).
  • HLA human lymphocyte antigens
  • platelet antigens antigens expressed on transplanted organs
  • Rh pregnancy
  • hemophilic factors e.g. Factor VTfl and Factor IX
  • the self-peptide antigen or the foreign antigen is soluble.
  • the cAMP activator added in the culture allows the activation of the cAMP pathway.
  • cAMP activator include, but are not limited to PGE2 (prostaglandin E2), an EP2 or EP4 agonist, a membrane adenine cyclase activator such as forskolin, or metabotropic glutamate receptors agonists.
  • PGE2 examples include, but are not limited to, PGE2 of ref P5640 or P0409 (Sigma- Aldrich), PGE2 of ref 2296 (R&D Systems), PGE2 of ref 2268 (Bio Vision), PGE2 of ref 72192 (Stemcell), PGE2 of ref abl44539 (Abeam ), and PGE2 of ref 14010 (Cayman Chemical).
  • the cAMP activator preferably PGE2 is used at a concentration ranging from 0.01 ⁇ to 10 ⁇ .
  • the expression "from 0.01 ⁇ to 10 ⁇ " includes, without limitation 0.02 ⁇ , 0.03 ⁇ , 0.04 ⁇ , 0.05 ⁇ , 0.06 ⁇ , 0.07 ⁇ , 0.08 ⁇ , 0.09 ⁇ , 0.1 ⁇ , 0.2 ⁇ , 0.3 ⁇ , 0.4 ⁇ , 0.5 ⁇ , 0.6 ⁇ , 0.7 ⁇ , 0.8 ⁇ , 0.9 ⁇ , 1 ⁇ , 1.5 ⁇ , 2 ⁇ , 2.5 ⁇ , 3 ⁇ , 3.5 ⁇ , 4 ⁇ , 4.5 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ .
  • PGE2 is at a concentration ranging from 0.03 ⁇ to 1.5 ⁇ .
  • the TGFP pathway activator added in the culture allows the activation of the TGFP pathway.
  • TGFP pathway activators include, but are not limited to, TGFP family (TGFpi, TGFP2, TGFP3), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, and nodal.
  • TGFP examples include, but are not limited to, TGFpi of ref T7039 (Sigma- Aldrich), TGFp2 of ref T2815 (Sigma- Aldrich), TGFp3 of ref T5425 (Sigma- Aldrich), human TGFpl of ref P01137 (R&D system), human TGFpl of ref 580702 (Biolegend), TGFpl of ref HZ-1011 (HumanZyme), human TGFpl of ref 14- 8348-62 (Affymetrix eBioscience).
  • the pathway activator is used at a concentration ranging from 1 ng/ml to 20 ng/ml.
  • the expression "from 1 ng/ml to 20 ng/ml” includes, without limitation 2 ng/ml, 2.5 ng/ml, 3 ng/ml, 3.5 ng/ml, 4 ng/ml, 4.5 ng/ml, 5 ng/ml, 5.5 ng/ml, 6 ng/ml, 6.5 ng/ml, 7 ng/ml, 7.5 ng/ml, 8 ng/ml, 8.5 ng/ml, 9 ng/ml, 9.5 ng/ml, 10 ng/ml, 11 ng/ml, 12 ng/ml, 13 ng/ml, 14 ng/ml, 15 ng/ml, 16 ng/ml, 17 ng/ml, 18 ng/ml, 19 ng/ml.
  • TGF is at a concentration ranging from 2.5 ng/ml to 7.5 ng/ml.
  • the mTOR inhibitor added in the culture allows the inhibition of the mTOR pathway.
  • mTOR inhibitor include, but are not limited to, rapamycin (also named sirolimus) and its analogs (termed rapalogs); wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG); curcumin; resveratrol; genistein; 3, 3-diindolylmethane (DIM); LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l- benzopyran-4-one); PP242; PP30; Torinl ; Ku-0063794; WAY-600; WYE-687; WYE- 354; and mTOR and PI3K dual-specificity inhibitors such as GNE477, NVP-BEZ235, PI-103, XL765 and WJD008.
  • rapamycin examples include, but are not limited to, rapamycin of ref R0395 (Sigma- Aldrich), rapamycin of ref SI 039 (Selleckchem), rapamycin of ref 1292 (Tocris), rapamycin of ref R-5000 (LC Laboratories), rapamycin of ref tlrl-rap (InvivoGen), rapamycin of ref abl20224 (Abeam), rapamycin of ref R0395 (Sigma-Aldrich).
  • Examples of compounds of the same chemical class than rapamycin used clinically include, but are not limited to, Everolimus (code name RAD001), Temsirolimus (code name CCI-779, NSC 683864), Zotarolimus (code name ABT-578).
  • the mTOR inhibitor preferably rapamycin
  • the expression "from 0.1 nM to 50 nM” includes, without limitation 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 26 nM, 27 nM, 28 nM, 29 nM
  • At least one cytokine selected from IL-2, IL-7, IL-15 and TSLP can be added in the culture.
  • IL-2 is used at a concentration ranging from 10 IU/ml to 1000 IU/ml.
  • the expression "from 10 IU/ml to 1000 IU/ml” includes, without limitation 15 IU/ml, 20 IU/ml, 25 IU/ml, 30 IU/ml, 35 IU/ml, 40 IU/ml, 45 IU/ml, 50 IU/ml, 55 IU/ml, 60 IU/ml, 65 IU/ml, 70 IU/ml, 75 IU/ml, 80 IU/ml, 85 IU/ml, 90 IU/ml, 95 IU/ml, 100 IU/ml, 150 IU/ml, 200 IU/ml, 250 IU/ml, 300 IU/ml, 350 IU/ml, 400 IU/ml, 450 IU/ml, 500 IU/ml, 550 IU/ml, 600 IU/m
  • IL-7 is used at a concentration ranging from 1 ng/ml to 100 ng/ml.
  • the expression "from 1 ng/ml to 100 ng/ml” includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
  • IL-15 is used at a concentration ranging from 1 ng/ml to 50 ng/ml.
  • the expression "from 1 ng/ml to 50 ng/ml” includes, without limitation 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml,
  • IL-15 is used at a concentration ranging from
  • TSLP is used at a concentration from ranging 1 ng/ml to 100 ng/ml.
  • the expression "from 1 ng/ml to 100 ng/ml” includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
  • neutralizing antibodies can be added to the culture to prevent the generation of other populations of regulatory T cells.
  • neutralizing antibodies include, but are not limited to, anti-IFNy, anti-IL-4, and/or anti-IL12 antibodies.
  • anti-IFNy antibodies examples include, but are not limited to, Affymetrix eBioscience (Ref 14-7318), R&D systems (Ref MAB285), Novus Biologicals (Ref AF- 485-NA).
  • anti-IL-4 antibodies include, but are not limited to, R&D Systems (Ref MAB304, MAB204, or MAB204), Affymetrix eBioscience (Ref 14-7048), GeneTex (Ref GTX10755).
  • anti-IL-12 antibodies include, but are not limited to, Affymetrix eBioscience (Ref 16-7129 or 16-8126), Biolegend (Ref 508803), R&D systems (Ref MAB219, AF-219, or AB-219).
  • the culture medium used in the culture of the invention comprises (i) one or more pH buffering system(s); (ii) inorganic salt(s); (iii) trace element(s); (iv) free amino acid(s); (v) vitamin(s); (vi) hormone(s); (vii) carbon/energy source(s).
  • inorganic salts include, but are not limited to, calcium bromide, calcium chloride, calcium phosphate, calcium nitrate, calcium nitrite, calcium sulphate, magnesium bromide, magnesium chloride, magnesium sulphate, potassium bicarbonate, potassium bromide, potassium chloride, potassium dihydrogen phosphate, potassium disulphate, di- potassium hydrogen phosphate, potassium nitrate, potassium nitrite, potassium sulphite, potassium sulphate, sodium bicarbonate, sodium bromide, sodium chloride, sodium disulphate, sodium hydrogen carbonate, sodium dihydrogen phosphate, di-sodium hydrogen phosphate, sodium sulphate and a mix thereof.
  • trace elements include, but are not limited to, cobalt (Co), copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), selenium (Se), zinc (Zn) and the salts thereof.
  • free amino acids include, but are not limited to, L-alanine, L-arginine, L- asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, taurine, L-threonine, L-tryptophan, L-tyrosine, L-valine and a mix thereof.
  • vitamins include, but are not limited to, biotin (vitamin H); D-calcium- pantothenate; choline chloride; folic acid (vitamin B9); myo-inositol; nicotinamide; pyridoxal (vitamin B6); riboflavin (vitamin B2); thiamine (vitamin Bl); cobalamin (vitamin B 12); acid ascorbic; a-tocopherol (vitamin E) and a mix thereof.
  • biotin vitamin H
  • D-calcium- pantothenate choline chloride
  • folic acid vitamin B9
  • myo-inositol nicotinamide
  • pyridoxal vitamin B6
  • riboflavin vitamin B2
  • thiamine vitamin Bl
  • cobalamin vitamin B 12
  • acid ascorbic a-tocopherol (vitamin E) and a mix thereof.
  • carbon/energy sources include, but are not limited to, D-glucose; pyruvate; lactate; ATP; creatine; creatine phosphate; and a mix thereof.
  • the culture medium is a commercially available cell culture medium, in particular selected in a group comprising the IMDM (Iscove's Modified Dulbecco's Medium) from GIBCO® or the RPMI 1640 medium from GIBCO®.
  • IMDM Iscove's Modified Dulbecco's Medium
  • the culture medium is a serum-free culture medium such as the AIM-V medium from GIBCO®, the X-VIVO 10, 15 and 20 media from LONZA.
  • the culture medium can be further supplemented with additional compound(s), in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
  • additional compound(s) in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
  • the culture medium is IMDM.
  • the culture medium comprises IMDM cell culture medium; from 1% (w/w) to 5% (w/w) of foetal bovine serum; from 10 IU/ml to 200 IU/ml of penicillin; from 10 IU/ml to 200 IU/ml of streptomycin; from 0.1 mM to 10 mM of a mixture of non-essential amino acids, in particular amino acids selected in a group comprising alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine; from 0.5 mM to 10 mM of glutamine from 10 mM to 25 mM of HEPES pH 7.6-7.8.
  • the culture for generating the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days.
  • the expression "at least 5 days” includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days.
  • a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium.
  • portion is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium.
  • fresh culture medium refers to a culture medium that has not been in contact with any CD3+ T cells.
  • the medium is a nTreg polarizing medium.
  • the inventors define a "nTreg polarizing medium" as a medium such as RPMI medium comprising at least one cAMP activator as described hereabove, at least one TGF pathway activator as described here above and at least one mTor inhibitor as described hereabove.
  • the "nTreg polarizing medium” refers to a RPMI medium comprising TGF , rapamycin and PGE2.
  • the medium is an inflammatory medium.
  • an "inflammatory medium” as a medium such as IMDM comprising inflammatory cytokines such as for example IL- ⁇ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml).
  • IL- ⁇ 10 ng/ml
  • IL-6 (30 ng/ml
  • IL-21 50 ng/ml
  • IL- 23 (30 ng/ml
  • IL-2 100 Ul/ml
  • the method for generating ex vivo MHCII restricted CD4 + Foxp3 + regulatory T cells comprises: culturing CD3 + CD4 + CD25 " T cells in the presence of autologous ACD3 feeder cells and coated anti-CD3 antibody and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, - thereby obtaining a population of MHCII restricted CD4 + Foxp3 + regulatory
  • T cells ex vivo generated.
  • the method for generating ex vivo MHCII restricted CD4 + Foxp3 + regulatory T cells comprises: culturing CD3 + CD4 + CD25 " T cells in the presence of tolerogenic DC that have been pulsed with at least one self-peptide antigen during about 24h and in the presence of ACD3 feeder cells and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, thereby obtaining a population of MHCII restricted CD4 + Foxp3 + regulatory T cells.
  • the present invention also relates to an ex vivo method of generation and expansion of MHCII restricted CD4 + Foxp3 + regulatory T cells, comprising: generating the MHCII restricted CD4 + Foxp3 + regulatory T cells as described here above, expanding the MHCII restricted CD4 + Foxp3 + regulatory T cells generated by contacting them in the presence of an TCRaP cell activator (preferably either autologous ACD3 feeder cells and coated anti-CD3 antibody or tolerogenic DC that have been pulsed with at least one self-peptide antigen during about 24h and in the presence of ACD3 feeder cells) and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator (preferably PGE2), ii) a TGF (Transforming growth factor beta) pathway activator (preferably TGFP), iii) a mTOR inhibitor (preferably rapamycin), and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15
  • the MHCII restricted CD4 + Foxp3 + regulatory T cell population generated ex vivo is isolated by flow cytometry based on the following phenotype: CD3 + TCRap + CD45RO + Foxp3 + .
  • the isolated MHCII restricted CD4 + Foxp3 + regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of a polyclonal ⁇ T cell activator.
  • polyclonal ⁇ T cell activator examples include, but are not limited to, mitogen such as PMA/ionomycin, super-antigen, anti-CD3 antibody...
  • the anti-CD3 monoclonal antibody is coated.
  • the polyclonal T cell activator can be used in the presence of feeder cells as described here above.
  • the isolated MHCII restricted CD4 + Foxp3 + regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of antigen-specific TCRaP cell activator as described here above.
  • the antigen-specific TCRaP cell activator can be used in the presence of feeder cells as described here above.
  • the culture for expanding the ex vivo generated MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days.
  • the expression "at least 5 days” includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days or more.
  • a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium.
  • portion is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium.
  • fresh culture medium refers to a culture medium that has not been in contact with any CD3+ T cells.
  • MHCII restricted CD4 + Foxp3 + regulatory T cells are generated ex vivo by culturing CD3 + TCR ap + CD45RA + , preferably CD3 + TCR a + CD45RA + CD25 " , T cells obtained from PBMCs by negative selection (5.10 3 cells/ml) in the presence of autologous ACD3 feeder cells (125 10 5 cells/ml) and coated anti-CD3 antibody (2 ⁇ g/ml) in the presence of PGE2 (1 ⁇ ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5.
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • PGE2 50 nM
  • TGF 5 ng/ml
  • Rapamycin 1 nM
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • MHCII restricted CD4 + Foxp3 + regulatory T cells are generated ex vivo by culturing CD3 + TCR a + CD45RA + T cells, preferably CD3 + TCR a + CD45RA + CD25 " , obtained from PBMCs by negative selection (5.10 3 cells/ml) in the presence of tolerogenic DCs, that have been pulsed with at least one self-peptide antigen during about 24h, and in the presence of ACD3 feeder cells (125 10 5 cells/ml), PGE2 (1 ⁇ ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5.
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • TGF 5 ng/ml
  • PGE2 50 nM
  • TGF 5 ng/ml
  • Rapamycin 1 nM
  • IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • tolerogenic DCs were obtained by culturing CD14 + monocytes isolated from PBMCs in the presence of AIMV supplemented with GMCSF (100 ng/ml) and IL-4 (10 ng/ml). At day 3 and 6, the medium is discarded and replaced by fresh medium comprising GM-CSF and IL-4. On day 6, the tolerogenic DCs are pulsed for 24 hours in the presence of self-peptide antigen.
  • the present invention relates to an isolated population of invariant Foxp3 + regulatory T cells having the following phenotype: CD3 + Va24 + Foxp3 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD 127 " CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD45RO + CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD 127 " CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD 161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD127 CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD45RO + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CD45RO + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD45RO + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD 127 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD 161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD127 " CD161 CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD 127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD 127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD45RO + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD 127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CTLA4 + CD45RO + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CTLA4 + CD45RO + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CD45RO + CD 127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD127 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD 127 " CD161 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD127 " CD161 " CD56 " .
  • the invariant Foxp3 + regulatory T cells are CD3 + Va24 + Foxp3 + CD4 + CD25 + CTLA4 + CD45RO + CD 127 " CD161 " CD 127 " .
  • the invariant Foxp3 + regulatory T cells are Va24 + Jal8 + .
  • the invariant Foxp3 + regulatory T cells CD3 + Va24 + Foxp3 + CD4 + express the TCR ⁇ chain. In one embodiment, the invariant Foxp3 + regulatory T cells CD3 + Va24 + Foxp3 + CD4 + express the TCR ⁇ 2 chain. In one embodiment, the invariant Foxp3 + regulatory T cells CD3 + Va24 + Foxp3 + CD4 + express the TCR ⁇ 4 chain. In one embodiment, the invariant Foxp3 + regulatory T cells CD3 + Va24 + Foxp3 + CD4 + express the TCR ⁇ 8 chain. In one embodiment, the invariant Foxp3 + regulatory T cells CD3 + Va24 + Foxp3 + CD4 + express the TCR ⁇ 5.1 chain.
  • the invariant Foxp3 + regulatory T cells of the invention are human cells. yd Foxp3 + regulatory T cells
  • the present invention also relates to ⁇ Foxp3 + regulatory T cells having the following phenotype: CD3 + TCRy5 + Foxp3 + .
  • the ⁇ Foxp3 + regulatory T cells of the invention express the V52 isotype.
  • the ⁇ Foxp3 + regulatory T cells of the invention express the Vy9 isotype.
  • the ⁇ Foxp3 + regulatory T cells of the invention express the Vy9V02 isotype.
  • the ⁇ Foxp3 + regulatory T cells of the invention do not express the ⁇ 2 isotype.
  • the ⁇ Foxp3 + regulatory T cells of the invention do not express the Vy9 isotype.
  • the ⁇ Foxp3 + regulatory T cells of the invention do not express the Vy9V02 isotype. In one embodiment, the ⁇ Foxp3 + regulatory T cells of the invention express the ⁇ 3 isotype. In one embodiment, the ⁇ Foxp3 + regulatory T cells of the invention express the ⁇ 4 isotype. In one embodiment, the ⁇ Foxp3 + regulatory T cells of the invention express the ⁇ 5 isotype. In one embodiment, the ⁇ Foxp3 + regulatory T cells of the invention express the ⁇ 6 isotype. In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy0 + CD25 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy0 + IL-1R1 " . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy0 + IL-6R " . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy0 + IL-23R " . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy0 + IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy0 + CTLA4 + . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD45RO + . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD127 + . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-1R1 " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CD45RO + . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CD127 + . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-1R1 " CTLA4 + . In one embodiment, said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-6R " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-23R " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-33R " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-1R1 " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-6R " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-23R " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL- 33R " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-lRl " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-6R " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-23R " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-33R " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-lR IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-lRl " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-lRl " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-6R " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL- 6R ⁇ IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD45RO + CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-lRl " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-6R " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-23R " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-33R " CTLA4 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-lRl " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-6R " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-23R " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-33R " CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-lRl " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-6R " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-23R " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-33R " CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-1R1 " IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-1R1 " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-1R1 " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-6R " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-6R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CD45RO + CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL- 1R1 " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-lRl " IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-6R " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-6R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-6R " IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-1R1 IL-23R IL-33R- .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-6R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-6R " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CTLA4 + CD45RO + CD127 + IL-lRl " IL-6R " IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-lRl " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-6R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL- 1R1 " IL-23R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-6R " IL-33R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 IL-6R IL-23R " .
  • said population of ⁇ Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + TCRy5 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 IL-6R IL-23R IL-33R-.
  • the present invention also relates to a population of MHCII restricted CD4 + Foxp3 + regulatory T cells having the following phenotype: CD3 + TCRa,p + Foxp3 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-1R1 " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-6R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-23R " . In one embodiment, said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-33R " . In one embodiment, said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CTLA4 + . In one embodiment, said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-lRl " CTLA4 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-6R " CTLA4 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-23R " CTLA4 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-33R " CTLA4 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-lRl " CD25 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-6R " CD25 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-23R " CD25 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-33R " CD25 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-lRl " CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-6R " CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-23R " CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-33R " CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-lRl " IL-6R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-lRl " IL-23R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-lRl " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-6R " IL- 23R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-6R " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + IL-23R " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CTLA4 + CD45RO + .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-23R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-lRl " IL-6R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-lRl " IL-23R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-lRl " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " IL- 23R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-23R " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-6R " IL-23R " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 IL-23R IL-33R ⁇
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-6R " IL-33R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 " IL-6R " IL-23R " .
  • said population of MHCII restricted CD4 + Foxp3 + regulatory T cells has the following phenotype: CD4 + Foxp3 + CD25 + CTLA4 + CD45RO + CD127 + IL-1R1 IL-6R-IL-23R IL-33R ⁇
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) at least equivalent to the Foxp3 MFI measured in naive regulatory T cells.
  • MFI median fluorescence intensity
  • na ' ive regulatory T cells refers to T cells having for phenotype Foxp3 + CD45RA + CD4 + CD25 + CD127 ⁇
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) of at least 2000.
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) of at least 2 or 3 fold the Foxp3 MFI measured in na ' ive regulatory T cells.
  • MFI median fluorescence intensity
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) of at least 2000, 3000, 4000, 5000, 10000, 20000, 30000, 40000, 50000, 60000, 70000.
  • MFI median fluorescence intensity
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention comprise at least 65% of the CD3 + Va24 + cells expressing Foxp3.
  • the expression "at least 65%” includes, without limitation 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 752%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 82%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and 100%.
  • the term "expression" may refer alternatively to the transcription of a molecule (i.e. expression of the mRNA) or to the translation (i.e. expression of the protein) of a molecule.
  • detecting the expression may correspond to an intracellular detection.
  • detecting the expression may correspond to a surface detection, i.e. to the detection of molecules expressed at the cell surface.
  • detecting the expression may correspond to an extracellular detection, i.e. to the detection of secretion.
  • detecting the expression may correspond to intracellular, surface and/or extracellular detections.
  • Methods for determining the expression level include, without limitation, determining the transcriptome (in an embodiment wherein expression relates to transcription of a molecule) or proteome (in an embodiment wherein expression relates to translation of a cytotoxic molecule) of cells.
  • the expression of the molecules is assessed at the mRNA level.
  • Methods for assessing the transcription level of a molecule are well known in the prior art. Examples of such methods include, but are not limited to, RT- PCR, RT-qPCR, Northern Blot, hybridization techniques such as, for example, use of microarrays, and combination thereof including but not limited to, hybridization of amplicons obtained by RT-PCR, sequencing such as, for example, next-generation DNA sequencing (NGS) or RNA-seq (also known as "Whole Transcriptome Shotgun Sequencing") and the like.
  • NGS next-generation DNA sequencing
  • RNA-seq also known as "Whole Transcriptome Shotgun Sequencing
  • determining the expression level of at least one molecule corresponds to detecting and/or quantifying binding of a ligand to a molecule.
  • said ligand is an antibody specific of said molecule, and the method of the invention comprises detecting and/or quantifying a complex formed between said antibody and said molecule.
  • the complex can be detected if the ligand has been for example, but not limited to, covalently coupled with a detectable molecule such as an antibody constant fragment (Fc) or a fluorescent compound (e.g. Cyanine dye, Alexa dye, Quantum dye, etc).
  • a detectable molecule such as an antibody constant fragment (Fc) or a fluorescent compound (e.g. Cyanine dye, Alexa dye, Quantum dye, etc).
  • Fc antibody constant fragment
  • a fluorescent compound e.g. Cyanine dye, Alexa dye, Quantum dye, etc.
  • the complex can also be detected if the ligand has been tagged with different means well known to the person skilled in the art.
  • a tag used with the invention can be a tag selected from the group comprising or consisting of Hemaglutinin Tag, Poly Arginine Tag, Poly Histidine Tag, Myc Tag, Strep Tag, S-Tag, HAT Tag, 3x Flag Tag, Calmodulin-binding peptide Tag, SBP Tag, Chitin binding domain Tag, GST Tag, Maltose-Binding protein Tag, Fluorescent Protein Tag, T7 Tag, V5 Tag and Xpress Tag.
  • the use of the ligand therefore allows on the one hand the identification and detection of the molecule depending on the ligand used, and on the other hand the quantification of the complex formed.
  • determining the expression level of molecules is conducted by flow cytometry, immunofluorescence or image analysis, for example high content analysis.
  • the determination of the expression level of molecules is conducted by flow cytometry.
  • cells are fixed and permeabilized, thereby allowing detecting intracellular proteins.
  • determining the expression level of a molecule in a cell population comprises determining the percentage of cells of the cell population expressing the molecule (i.e. cells "+" for the molecule). Preferably, said percentage of cells expressing the molecule is measured by FACS.
  • expressing (or +) and “not expressing (or -)” are well known in the art and refer to the expression level of the cell marker of interest, in that the expression level of the cell marker corresponding to "+” is high or intermediate, also referred as "+/-".
  • the cell marker corresponding to "-” is a null expression level of the cell marker or also refers to less than 10 % of a cell population expressing the said cell marker.
  • the expression level of the cell marker of interest is determined by comparing the Median Fluorescence Intensity (MFI) of the cells from the cell population stained with fiuorescently labeled antibody specific for this marker to the fluorescence intensity (FI) of the cells from the same cell population stained with fiuorescently labeled antibody with an irrelevant specificity but with the same isotype, the same fluorescent probe and originated from the same specie (referred as Isotype control).
  • the cells from the population stained with fiuorescently labeled antibody specific for this marker and that show equivalent MFI or a lower MFI than the cells stained with the isotype controls are not expressing this marker and then are designated (-) or negative.
  • the cells from the population stained with fiuorescently labeled antibody specific for this marker and that show a MFI value superior to the cells stained with the isotype controls are expressing this marker and then are designated (+) or positive.
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention do not present a regulatory T cells specific demethylated region (TSDR) of the gene Foxp3.
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention present a regulatory T cells specific demethylated region (TSDR) of the gene Foxp3.
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention present a percentage of demethylation of the TSDR of the gene FOXP3 superior to at least 30%, 40%>, 50%>.
  • a protocol for measuring promoter demethylation percentage is shown in the Material and Method part of the Examples.
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention present a percentage of enrichment of acetylated histone in Foxp3 promoter region superior to at least 10%>, 20%>, 30%>, 40%> or 50%>.
  • a protocol for measuring enrichment of acetylated histones in percentage is shown in the Material and Method part of the Examples.
  • the invariant Foxp3 + regulatory T cells of the invention, the ⁇ Foxp3 + regulatory T cells of the invention and the MHCII restricted CD4 + Foxp3 + regulatory T cells of the invention are capable of suppressive activity similar to the suppressive activity of na ' ive CD4 + CD25 + CD45RA + CD 127 " regulatory T cells. Determination of the suppressive activity of a cell population is well known in the art and can be performed by conventional assays such as the standard polyclonal cell-cell contact Treg suppression assay or the autologous MLR suppression assay as described in the Examples.
  • the present invention also relates to invariant Foxp3 + regulatory T cells, ⁇ Foxp3 + regulatory T cells and MHCII restricted CD4 + Foxp3 + regulatory T cells obtainable or obtained by the ex vivo generation method as described here above.
  • the present invention also relates to invariant Foxp3 + regulatory T cells, ⁇ Foxp3 + regulatory T cells and MHCII restricted CD4 + Foxp3 + regulatory T cells obtainable or obtained by the ex vivo generation and expansion method as described here above.
  • the populations of invariant Foxp3 + regulatory T cells, ⁇ Foxp3 + regulatory T cells and MHCII restricted CD4 + Foxp3 + regulatory T cells obtained by the generation and expansion method of the invention comprises at least 10 6 , 10 7 , 10 8 , 10 9 , 10 10 cells.
  • the populations of invariant Foxp3 + regulatory T cells, ⁇ Foxp3 + regulatory T cells and MHCII restricted CD4 + Foxp3 + regulatory T cells obtained by the generation and expansion method of the invention has the property to remain stable when placed in inflammatory conditions.
  • stable refers to no secretion or a low secretion of IL-17, i.e. inferior to 200 ng/ml, 100 ng/ml, 50 ng/ml and still capable of suppressive capacity, i.e. inhibiting proliferation of conventional T cells as shown in the Examples.
  • inflammatory condition refers to a medium enriched in aromatic acid, preferably in tryptophan, such as for example IMDM, comprising inflammatory cytokines such as for example IL- ⁇ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml).
  • a method for determining if a population of regulatory T cells remains stable in inflammatory condition comprises culturing the regulatory T cells in the inflammatory condition medium as described here above in the presence of anti-CD3 (4 ⁇ g/ml), preferably coated, and anti-CD28 (4 ⁇ g/ml), preferably in a soluble form.
  • IL-17 in the culture supernatant is measured.
  • the recognition of IL-17 in the culture supernatant may be carried out by conventional methods known in the art such as, for example, a sandwich ELISA anti-IL-17. Briefly, after coated the plate with a capture anti-IL-17 antibody, the culture supernatant is added to each well with a dilution series. After incubation, a detection anti-IL-17 antibody is added to each well.
  • the ELISA is developed by any colorimetric means known in the art such as, for example, using detection antibody labelled with biotin, a poly-streptavidin HRP amplification system and an o- phenylenediamine dihydro chloride substrate solution.
  • An IL-17 level inferior to 200 ng/ml, 100 ng/ml, 50 ng/ml corresponds to no secretion or low secretion of IL-17.
  • the inventors state that the stroma of malignant tumor cells comprises TILs (Tumor-infiltrating lymphocytes) that are highly enriched in regulatory T cells and that exert an immune suppressive activity, in particular on NK cells, which likely accounts on the local cancer immune escape.
  • TILs Tumor-infiltrating lymphocytes
  • the at least one inactivated regulatory T cells population may represent an antigenic target to induce an immune response directed against the at least one regulatory T cells population present in the TILs, thereby preventing their immune suppressive activity and allowing the cytotoxic activity of effector cells such as NK cells against the tumor cells.
  • the inventors thus suggest using a vaccine composition comprising as active principle at least one inactivated regulatory T cells population among peripheral regulatory T cells population, ⁇ regulatory T cells population and invariant regulatory T cells population.
  • One object of the invention is an immunogenic product comprising, consisting essentially of or consisting of at least one inactivated Foxp3 + regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population.
  • the immunogenic product comprises, consists essentially of or consists of at least one inactivated ex vivo generated Foxp3 + regulatory T cells population among an ex vivo generated MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated ⁇ Foxp3 + regulatory T cells population and an ex vivo generated invariant Foxp3 + regulatory T cells population.
  • the term "consisting essentially of, with reference to an immunogenic product, pharmaceutical composition, vaccine or medicament means that the at least one ex vivo generated Foxp3 + regulatory T cells population or antibody of the invention is the only one therapeutic agent or agent with a biologic activity within said immunogenic product, pharmaceutical composition, vaccine or medicament.
  • the immunogenic product comprises, consists essentially of or consists of at least one inactivated ex vivo generated and optionally expanded regulatory T cells population among an ex vivo generated and optionally expanded MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated and optionally expanded ⁇ Foxp3 + regulatory T cells population and an ex vivo generated and optionally expanded invariant Foxp3 + regulatory T cells population.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, consisting essentially of or consisting of the immunogenic product as described here above and at least one pharmaceutically acceptable excipient.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, consisting essentially of or consisting of at least one inactivated Foxp3 + regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, consisting essentially of or consisting of at least one inactivated ex vivo generated Foxp3 + regulatory T cells population among an ex vivo generated MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated ⁇ Foxp3 + regulatory T cells population and an ex vivo generated invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient.
  • excipient refers to any and all conventional solvents, dispersion media, fillers, solid carriers, aqueous solutions, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • preparations should meet sterility, pyrogenicity, general safety and purity standards as required by regulatory offices, such as, for example, FDA Office or EMA.
  • pharmaceutically acceptable is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the subject to which it is administered.
  • pharmaceutically acceptable excipient include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like or combinations thereof.
  • Vaccine composition include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like or combinations thereof.
  • Another object of the invention is a vaccine composition
  • a vaccine composition comprising, consisting essentially of or consisting of the immunogenic product as described here above.
  • Another object of the invention is a vaccine composition
  • a vaccine composition comprising, consisting essentially of or consisting of at least one inactivated regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population.
  • Another object of the invention is a vaccine composition
  • a vaccine composition comprising, consisting essentially of or consisting of at least one inactivated ex vivo generated regulatory T cells population among an ex vivo generated MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated ⁇ Foxp3 + regulatory T cells population and an ex vivo generated invariant Foxp3 + regulatory T cells population.
  • inactivated T cells refers to T cells that are viable but has reduced or no effector function, i.e. have lost any pathogenic potential.
  • cell surface markers of inactivated T cells include, but are not limited to, 7-Aminoactinomycin D (7- AAD), calreticulin and heat shock protein 90 (HSP-90). Therefore, inactivated T cells express 7-AAD and/or calreticulin and/or HSP-90.
  • the inactivated regulatory T cells of the invention have lost their suppressive activity but are still immunogenic.
  • An example of T cell effector function assay is, but not limited to, T-cell proliferation assay. T-cell proliferation may be assessed on fixed T cells versus non-fixed T cells.
  • the T- cell proliferation assay aims at determining the percentage of living proliferating cells in fixed versus non-fixed T cells by flow cytometry. After staining the T cells with CFSE, anti-CD3 antibody and 7-AAD, the living proliferating cells are defined as the CFSE low fraction in gated CD3 + 7-AAD " cells.
  • the regulatory T cells are inactivated by any method well-known in the art.
  • methods for inactivating cells include, but are not limited to, irradiation, preferably with about 2500 to 3000 rads and/or chemical inactivation such as exposure to cisplatin, carboplatin, oxaliplatin, mitomycine C or antracycline.
  • the vaccine composition of the invention further comprises at least one adjuvant.
  • adjuvant examples include, but are not limited to, ISA51; emulsions such as CFA, MF59, montanide, AS03 and AF03; mineral salts such as alum, calcium phosphate, iron salt, zirconium salt, and AS04; TLR ligands such as TLR2 ligands (such as outer-surface protein A or OspA), TLR3 ligands (such as poly I:C), TLR4 ligands (such as MPL and GLA), TLR5 ligands, TLR7/8 ligands (such as imiquimod), TLR9 ligands (such as CpG ODN); polysacharrides such as chitin, chitosan, a-glucans, ⁇ -glucans, fructans, mannans, dextrans, lentinans, inulin-based adjuvants (such as gamma inulin); TLR9 and STING ligands such as TLR9 and STING
  • the inactivated Foxp3 + regulatory T cells present in the immunogenic product, pharmaceutical composition or vaccine composition of the invention are human Foxp3 + regulatory T cells. In one embodiment, the inactivated Foxp3 + regulatory T cells present in the immunogenic product, pharmaceutical composition or vaccine composition of the invention are autologous Foxp3 + regulatory T cells. In one embodiment, the inactivated regulatory T cells present in the immunogenic product, pharmaceutical composition or vaccine composition of the invention are allogenic Foxp3 + regulatory T cells.
  • the immunogenic product, pharmaceutical composition or vaccine composition of the invention may be personalized for a patient.
  • a "personalized" immunogenic product or vaccine composition refers to the use of Foxp3 + regulatory T cells generated and expanded ex vivo with at least one patient specific epitope.
  • the Foxp3 + regulatory T cells to be used as immunogenic product or in the vaccine composition are generated and expanded ex vivo in the presence of apoptotic bodies of cancer cells obtained from the patient, thereby providing at least one patient specific epitope.
  • the immunogenic product, pharmaceutical composition or vaccine composition of the invention comprises, consists essentially of or consists of as active principle at least one inactivated Foxp3 + regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population.
  • the immunogenic product, pharmaceutical composition or vaccine composition of the invention comprises, consists essentially of or consists of at least 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 inactivated Foxp3 + regulatory T cells as active principle.
  • the immunogenic product, pharmaceutical composition or vaccine composition of the invention comprise, consist essentially of or consist of about 10 4 , 5xl0 4 , 10 5 , 5xl0 5 , 10 6 , 5xl0 6 , 10 7 , 5xl0 7 , 10 8 , 5xl0 8 , 10 9 , 5xl0 9 , 10 10 , inactivated Foxp3 + regulatory T cells as active principle.
  • the Foxp3 + regulatory T cells, the inactivated Foxp3 + regulatory T cells, the immunogenic product, the pharmaceutical composition or the vaccine composition of the invention are/is frozen.
  • the immunogenic product, pharmaceutical composition or vaccine composition of the invention may be administrated to the subject by subcutaneous, intramuscular, intraperitoneal or intravenous injection, or directly into the tumor.
  • the immunogenic product, pharmaceutical composition or vaccine composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times in a year.
  • Example of regime of administration includes, but is not limited to, administration of the immunogenic product or vaccine composition at day 0, 4 weeks after day 0, 8 weeks after day 0, 12 weeks after day 0 and 24 weeks after day O.
  • Another object of the invention is a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of at least one inactivated regulatory T cells population or of the immunogenic product, pharmaceutical composition or vaccine composition of the invention as described here above.
  • Another object of the invention is a method for eliciting an immune response against at least one Foxp3 + regulatory T cells population present in the TILs of a subject affected with a cancer, comprising administering to the subject a therapeutically effective amount of at least one inactivated Foxp3 + regulatory T cells population or of the immunogenic product, pharmaceutical composition or vaccine composition of the invention as described here above.
  • Another object of the invention is a method for inducing an immunogenic apoptosis of at least one Foxp3 + regulatory T cells population present in the TILs of a subject affected with a cancer, comprising administering to the subject a therapeutically effective amount of at least one inactivated Foxp3 + regulatory T cells population or of the immunogenic product, pharmaceutical composition or vaccine composition of the invention as described here above.
  • cancers that can be treated with the immunogenic product, pharmaceutical composition or vaccine composition of the invention include, but are not limited to, adrenocortical carcinoma, anal cancer, bladder cancer, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal, pineal tumors, hypothalamic glioma, breast cancer, carcinoid tumor, carcinoma, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, extrahepatic bile duct cancer, ewings family of tumors (pnet), extracranial germ cell tumor, eye cancer, intraocular melanoma, gallbladder cancer, gastric cancer, germ cell tumor, extragonadal, gestational trophoblastic tumor, head and neck cancer, hypopharyngeal cancer, islet cell carcinoma, laryngeal cancer, leukemia, acute lymphoblastic, leukemia, oral cavity cancer, liver cancer, lung cancer, small cell lymphoma, AIDS-related, lymph
  • Another object of the invention is a method for preparing the immunogenic product of the invention, comprising: identifying from a tumor sample or from TILs obtained from the subject the at least one regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population, that is overrepresented, ex vivo generating the at least one overrepresented regulatory T cells population, inactivating the at least one ex vivo generated regulatory T cell population.
  • the MHCII restricted CD4 + Foxp3 + regulatory T cells population is considered overrepresented when superior to 5 to 10 % of total cells present in the sample. In one embodiment, there is a positive correlation between the percentage of Foxp3 expression in MHCII restricted CD4 + Foxp3 + regulatory T cells in the sample and a poor clinical outcome for breast cancer.
  • the ⁇ Foxp3 + regulatory T cells population is considered overrepresented when superior to 5 to 10 % of total cells present in the sample. In one embodiment, there is a positive correlation between the percentage of Foxp3 expression in MHCII restricted CD4 + Foxp3 + regulatory T cells in the sample and a poor clinical outcome for breast cancer.
  • the invariant Foxp3 + regulatory T cells population is considered overrepresented when superior to 0.01 to 1 % of total cells present in the sample.
  • Another object of the invention is a method for treating cancer in a subject in need thereof, comprising administrating to the subject the immunogenic product, pharmaceutical composition or vaccine composition of the invention.
  • Another object of the invention is a method for treating cancer in a subject in need thereof, comprising:
  • the inventors suggest that the Foxp3 + regulatory T cells of the invention, which are committed to exert immune suppressive function, may be capable of inhibiting autoreactive pathogenic immune effector cells including CD4 + , CD8 + , B cells or innate NK cells, which, in turn, are no longer able to exert their cytotoxic properties towards the self-cells.
  • One object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one regulatory T cells population among a MHCII restricted CD4 + Foxp3 + regulatory T cells population, a ⁇ Foxp3 + regulatory T cells population and an invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient.
  • Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one ex vivo generated Foxp3 + regulatory T cells population among an ex vivo generated MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated ⁇ Foxp3 + regulatory T cells population and an ex vivo generated invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, consisting essentially of or consisting of at least one ex vivo generated Foxp3 + regulatory T cells population among an ex vivo generated and expanded MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated and expanded ⁇ Foxp3 + regulatory T cells population and an ex vivo generated and expanded invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising at least one ex vivo generated Foxp3 + regulatory T cells population among an ex vivo generated MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated ⁇ Foxp3 + regulatory T cells population and an ex vivo generated invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated regulatory T cells population remains stable when placed in inflammatory condition as described here above.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising at least one ex vivo generated Foxp3 + regulatory T cells population among an ex vivo generated and expanded MHCII restricted CD4 + Foxp3 + regulatory T cells population, an ex vivo generated and expanded ⁇ Foxp3 + regulatory T cells population and an ex vivo generated and expanded invariant Foxp3 + regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated and expanded regulatory T cells population remains stable when placed in inflammatory condition as described here above.
  • One object of the invention is the at least one ex vivo generated Foxp3 + regulatory T cells population or the pharmaceutical composition as described here above for use in adoptive therapy.
  • Another object of the invention is the at least one ex vivo generated Foxp3 + regulatory T cells population or the pharmaceutical composition as described here above for use in treating inflammatory or autoimmune diseases.
  • One object of the invention is the at least one ex vivo generated and expanded Foxp3 + regulatory T cells population or the pharmaceutical composition as described here above for use in adoptive therapy.
  • Another object of the invention is the at least one ex vivo generated and expanded Foxp3 + regulatory T cells population or the pharmaceutical composition as described here above for use in treating inflammatory or autoimmune diseases.
  • inflammatory or autoimmune diseases include, but are not limited to, acute disseminated encephalomyelitis, acute necrotizing haemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing spondylitis, anti-GBM/Anti-TBM nephritis, antiphospho lipid syndrome, autoimmune angioedema, autoimmune aplastic anaemia, autoimmune dysautonomia, autoimmune haemo lytic anaemia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease, autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura, autoimmune thyroid disease, autoimmune urticaria, axonal and neuronal neuropathies, Balo disease, Behcet's disease,
  • inflammatory or autoimmune diseases include, but are not limited to, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis.
  • Another object of the invention is the at least one ex vivo generated Foxp3 + regulatory T cells population or the pharmaceutical composition as described here above for use in preventing transplant rejection, graft versus host disease (GVHD).
  • GVHD transplant rejection, graft versus host disease
  • Another object of the invention is the at least one ex vivo generated and expanded Foxp3 + regulatory T cells population or the pharmaceutical composition as described here above for use in preventing transplant rejection, graft versus host disease (GVHD).
  • GVHD transplant rejection, graft versus host disease
  • the pharmaceutical composition of the invention comprises, consists essentially of or consists of at least 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 ex vivo generated Foxp3 + regulatory T cells as active principle.
  • the pharmaceutical composition of the invention comprises, consists essentially of or consists of about 10 4 , 5x10 4 , 10 5 , 5x10 5 , 10 6 , 5x10 6 , 10 7 , 5xl0 7 , 10 8 , 5xl0 8 , 10 9 , 5xl0 9 , 10 10 ex vivo generated Foxp3 + regulatory T cells as active principle.
  • the at least one ex vivo generated Foxp3 + regulatory T cells population or the pharmaceutical the invention are/is frozen.
  • the at least one ex vivo generated Foxp3 + regulatory T cells population present in the pharmaceutical composition of the invention is generated from human T cells.
  • the at least one ex vivo generated Foxp3 + regulatory T cells population present in the pharmaceutical composition of the invention is autologous.
  • the at least one ex vivo generated Foxp3 + regulatory T cells population present in the pharmaceutical composition of the invention is allogenic.
  • the pharmaceutical composition of the invention may be administrated to the subject by subcutaneous, intramuscular, intraperitoneal or intravenous injection.
  • the pharmaceutical composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times per week.
  • the pharmaceutical composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times per month.
  • the pharmaceutical composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times per 3 months.
  • Another object of the invention is a method for treating inflammatory or autoimmune diseases in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of at least one ex vivo generated Foxp3 + regulatory T cells or the pharmaceutical composition as described here above.
  • T cell vaccination induces regulatory networks that specifically suppress the immunogenic T cells by activating T cells specific for a clono type-specific determinant (anti- idiotypic response).
  • anti-ergotypic responses directed at activation markers may also partially account for the suppression of the regulatory T cell population targeted.
  • Another object of the invention is an antibody recognizing the TCR (T cell receptor) of the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention.
  • the antibody recognizing the TCR of the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention recognizes at least one of the CDR1, CDR2 and CDR3 (complementary determining region 1, 2 and 3) of the TCR.
  • the antibody recognizing the TCR of the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention recognizes the CDR3 of the TCR.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, consisting essentially of or consisting of said antibody and at least one pharmaceutically acceptable excipient.
  • Another object of the invention is the use of said antibody for treating cancer in a subject in need thereof.
  • the antibodies directed against the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention consist of antibodies produced following immunization of a mammal, including a human, with the immunogenic composition as described here above.
  • the antibodies may also be obtained by cloning the relevant DNA material encoding them, starting for example from B cells obtained from the said mammal, including from the said human.
  • the antibodies may also be obtained by sequencing the amino acid sequences of the antibodies collected from the said mammal, including from the said human, and then synthesize a DNA molecule encoding the antibody or a portion thereof comprising the CDR thereof, for producing relevant recombinant antibodies directed against the peripheral regulatory T cells of the invention.
  • Preparing antibodies directed against the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention by immunization with the immunogenic composition of the invention may be easily performed by a skilled in the art, using the common technical knowledge from the state in the art.
  • the antibodies directed against the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention may be obtained after immortalization of the human B lymphocytes producing them; their cDNA can also be cloned and used further for producing them or their derivatives through recombinant DAN technology.
  • antibody herein is used to refer to a molecule having a useful antigen binding specificity. Those skilled in the art will readily appreciate that this term may also cover polypeptides which are fragments of or derivatives of antibodies yet which can show the same or a closely similar functionality. Such antibody fragments or derivatives are intended to be encompassed by the term antibody as used herein.
  • antibody or “antibody molecule” for the purpose of passive immunotherapy, it is intended herein not only whole immunoglobulin molecules but also fragments thereof, such as Fab, F(ab')2, Fv and other fragments thereof that retain the capacity to bind and inactivate the peripheral regulatory T cells.
  • the term antibody includes genetically engineered derivatives of antibodies such as single chain Fv molecules (scFv) and domain antibodies (dAbs).
  • an antibody directed against the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention consists of a polyclonal antibody.
  • an antibody directed against the at least one ex vivo generated Foxp3 + regulatory T cells population of the invention consists of a monoclonal antibody.
  • monoclonal antibody is used herein to encompass any isolated Ab's such as conventional monoclonal antibody hybridomas, but also to encompass isolated monospecific antibodies produced by any cell, such as for example a sample of identical human immunoglobulins expressed in a mammalian cell line.
  • variable heavy (VH) and variable light (VL) domains of the antibody are involved in antigen recognition, a fact first recognized by early protease digestion experiments. Further confirmation was found by "humanization" of rodent antibodies. Variable domains of rodent origin may be fused to constant domains of human origin such that the resultant antibody retains the antigenic specificity of the rodent parented antibody (Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81, 6851-6855). That antigenic specificity is conferred by variable domains and is independent of the constant domains is known from experiments involving the bacterial expression of antibody fragments, all containing one or more variable domains.
  • Fab-like molecules (Better et al (1988) Science 240, 1041); Fv molecules (Skerra et al (1988) Science 240, 1038); single-chain Fv (ScFv) molecules where the V.sub.H and V.sub.L partner domains are linked via a flexible oligopeptide (Bird et al (1988) Science 242, 423; Huston et al (1988) Proc. Natl. Acad. Sci. USA 85, 5879) and single domain antibodies (dabs) comprising isolated V domains (Ward et al (1989) Nature 341, 544).
  • ScFv molecules encompasses molecules wherein the VH and VL partner domains are linked via a flexible oligopeptide.
  • Engineered antibodies, such as ScFv antibodies can be made using the techniques and approaches described in J. Huston et al, (1988) "Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single chain Fv analogue produced in E. coli", Proc. Natl. Acad. Sci. USA, 85, pp. 5879-5883, and in A. Pluckthun, (1991) "Antibody engineering; Advances from use of E. coli expression systems", Bio/technology 9 (6): 545-51, incorporated herein by reference.
  • Suitable monoclonal antibodies which are reactive as described herein may be prepared by known techniques, for example those disclosed in “Monoclonal Antibodies; A manual of techniques", H Zola (CRC Press, 1988) and in “Monoclonal Hybridoma Antibodies: Techniques and Application", S G R Hurrell (CRC Press, 1982).
  • a further embodiment encompasses humanized antibodies where the regions of the murine antibody that contacted the antigen, the Complementarity Determining Regions (CDRs) were transferred to a human antibody framework.
  • CDRs Complementarity Determining Regions
  • Such antibodies are almost completely human and seldom cause any harmful antibody responses when administered to patients.
  • Several chimeric or humanized antibodies have been registered as therapeutic drugs and are now widely used within various indications (Borrebaeck & Carlsson, 2001, Curr. Opin. Pharmacol. 1 : 404-408). It is preferred if the antibody is a humanized antibody.
  • Suitably prepared non- human antibodies can be "humanized” in known ways, for example by inserting the CDR regions of mouse antibodies into the framework of human antibodies. Humanized antibodies can be made using the techniques and approaches described in Verhoeyen et al (1988) Science, 239, 1534-1536, and in Kettleborough et al, (1991) Protein Engineering, 14 (7), 773-783.
  • antibodies also encompass completely human antibodies, which may be produced using recombinant technologies.
  • large libraries comprising billions of different antibodies are used.
  • this technology does not rely on immunization of animals to generate the specific antibody.
  • the recombinant libraries comprise a huge number of pre-made antibody variants wherein it is likely that the library will have at least one antibody specific for any antigen.
  • the frequency of administration may be determined clinically by following the decline of antibody titers in the serum of patients over time, but in any event may be at a frequency of 1 to 52 times per year, and most preferably between 1 and 12 times per year.
  • Quantities of antibody may vary according to the severity of the disease, or half- life of the antibody in the serum, but preferably will be in the range of 1 to 10 mg/kg of patient, and preferably within the range of 1 to 5 mg/kg of patient, and most preferably 1 to 2 mg/kg of patient.
  • Figure 1 Different frequencies and phenotypic characteristics between FOXP3 + and FOXP3 " CD3 + T cell populations, as defined by their variable TCR recognition in human peripheral blood (PBMCs) and in TIL isolated from breast tumor.
  • PBMCs peripheral blood
  • FIG. 2 Analysis of Foxp3 + expression in lymphocytes present in the TILs extracted from luminal A and B breast subtypes. Tumor tissue from patient with luminal-A and luminal B was minced with scalpels and enzymatically digested by overnight incubation in collagenase Type IV. Expression of FOXP3 marker in lymphocytes present in the isolated TIL was determined by flow cytometric analysis. Representation of the Foxp3 expression level by the MFI in the CD3 + CD4 + TCRaP + restricted T cells and in the CD3 + CD4 + TCRy5 + unrestricted T cells.
  • FIG. 3 Positive correlation of Foxp3 + expression in lymphocytes present in the TILs and a poor clinical outcome in breast cancer.
  • Expression of FOXP3 marker in lymphocytes present in the isolated TIL was determined by flow cytometric analysis. Representation of the percentage of FOXP3 expression in the CD3 + CD4 + TCRaP + restricted T cells and in the CD3 + CD4 + TCRy5 + unrestricted T cells.
  • FIG 4 Multiparametric flow cytometry analysis of lymphocytes present in the TILs from luminal A and B breast subtypes. Lymphocytes present in the TIL were stained at the cell surface using Abs directed against CD3, CD4, CD25, CD56, CD161. After fixation and permeabilization Foxp3 and CTLA4 were stained intracellularly.
  • Figure 5 Phenotype and functional suppressive capacity of ex vivo generated Ag specific CD3 + TCRy6 + T cells from stimulated naive CD3 + TCRy6 + T cells.
  • Naive CD3 + TCRy5 + T cells were stimulated with zoledronic acid-treated-autologous tDCs, in presence of the nTreg polarizing medium and IL-2 (100 IU/ml) and IL-15 (10 ng/ml).
  • IL-2 100 IU/ml
  • IL-15 10 ng/ml
  • FIG. 6 In vitro induction of tumor- Ag specific CD3 + TCR Va24 + CD 1 -restricted T cells (invTreg) from stimulated naive CD3 + TCR Va24 + T cells with different nTreg polarizing medium.
  • Naive CD3 + TCR Va24 + T cells were stimulated for 21 days with tumor - apoptotic breast tumor cell line pulsed autologous tDC as described in Fig 3 in presence of IL-2 (100 IU/ml) and IL-15 (10 ng/ml). Where indicated, TGF , RAP A and PGE2 were added.
  • FIG. 7 Combination of TGF , RAPA and PGE2 induce the establishment and the expansion of tumor Antigen specific FOXP3 + CD3 + TCR Va24 + CD 1 -restricted T cell cells committed to exclusively exert regulatory activity, with an autologous MLR assay.
  • CD3 + TCR Va24 + CD45RA + T cells were stimulated with autologous tolerogenic DC pulsed with apoptotic breast tumor cell lines in presence of IL-2, IL-15 and nTreg polarizing medium. After 21 days of in vitro expansion in nTreg polarizing medium, suppressive capacity of ex vivo generated Tumor Ag-specific invariant Foxp3 + Treg was evaluated in the presence of (A) a low or (B) high inflammatory medium. Fresh na ' ive Treg were used as control.
  • FIG. 8 Analysis of Foxp3 + expression in human MHCII restricted CD4 + Foxp3 + CD4 + regulatory T cells (Treg) generated ex vivo from polyclonally stimulated naive CD4 + T cells with different nTreg polarizing medium.
  • Naive CD4 + T cells were stimulated for 12 days with plate-bound anti-CD3 (4 ⁇ g/ml) in presence of IL-2 (100 IU/ml). Where indicated, TGF (5 ng/ml), RAPA (10 nM) and PGE2 (1 ⁇ ) were added.
  • A Overlay histogram displaying Foxp3 expression profiles of each of the generated pTreg.
  • FIG. 9 Comparative analysis of in vitro suppressive capacity of human Treg generated with different nTreg polarizing medium. Suppressive capacity of ex vivo generated Treg was evaluated (A) in quiescent and (B) in inflammatory context with the standard polyclonal nTreg assay. CFSE-labeled conventional T cells (Tconv) were cocultured with ex vivo generated Treg at different ratio. Percent inhibition of TconvCFSE proliferation by Treg was depicted. Fresh Treg and Tconv were used as control.
  • FIG. 10 Combination of TGF , RAPA and PGE2 induce the establishment and the expansion of cultured Treg committed to exclusively exert regulatory activity. After 21 days of ex vivo generation in nTreg or TH-17 polarizing medium, suppressive capacity of ex vivo generated OVA- specific Treg was evaluated in the presence of a high inflammatory context inducing medium. Fresh Treg were used as control.
  • FIG. 11 IL-17 production by stimulated OVA- ex vivo generated Treg.
  • Specific- Treg (A) induced after the first 21 days of culture in nTreg polarizing medium or (B) expanded for 3 weeks in nTreg or TH-17 polarizing medium were tested for their IL-17- producing capacity upon stimulation with aCD3 Ab and aCD28 Ab for 2 days in IMDM medium containing IL-2, IL-1, IL-6, IL-21, and IL-23 cytokines.
  • IL-17 was detected in supernatant culture by ELISA.
  • Tumor tissue sample originated from patient with luminal A and Luminal B Breast cancer (Institut Jean Godinot, Reims). Cell Purification and Culture.
  • PBMCs Peripheral blood mononuclear cells
  • PBMCs are isolated by density gradient centrifugation on Ficoll-Hypaque (Pharmacia). PBMCs are used either as fresh cells or stored frozen in liquid nitrogen.
  • T-cell subsets and T cell-depleted accessory cells are isolated from either fresh or frozen PBMCs.
  • T cell-depleted accessory cells are isolated by negative selection from PBMCs by incubation with anti-CD3-coated Dynabeads (Dynal Biotech) and are irradiated at 3000 rad (referred to as ACD3-feeder).
  • CD4 + T cells are negatively selected with a CD4 + T-cell isolation kit (Miltenyi Biotec, yielding CD4 + T-cell populations at a purity of 96-99%. Subsequently, selected CD4 + T cells are labeled with anti-CD4 (13B8.2)-FITC (Beckman Coulter), anti-CD25(4E3)- APC (Miltenyi Biotec), and anti-CD127(R34.34)-PE (Beckman Coulter) before being sorted into CD4 + CD127- /lo CD25 high (pTregs) and CD4 + CD127 + CD25 neg/dim [conventional helper CD4 T cells (Tconv)] subpopulations using a F ACS Aria III Cell Sorter (Becton Dickinson).
  • CD14 + monocytes are isolated from PBMCs by positive selection using a MACS system.
  • CD3 + CD4 + CD127 + CD45RA + CD25 TCRaP + MHCII restricted are isolated from PBMCs after magnetic enrichment (MACS system: CD4 microbeads) and FACs sorting. Before the sorting step, enriched CD3 + CD4 + T cells are stained with anti-CD4 (13B8.2)-FITC (Beckman Coulter), anti-CD25(4E3)- APC (Miltenyi Biotec), and anti-CD127(R34.34)-PE (Beckman Coulter), anti-TCR ⁇ - BV421 (IP26) (Bio legend).
  • CD3 + CD45RA + invTCR Va24 + CD 1 -restricted T cells are isolated from PBMCs after magnetic enrichment (MACS system: anti-iNKT microbeads) and FACS sorting. Before the sorting step, enriched CD3 + invTCR Va24 + T cells are stained with anti-CD3 (UCHT-1) V450 anti-invariant TCR Va24-JaQ (6B11)-PE (inv TCR Va24-JaQ (Becton Dickinson) and anti-CD45RA (T6D11)-FITC (Miltenyi Biotec).
  • CD3 + CD45RA + CD27 + TCRy5 + unrestricted T cells are isolated from PBMCs after magnetic enrichment (MACS system: TCRy5 + T cell isolation kit) and FACS sorting. Before the sorting step, enriched CD3 + TCRy5 + T cells are stained with anti-CD3 (UCHT-1) V450, anti- TCR pany5 + PE (IMMU510) (Beckman Coulter), anti-CD27- APC efluor 780 (0323) (ebioscience) and anti-CD45RA (T6D11)-FITC (Miltenyi Biotec).
  • T cell subsets are cultured either in IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES (IMDM-5 media) in hypoxia 2%.
  • Breast cancer cell line and culture The human breast cancer cell line MCF-7 was obtained from the American Type Culture Collection (USA). Cells are maintained in Dulbecco's modified Eagle's medium (DMEM; Invitrogen, USA) supplemented with 10% fetal bovine serum (FBS). MCF-7 cells are treated with 5 ⁇ g/ml Doxorubicin for 24 h or by ⁇ irradiation (20 Gy). Extent of apoptosis is monitored by flow cytometric analysis (FACS). Cells are extensively washed prior to feeding DCs.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • TIL isolation Tumor tissue was minced with scalpels and enzymatically digested by overnight incubation in collagenase Type IV (2 mg/ mL, Roche Diagnostic GmbH) in DMEM High Glucose medium supplemented with 2 mM glutamine (Gibco), 50 mg/mL gentamycin and 0.25 % Human Serum Albumin, at 37 °C on a rotary shaker. Ex vivo generation of polyclonal functionally committed FOXP3 expressing regulatory T cells.
  • T cells are seeded at 2.5 10 5 /well in 48- well plates and stimulated with plate-bound anti-CD3 mAb (4 ⁇ g/ml) in the presence of ACD3-feeder (1 M).
  • Cells are cultured in IMDM-5 media (IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES) with PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM.
  • IL-2 (lOOIU/ml) are added to the culture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml).
  • these CD4 + T-cell lines were further expanded by restimulation with plate-bound anti-CD3 Abs (4 ⁇ / ⁇ 1). The restimulations were performed in the presence of ACD3-feeder, PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM and IL-2 (100 Ul/ml). Then every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml).
  • the phenotype of the expanded CD4 + T cells was assessed by flow cytometry. 75% of the stimulated naive conventional T cells that became CD45RO + express FOXP3 + .
  • T cells are seeded at 1 x 10 3 /well in 96-well plates and stimulated with plate-bound anti-inv TCR Va24-JaQ (6B11) mAb (2 ⁇ / ⁇ 1) in the presence of ACD3- feeder (2.5x10 5 ).
  • Cells are cultured in IMDM-5 media with PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Every three days, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture.
  • T cells are further expanded by restimulation with plate-bound anti- anti-inv TCR Va24-JaQ (6B11) mAb (2 in the presence of ACD3-feeder, PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM IL- 2 (100 Ul/ml) and IL-15 (10 ng/ml). Then every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). On day 21, cells are analyzed by flow cytometry. 70% of the stimulated CD3+ invTCR Va24 + RA + T cells that became CD45RO + express Foxp3 + .
  • T cells are seeded at 1 x 10 3 /well in 96-well plates and stimulated with plate-bound anti-TCRy5 mAb (2 ⁇ g/ml) in the presence of ACD3-feeder (2.5X10 5 ).
  • IMDM-5 media IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES
  • PGE2 1 ⁇ PGE2 1 ⁇
  • TGF 5 ng/ml TGF 5 ng/ml
  • Rapa 10 nM IL- 2 (100 Ul/ml)
  • IL-15 10 ng/ml
  • T cells were further expanded by restimulation with plate-bound anti-pan TCR ⁇ Abs (2 ⁇ g/ml).
  • the restimulations were performed in the presence of ACD3-feeder, PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM and IL-2 (100 Ul/ml) and IL-15 (10 ng/ml).
  • ACD3-feeder PGE2 1 ⁇
  • TGF 5 ng/ml TGF 5 ng/ml
  • Rapa 10 nM and IL-2 100 Ul/ml
  • IL-15 10 ng/ml
  • cells are analyzed by flow cytometry. 65% of the stimulated CD3 + CD45RA + CD27 + TCRy5 + T cells that became CD45RO + express Foxp3 + .
  • Tol-Mo-DC are 1) removed from the wells, washed twice with IMDM-5 (IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES, 2) added to wells of a 48-well plate at a concentration of 3 X 10 5 /ml in IMDM-5 and 3) pulsed in IMDM-5 with specific
  • ovalbumin pulsed tDC are 1) washed twice with IMDM-5 and 2) added to wells of a 48-well plate at a concentration of 3 X 10 5 /ml in IMDM-5 in the presence of 2 X 10 5 irradiated autologous feeders, PGE2 1 ⁇ , and Rapa 10 nM.
  • Purified naive conventional CD4 + T cells are added to the pulsed tDC.
  • T cell cloning medium On day 1 , IL-2 (lOOIU/ml) and TGF (5ng/ml) are added to the coculture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml (T cell cloning medium). On day 12, these T-cells are further expanded by restimulation with ova-pulsed tDC in the presence of ACD3-feeder, PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM, IL-2 (100 Ul/ml). Once T cells begin to expand, they can be split every 2 to 3 days with T cell cloning medium and irradiated feeder. On day 21, cells are analyzed by flow cytometry.
  • nTreg polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin
  • TH-17 polarizing medium IMDM medium containing IL-2 IL-1 IL-6, IL-21 IL- 23 cytokines
  • the 21 -day-expanded- Foxp3 expressing CD3 + CD4 + TCRa,p + MHCII restricted T cells are stimulated with plate-bound anti-CD3 mAb (4 ⁇ g/ml) in the presence of ACD3-feeder (1 M) in 48-well plates and every three days, half of the supernatant volume is discarded and replaced with fresh T cell cloning medium or TH-17 polarizing medium for 21 days.
  • tDC tumor-loaded Tolerogenic DC from CD14 + monocytes
  • monocytes are cultured in 48-well flat-bottom plates containing 0,5 ml of AIMV per well supplemented with 100 ng/ml recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF) and 10 ng/ml human recombinant IL-4 and AM580 (100 nM) for the generation of immature DC expressing CD Id.
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • AM580 100 nM
  • tDCs are co-cultured with apoptotic MCF-7 cells at a DC/tumor cell ratio of 1 :2 for 24h in AIMV with GM-CSF (100 ng/niL), IL-4 (10 ng/mL).
  • Another portion of tDC are freezed at 2 x 10 6 / per vial vial- in 90% FBS -10% DMSO.
  • tumor-antigen pulsed tDC are 1) washed twice with IMDM-5 and 2) added to wells of a 48-well plate at a concentration of 3 X 10 5 /ml in
  • IMDM-5 in the presence of 2 X 10 5 irradiated autologous feeders, PGE2 1 ⁇ , and Rapa 10 nM.
  • Purified CD3 + CD45RA + invTCR Va24 + CD1- restricted T cells are added to the pulsed tDC.
  • IL-2 (lOOIU/ml)
  • IL-15 (10 ng/ml)
  • TGF (5ng/ml)
  • T-cells are further expanded by restimulation with tumor Ag-pulsed tDC in the presence of ACD3-feeder, PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM, IL- 2 (100 Ul/ml) and IL-15 (10 ng/ml).
  • ACD3-feeder PGE2 1 ⁇
  • TGF 5 ng/ml TGF 5 ng/ml
  • Rapa 10 nM IL- 2 (100 Ul/ml)
  • IL-15 10 ng/ml
  • Tolerogenic DC from CD14 + monocytes
  • Tol-Mo-DC monocytes are cultured in 48-well flat-bottom plates containing 0,5 ml of AIMV per well supplemented with 100 ng/ml recombinant human granulocyte- macrophage colony- stimulating factor (GM-CSF) and 10 ng/ml human recombinant IL-4 for the generation of immature DC.
  • GM-CSF granulocyte- macrophage colony- stimulating factor
  • IL-4 human recombinant IL-4
  • IMDM-5 IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES, freezed or used for the generation and expansion of phospho- antigen specific functionally committed FOXP3 expressing CD3 + TCRy ⁇ T unrestricted T cells.
  • IL-2 (lOOIU/ml), IL-15 (10 ng/ml) and TGF (5ng/ml) are added to the coculture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) (T cell cloning medium). On day 12, these T-cells are further expanded by restimulation with tDC in the presence of ACD3-feeder, PGE2 1 ⁇ , TGF 5 ng/ml, Rapa 10 nM, IL-2 (100 Ul/ml), IL-15 (10 ng/ml) and zoledronic acid (100 nM).
  • T cells Once T cells begin to expand, they can be split every 2 to 3 days with T cell cloning medium and irradiated feeder. On day 21, cells are analyzed by flow cytometry. 75 % of the stimulated CD3 + CD45RA + TCRy5 + T cells that became CD45RO + express Foxp3 + .
  • monocytes are cultured in 48- well flat-bottom plates containing 0.5 ml of RPMI-5 per well supplemented with 20 ng/ml recombinant human granulocyte-macrophage colony- stimulating factor (GM- CSF) and 20 ng/ml human recombinant IL-4 for the generation of immature DC (iDC).
  • GM- CSF granulocyte-macrophage colony- stimulating factor
  • IL-4 immature DC
  • iDC a portion of iDC are co-cultured with apoptotic MCF-7 cells at a DC/tumor cell ratio of 1 :2 for 24h in RPMI 1640 supplemented with GM-CSF (20 ng/mL), IL-4 (20 ng/niL) and 5 % FBS.
  • Another portion of iDC are freezed at 2 x 10 6 / per vial - in 90% FBS -10% DMSO.
  • pulsed DCs are matured with tumor necrosis factor a (TNF-a; 20 ng/mL final) and PGE2 (1 ⁇ ) for 2 days (mDC).
  • TNF and PGE2 at the same concentrations
  • lipopolysaccharide LPS; 10-1000 ng/mL; Sigma
  • Antigen- loaded DC stimulators are irradiated at 30 Gy.
  • the presence of IL-17 in the culture supernatant is measured by ELISA.
  • the recognition of IL-17 by an anti-IL-17 antibody may be carried out by conventional methods known in the art such as a sandwich ELISA anti-IL-17.
  • the ELISA is developed by any colorimetric means known in the art such as for example using a detection antibody labelled with biotin, a poly-streptavidin HRP amplification system and an o-phenylenediamine dihydrochloride substrate solution.
  • One example of said method is the following:
  • a plate with the capture antibody, such as for example an anti-IL17 antibody,
  • a blocking buffer such as, for example, casein 2% in
  • the detection antibody such as for example a biotinylated anti-IL-17 antibody
  • IL-17 level inferior to 200 ng/ml, 100 ng/ml, 50 ng/ml corresponds to no secretion or low secretion of IL-17 after calculation with the standard curve.
  • CD3 + T cells anti-CD4(SK3)-PerCP-eFluor 710, anti-TCRap(IP26)-APC (ebioscience), anti-CD25 (B1.49.9)-PeCy55, anti-CD 127(R34.34)-APC-AF700 (Beckman Coulter), anti-CD3(UCHTl)- BB515 anti-invariant TCR Va24-JaQ (6B11)- PE, anti-Foxp3 (259D/C7)-PE-CF594 and anti-CD 152 (BNI3)-BV421, anti-CD 161 (DX12) BV605 and anti-CD56(NCAM 16.2) BU395 (Becton Dickinson), anti-TCR ⁇ - BV421 (IP26) (Bio legend), anti- TCR pan ⁇ + PE (IMMU510) (Beckman Coulter) and anti-CD27- APC e
  • Cells are stained for surface markers (at 4°C in the dark for 30 min) using mixtures of Ab diluted in PBS containing BSA/NaNs (0.5% BSA, 0.01% NaN 3 ) (FACS buffer). Foxp3 and CTLA-4 intracellular staining are performed with FOXP3 staining kit obtained from ebioscience according to the manufacturer's instructions. Appropriate isotype control Abs are used for each staining combination. Samples are acquired on a BD LSR FORTESSA flow cytometer using BD FACSDIVA 8.0.1 software (Becton Dickinson). Results are expressed in percentage (%) or in mean fluorescence intensity (MFI).
  • CFSE staining Tconv are stained with 1 ⁇ carboxy- fluorescein succinimidyl ester (CFSE) (CellTrace cell proliferation kit; Molecular Probes/Invitrogen) in PBS for 8 min at 37 °C at a concentration of 1 x 10 7 cells/mL. The labeling are stopped by washing the cell twice with RPMI 1640 culture medium containing 10% FBS. Cells are then resuspended at the desired concentration and subsequently used for proliferation assays.
  • CFSE carboxy- fluorescein succinimidyl ester
  • 7-AAD (7-amino-actinomycin D) staining Apoptosis of stimulated CFSE- labeled or unlabeled nTregs and Tconv was determined using the 7-AAD assay. Briefly, cultured cells are stained with 20 ⁇ g/mL nuclear dye 7-AAD (Sigma- Aldrich) for 30 min at 4 °C. FSC/7-AAD dot plots distinguish living (FSC high /7-AAD " ) from apoptotic (FSC high /7- AAD + ) cells and apoptotic bodies (FSC low /7- AAD + ) and debris ((FSC low /7-AAD " ). Living cells are identified as CD3 + 7-AAD " FSC + cells.
  • T-cell proliferation is assessed CFSE dilution assay in RPMI supplemented with 5% FBS, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES (RPMI-5 media) in normoxia.
  • stimulated CFSE-labeled Tconv are harvested, costained with anti-CD3 mAb and 7-AAD, and the percentage of living proliferating cells (defined as CFSE low fraction) in gated CD3 + 7-AAD- cells is determined by flow cytometry.
  • Standard polyclonal cell-cell contact Treg suppression assay CFSE-labeled Tconv (4 x 10 4 per well), used as responder cells, are cultured with ACD3- feeder (4 ⁇ 10 4 per well) in the presence or absence of defined amounts of Foxp3 T cells (blood Treg or ex vivo generated T cells) for 4 to 5 d. Cultures are performed in round-bottom plates coated with 0.2 ⁇ g/mL anti-CD3 mAb in 200 of complete RPMI medium.
  • Results are expressed as the percentage of proliferating CFSE low T cells or as a percentage of suppression calculated as follows: (100 x [(percentage of Tconv CFSE low cells - percentage of Tconv CFSE low in coculture with nTregs)/percentage of Tconv CSFE low cells.
  • TSDR Treg specific demethylation region
  • DNA standards originated from unmethylated bisulfite-converted human EpiTect control DNA (Qiagen) or universally methylated bisulfite-converted human control DNA (Zymo Research).
  • the TSDR was PCR-amplified using the following reaction: 50 ⁇ reaction volume containing 25 ⁇ of ZymoTaq PreMix buffer (Zymo Research) and 0.5 ⁇ each of the primers FOXP3_TSDRfwd (5'- ATATTTTTAGATAGGGATATGGAGATGATTTGTTTGG-3' SEQ ID NO: 1) and FOXP3_TSDRrev (5'-AATAAACATCACCTACCACATCCACCAACAC-3' - SEQ ID NO: 2). After incubation at 95°C for 10 min, amplification was performed as follows: 50 cycles at 95°C for 30 s, 55°C for 30 s, and 72°C for 1 min.
  • Amplified PCR products were purified with the QIAquick Gel Extraction Kit (Qiagen). The concentration of purified control TSDR DNA was determined with a GE NanoVue spectrophotometer (GE Healthcare Life Sciences). TSDR real-time PCR was performed with probes that targeted methylated or demethylated target sequences. The reaction was performed in 96-well white trays with a Roche LightCycler 480 system (Roche Diagnostics). Each reaction contained 10 ⁇ LightCycler 480 Probes Master Mix (Roche), 10 ng of bisulfite converted DNA sample or standards, 1 ⁇ of each primer, and 150 nM of each probe with a final reaction value of 20 ⁇ .
  • the probes used for amplification were TSDR- Forward 5 '-GGTTTGTATTTGGGTTTTGTTGTTAT AGT-3 ' (SEQ ID NO: 3) and TSDR-Reverse 5 '-CT AT AAAAT AAAAT ATCTACCCTCTTCTCTTCCT-3 ' (SEQ ID NO: 4).
  • the probes for target sequence detection were FAM-labeled methylated probe, FAM-CGGTCGGATGCGTC-MGB-NFQ (SEQ ID NO: 5), or VIC-labeled unmethylated probe, VIC-TGGTGGTTGGATGTGTTG-MGB-NFQ (SEQ ID NO: 6). All samples were tested in triplicate.
  • the protocol for real-time amplification is as follows: after initial denaturation at 95°C for 10 min, the samples were subjected to 50 cycles at 95°C for 15 s and at 61°C for 1 min. Fourteen different ratios of fully methylated and demethylated template were used as real-time standards. A six-order polynomial equation was used to extrapolate the percentage of cells demethylated at the TSDR for each sample.
  • Histone acetylation analysis of the four different sites of FOXP3 gene was evaluated by ChIP assay, as previously described by Ling Lu (Ling Lu et al, PNAS 2014). Briefly, 50,000 cells of each treated nTreg cell sample were harvested and cross-linked with 1% formaldehyde, and then lysed with 120 ⁇ of lysis buffer [50 mM Tris-HCl, pH 8.0, 10 mM EDTA, 1% (wt/vol) SDS, protease inhibitor mix (1 : 100 dilution; Sigma), 1 mM PMSF, 20 mM Na-butyrate].
  • lysis buffer 50 mM Tris-HCl, pH 8.0, 10 mM EDTA, 1% (wt/vol) SDS, protease inhibitor mix (1 : 100 dilution; Sigma), 1 mM PMSF, 20 mM Na-butyrate.
  • the chromatin in the lysate was sonicated to 500-800-bp fragments and then diluted with 800 ⁇ of RIP A ChIP buffer [10 mM Tris-HCl, pH 7.5, 140 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 1% (vol/vol) Triton X-100, 0.1% (wt/vol) SDS, 0.1% (wt/vol) Na- deoxycholate, protease inhibitor mix (1 : 100 dilution; Sigma), 1 mM PMSF, and 20 mM Na- butyrate].
  • RIP A ChIP buffer 10 mM Tris-HCl, pH 7.5, 140 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 1% (vol/vol) Triton X-100, 0.1% (wt/vol) SDS, 0.1% (wt/vol) Na- deoxycholate, protease inhibitor mix (1 : 100 d
  • Dynabeads protein G ( ⁇ ; Invitrogen) was incubated with ⁇ g ofH3K4me3 (Abeam) or H3K9ac (Cell Signaling) or normal rabbit IgG negative control ChlP-grade antibodies for 2 h separately. Then, ⁇ of the sheared chromatin was immunoprecipitated with pretreated antibody-bead complexes and another ⁇ of the sheared chromatin for total input DNA extraction separately.
  • Immunoprecipitated DNA was quantified by real-time PCR with following primers: promoter, 5'-ACC GTA CAG CGT GGT TTT TC-3' (SEQ ID NO: 7) and 5'-CTA CCT CCC TGC CAT CTC CT-3' (SEQ ID NO: 8); CNS1, 5'- CCC AAG CCC TAT GTG TGATT-3' (SEQ ID NO: 9) and 5'-GTG TGT CAG GCC TTG TGC TA-3' (SEQ ID NO: 10) ; CNS2, 5'-GTC CTC TCC ACAACC CAA GA-3' (SEQ ID NO: 11) and 5'-GAC ACC ACG GAG GAA GAG AA -3' (SEQ ID NO: 12); and CNS3, 5'-AGG TGC CGA CCT TTA CTG TG-3' (SEQ ID NO: 13) and 5'- ACA ATA CGG CCT CCT CCT CT-3' (SEQ ID NO: 14). Results a
  • Luminal A and B subtypes are both estrogen-receptor-positive (ER+) and low-grade, with luminal A tumors growing very slowly and luminal B tumors growing more quickly. Luminal A tumors have the best prognosis. Luminal B tumors are associated with a poor clinical outcome.
  • Foxp3 expressing CD3 + TCRy5 + T cells present a same phenotypic profile as Foxp3 + CD3 + TCRaP + T cells. These Foxp3 + TCRy5 + T cell population express levels of Foxp3, CD25 and CTLA4 similar to those of Foxp3 + CD3 + TCRaP + T cells ( Figure 4). b) Ex vivo generation and expansion of specific CD3 + TCRy5 + expressing Foxp3 committed to exclusively exert regulatory activity.
  • Figure 5 shows that naive CD3 + TCRy5 + T cells (CD3 + CD45RA + CD27 + TCRy5 + T cells) stimulated with zoledronic acid-treated-autologous tDCs, in presence of the nTreg polarizing medium comprising the combination of IL-15, IL-2, TGF , PGE2 and rapamycin, express Foxp3 after 21 days expansion and exhibit significant functional suppressive activity, as assessed by the standard polyclonal cell-cell contact Treg suppression assay.
  • the 21 -day-expanded FOXP3 expressing CD3 + TCRy5 + T cells maintain their Foxp3 level and their suppressive activity, after a further 21 -day- culture in nTreg polarizing medium.
  • nTreg polarizing medium comprising the combination of IL-15, IL-2, TGF , PGE2 and rapamycin
  • nTreg polarizing medium were assessed for their capacity to induce the expression the differentiation of Foxp3 + cells with suppressive function.
  • Figure 6 shows that cultured naive CD3 + invTCR Va24 + T cells exhibit a variable level of Foxp3 dependent on their culture condition of stimulation.
  • Polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin results in a higher Foxp3 expression over combinations of IL-2, TGF and rapamycin, IL-2 and PGE2, or IL-2 alone.
  • the combination of IL-2, TGF , PGE2 and rapamycin results in an optimal intensity of Foxp3 expression in the invTCR Va24 + T cells, as compared to the other combinations.
  • CD3 + invTCR Va24 + T cells stimulated with the polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin express level and intensity of Foxp3 similar or higher to those of blood naive regulatory T cells (CD3 + TCRa CD4 + CD127 /low CD45RA + CD25 + ), corresponding to our positive control.
  • FIG. 7A shows that tumor Ag-specific memory invTCR Va24 + T cells ex vivo generated and expanded in the presence of the nTreg polarizing medium above described are endowed of a higher suppressive activity than fresh Foxp3 expressing CD3 + CD4 + TCRaP + MHCII restricted T cells when using an autologous MLR coculture assay.
  • Figure 7B shows that these tumor Ag-specific invTCR Va24 + T cells still maintain their suppressive activity, when the autologous MLR coculture assay are performed in presence of a high inflammatory medium containing IL-2 IL-1 IL-6, IL-21 IL-23 cytokines, while fresh Foxp3 expressing CD3 + CD4 + TCRaP + MHCII restricted T cells lose their suppressive activity.
  • nTreg polarizing medium were assessed for their capacity to induce the differentiation of Foxp3 + cells with suppressive function.
  • Figure 8 shows that, when ex vivo activated polyclonally with anti-CD3 mAbs, naive conventional CD4 + T cells exhibit a variable level of Foxp3 dependent on their culture condition of stimulation.
  • Polarizing medium comprising the combination of IL-2, TGF and rapamycin or IL-2, TGF , rapamycin and PGE2 results in a higher Foxp3 expression over combinations of IL-2 and PGE2, or IL-2 alone (B).
  • the combination of IL-2, TGF , rapamycin and PGE2 results in an optimal intensity of Foxp3 expression in the CD3 + CD4 + TCRa,p + MHCII restricted T cells, as compared to the other combinations (C).
  • CD4 + T cells stimulated with the polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin, express level and intensity of Foxp3 similar or higher to those of blood naive regulatory T cells (CD3 + TCRa + CD4 + CD127 /low CD45RA + CD25 + ), corresponding to our positive control.
  • FIG. 9A shows that CD3 + CD4 + TCRaP + MHCII restricted T cells, ex vivo generated and expanded for 21 days, using polyclonal stimulation, in the presence of the nTreg polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin, display a higher suppressive activity compared with both those generated in the presence of the nTreg polarizing medium comprising the combination of IL-2, TGF , rapamycin without PGE2 and fresh FOXP3 expressing CD3 + CD4 + TCRaP + MHCII restricted T cells, when using the standard polyclonal cell-cell contact Treg suppression assay.
  • Figure 9B shows that these 21-day-expanded-FOXP3 expressing CD3 + CD4 + TCRa,p + MHCII restricted T cells still maintain their suppressive activity, when the functional suppressive assay is performed in presence of a highly- inflammatory medium containing IL-2 IL-1 IL-6, IL-21 IL-23 cytokines, while fresh FOXP3 expressing CD3 + CD4 + TCRa,p + MHCII restricted T cells lose their suppressive capacity under these culture condition of stimulation.
  • the ova-specific- pTreg are further cultured for 3 weeks either in nTreg or TH-17 polarizing medium (IMDM medium containing IL-2 IL-1 IL-6, IL-21 IL-23 cytokines) and were tested for 1) their functional suppressive capacity in the presence of a high inflammatory context ( Figure 10) and 2) for their IL-17-producing capacity when stimulated through CD3 and CD28 as described above ( Figure 11).
  • Ova-specific CD3 + TCRa,p + MHCII restricted T cells After a further 21 -day-culture either in nTreg or TH-17 polarizing medium, Ova-specific CD3 + TCRa,p + MHCII restricted T cells not only still retain, in a high inflammatory context, functional suppressive activity (Figure 10), but also produce low level of IL-17 ( Figure 11B).
  • fresh Foxp3 expressing CD3 + TCRa,p + MHCII restricted T cells lose their suppressive function while producing IL-17 in this inflammatory context.

Abstract

The present invention relates to therapeutic uses of ex vivo generated Foxp3+ regulatory T cells. The inventors showed the presence of Foxp3+ expressing T cells in tumor infiltrating lymphocytes (TILs) isolated from luminal-B breast cancer. The inventors performed an ex vivo generation and expansion of specific CD3+ TCRy8+ expressing Foxp3: CD3+ TCRy8+ T cells maintain their Foxp3 level and their suppressive activity, after a further 21-day-culture. They also showed that tumor Ag-specific CD3+ TCR Va24+ T cells maintain their ability to perform suppressive function in pro-inflammatory conditions. In particular, the present invention relates to immunotherapeutic uses of at least one of ex vivo generated Foxp3+regulatory T cells population selected among a MHCII restricted CD4+Foxp3+regulatory T cells population, a y8 Foxp3+regulatory T cells population and an invariant Foxp3+regulatory T cells population.

Description

IMMUNOTHERAPEUTIC USES OF EX VIVO GENERATED FOXP3+
REGULATORY T CELLS
FIELD OF INVENTION The present invention relates to therapeutic uses of ex vivo generated Foxp3+ regulatory T cells.
BACKGROUND OF INVENTION
The focal point for cancer treatment is using a combination of conventional therapies: chemotherapy, radiotherapy and surgery. In most cases, they are effective in treating primary tumors; however, they inefficiently prevent metastasis through disseminated tumor cells. In recent years, immunotherapy has emerged as an alternative therapeutic strategy for the treatment of cancer, largely due to the clinical development of novel agents including cytokines, monoclonal antibodies (mAbs), genetically engineered chimeric antigen receptor and immune checkpoint blockade inhibitors. Immune checkpoint therapy, based on the passive administration of mAbs blocking negative regulators of the activation of effector T cells such as cytotoxic T lymphocyte- associated antigen 4 (CTLA-4) (Ipilimumab) and programmed death- 1 (PD1) (Pembrolizumab and Nivolumab), have indeed achieved durable responses in some patients, including long-term remissions with no clinical signs of cancer. However, interruption of immune checkpoints with mAbs is commonly associated with immune mediated toxicities such as auto-immune sequelae and inflammatory damage to normal parenchyma. The most common damages related to Ipilimumab, include rash/pruritus, enterocolitis, uveitis, pancreatitis, hypophysisitis and leucopenia. These adverse events could be anticipated because of the systemic activity of the administered mAbs, targeting thus all tissues instead of acting locally.
These observations have emphasized the need of alternative therapeutic strategies aiming to target specifically the cancer cells and/or their microenvironment. Such requirements could be achieved by therapeutic anti-cancer vaccinations. Anti-cancer vaccines are designed to elicit an immune response against tumor antigens or cells allowing a protection against tumor recurrence or metastatic disease. There are two general types of antigen associated with tumor cells: tumour-associated (TAA) or tumour-specific (TSA). While TAA expression is a function of inappropriate production of embryonic gene-encoded oncofetal antigens or overexpression of normal protein, TSA are either new antigens induced mostly by chemical carcinogens and sometimes by virus or viral antigens. Many tumor antigens have been identified and over hundred therapeutic cancer vaccines trials have been initiated since 20 years, using as vaccine immunogen clearly defined derived peptide or full-length recombinant tumor antigens. Though safety and immunogenicity have been thoroughly documented, none of these vaccines have yet been reported to be effective.
To improve these therapeutic anti-cancer vaccines, whole tumor cell-based vaccines have been developed. The advantage of using whole tumor cells as immunogen is that the cells provide a source of several TAAs or TSAs, some identified and some as yet undefined. These anti-cancer vaccines used as active principle 1) whole tumor cells or genetically modified tumor cell lines to express cytokines, chemokines or costimulatory molecules to enhance their immunogenicity, 2) cellular lysates or 3) immunogenic apoptotic bodies derived from tumor cells. However, immunization with cancer cell- based vaccines failed to induce long-lasting anti-tumor responses and has thus not resulted in significant long-term therapeutic benefits. Today the only therapeutic anticancer vaccine that has been licensed for use in clinical practice is sipuleucel-T (Provenge), a dendritic cell vaccine used in men with metastatic hormone-refractory prostate cancer. The absence of vaccine efficacy could be accounted for by a deficient cellular immune response to cancer antigens observed in the cancer patients. Immune escape i.e. paralysis of immune cells surrounding cancer cells was further substantiated by the demonstration that intratumoral lymphocytes exhibited immune suppressive activities. The clinical success of therapeutic antibodies targeting immune checkpoint receptors on T cells, as described above and their ligands, has stressed the role of peripheral tolerance mechanism in limiting vaccine responses. The targeted negative regulator CTLA-4 and PD1 molecules, are indeed also expressed by regulatory T cells. Therefore, it has been suggested that in addition to blocking negative signaling in effector T cells, allowing then greater expansion of all T-cell populations, presumably including those with antitumor reactivity, anti-CTLA-4 or anti-PD-1 mAbs also eliminate regulatory T cells (Tregs) present in tumor microenvironment. The immune suppressive microenvironment of a tumor seems to be one of the major impediments to the development of effective immune therapy for cancer.
As Treg-induced immune suppression is one of the major impediments to the development of effective immune therapy for cancer, approaches to eliminating immunosuppressive regulatory T cells before vaccination are also being tested.
Indeed, a considerable body of recent literature deals with various methods for Treg depletion. Treg express surface molecules that can be specifically targeted by antibodies (Abs) or pharmacologic inhibitors. For example, CD25, TGF β pathway, CTLA-4, PD-1, CD73, CD390... are targeted for Treg silencing either by Treg removal or impairment of Treg suppressor functions. To date, a variety of agents, including Abs (daclizumab: anti-CD25 Ab), IL-2 fusion toxins such as denileukin diftitox (Ontak), or drugs such as cyclophosphamide or tyrosine kinase inhibitors (sunitinib), have been tested in preclinical in vitro studies with human cells. These clinical trials have shown no convincing evidence of a relation between Treg depletion and clinical benefits in patients with cancer.
Considering the current background, which includes that 1) conventional anti-cancer vaccine preparations based on immunogenic apoptotic cancer cells, as active principle, may be effective, though as yet transitory, 2) targeting immune regulatory molecules such as CTLA-4 or PD1 with mAbs appear to be therapeutically effective, though with antibody therapy side effects, and 3) the cancer's immune system remain quite normal, up to late advanced stages of disease, the present invention concerns the development of a vaccine directed against the immune regulatory T cells specifically activated by cancer cells. The stroma of malignant tumors is indeed comprised of TILs (Tumor-infiltrating lymphocytes) known 1) to be highly enriched on regulatory T cells and 2) to exert an immune suppressive activity, which likely accounts for the local cancer immune escape.
The inventors thus suggest using a vaccine composition comprising inactivated Foxp3+ regulatory T cells as active principle. The inactivated Foxp3+ regulatory T cells may represent an antigenic target to induce an immune response directed against the Foxp3+ regulatory T cells present in the TILs, thereby preventing their immune suppressive activity and allowing the cytotoxic activity of effector cells such as NK cells against the tumor cells. In addition, as the TILs may comprise one or more different Foxp3+ regulatory T cells populations, the inventors suggest determining the one or more Foxp3+ regulatory T cells population overrepresented in the TILs, to ex vivo generate and expand the corresponding Foxp3+ regulatory T cells populations and thus prepare a treatment adapted to the tumor to be treated.
SUMMARY The present invention relates to an immunogenic product comprising at least one inactivated ex vivo generated Foxp3+ regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population.
Another object of the present invention is a pharmaceutical composition comprising at least one inactivated ex vivo generated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient.
A further object of the present invention relates to a vaccine composition comprising at least one inactivated ex vivo generated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3 +regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one adjuvant. The present invention also relates to a pharmaceutical composition comprising at least one ex vivo generated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated regulatory T cells population remains stable when placed in inflammatory condition.
In one embodiment, the immunogenic product, pharmaceutical composition or vaccine composition according to the invention is for use in treating cancer.
The present invention further relates to a method for preparing the immunogenic product, pharmaceutical composition or vaccine composition according to the invention, comprising: identifying from a tumor sample obtained from the subject the at least one regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population, that is overrepresented, ex vivo generating the at least one overrepresented regulatory T cells population, inactivating the at least one ex vivo generated regulatory T cell population.
Another object of the present invention is a pharmaceutical composition according to the invention for use in cell therapy. A further object of the present invention is a pharmaceutical composition according to the invention for use in treating inflammatory or autoimmune diseases or for preventing transplant rejection or graft versus host disease (GVHD).
In one embodiment, the at least one ex vivo generated regulatory T cells population of the immunogenic product, pharmaceutical composition or vaccine composition according to the invention is obtained by a method comprising: for the MHCII restricted CD4+ Foxp3+ regulatory T cells population: culturing CD3+ CD4+ CD25" T cells in the presence of a TCRa-β cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the γδ Foxp3+ regulatory T cells population: culturing CD3+ TCR γδ + T cells in the presence of a γδ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the invariant Foxp3+ regulatory T cells population: culturing CD3+ Va24+ T cells in the presence of an invariant T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days.
In one embodiment, the TCRa-β cell activator is a polyclonal TCRa-β cell activator, preferably an anti-CD3 antibody or an anti-TCRaP antibody; the γδ T cell activator is a polyclonal γδ T cell activator, preferably an anti-TCR γδ antibody or a non peptide phosphoantigen; and the invariant T cell activator is a polyclonal invariant T cell activator, preferably a Va24 activator.
In another embodiment, the TCRa-β cell activator is an antigen-specific TCRa-β cell activator, preferably tolerogenic dendritic cells (DCs) and pulsed with at least one self- peptide antigen; the γδ T cell activator is an antigen-specific γδ T cell activator, preferably tolerogenic dendritic cells (DCs) and pulsed with at least one bisphosphonate, preferably at least one aminobiphosphonate and the invariant T cell activator is an antigen-specific invariant T cell activator, preferably tolerogenic dendritic cells (DCs) expressing CDl and pulsed with at least one non peptide lipid antigen. In one embodiment of the present invention, the cAMP activator is selected from the group comprising prostaglandin E2 (PGE2), an EP2 or EP4 agonist, a membrane adenine cyclase activator or a metabotropic glutamate receptors agonist.
In one embodiment, the TGF pathway activator is selected from the group comprising TGF , bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin and nodal .
In one embodiment of the present invention, the mTOR inhibitor is selected from the group comprising rapamycin, rapamycin analogs, wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG), curcumin, resveratrol; genistein, 3, 3- diindolylmethane (DIM), LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l-benzopyran-4- one), PP242, PP30, Torinl, Ku-0063794, WAY-600, WYE-687, WYE-354, GNE477, NVP-BEZ235, PI- 103, XL765 and WJD008.
In one embodiment, the at least one ex vivo generated Foxp3+ regulatory T cells population are expanded by a method comprising: - for the MHCII restricted CD4+ Foxp3+ regulatory T cells population: culturing
CD3+ CD4+ CD25" T cells in the presence of a TCRaP cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL- 15 and TSLP, for at least 5 days; for the γδ Foxp3+ regulatory T cells population: culturing CD3+ TCRy5 + T cells in the presence of a γδ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGFP (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least
5 days; for the invariant Foxp3+ regulatory T cells population: culturing CD3+ Va24+ T cells in the presence of an invariant T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days.
DEFINITIONS
As used herein, "regulatory T cells" or "Treg" refers to cells capable of suppressive activity (i.e. inhibiting proliferation of conventional T cells), either by cell-cell contact or by MLR suppression (Mixed Lymphocytes Reaction). These cells include different subpopulations including but not limited to, peripheral regulatory T cells, γδ regulatory T cells and invariant regulatory T cells.
As used herein, "invariant Foxp3+ regulatory T cells" refers to cells having the following phenotype: CD3+ Va24+ Foxp3+. The term "invariant" as used herein includes the term "semi-invariant", where the semi-invariant T cells are T cells not expressing νβΐ 1. As used herein the isolated population of the invention is a population of semi-invariant Foxp3+ T cells having the following phenotype: CD3+ Va24+ Foxp3+Vpi Γ. These cells recognize non peptide lipid antigens under CD1 restriction.
As used herein, "y5Foxp3+ regulatory T cells" refers to cells having the following phenotype: y5TCR+ Foxp3+. These cells recognize non peptide phospho antigens with no MHC (major histocompatibility complex) restriction.
As used herein, "MHCII restricted CD4+ Foxp3+ regulatory T cells" refers to cells having the following phenotype: CD4+CD25+Foxp3+. These cells are thymic derived or peripherally induced. These cells can be identified by their a TCR (T cell receptor) and recognize peptides (including foreign or self peptides) presented by restricted MHC class II (major histocompatibility complex class II) molecules.
As used herein, the term "treatment" refers to therapeutic treatment and prophylactic and preventive measures, wherein the object is to prevent or slow down (lessen, diminish) the targeted pathological disorder or condition. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. A subject or mammal is successfully "treated" for a disease if, after receiving a therapeutic amount of Foxp3+ regulatory T cells or a therapeutically amount of inactivated Foxp3+ regulatory T cells according to the present invention, the patient shows observable and/or measurable reduction in or absence of one or more of the following: reduction in the number of pathogenic cells; reduction in the percent of total cells that are pathogenic; and/or relief to some extent, of one or more of the symptoms associated with the specific disease or condition; reduced morbidity and mortality, and improvement in quality of life issues. The above parameters for assessing successful treatment and improvement in the disease are readily measurable by routine procedures familiar to a physician.
As used herein, "therapeutically effective amount" refers to the number of Foxp3+ regulatory T cells or of inactivated Foxp3+ regulatory T cells that is aimed at inducing a therapeutic response, without causing significant negative or adverse side effects to the target. A therapeutically effective amount may be administered prior to the onset of the disease to be treated, for a prophylactic or preventive action. Alternatively or additionally, the therapeutically effective amount may be administered after initiation of the disease to be treated, for a therapeutic action.
As used herein, "therapeutic response" refers to a therapeutic benefit induced by the Foxp3+ regulatory T cell therapy or the Foxp3+ regulatory T cell vaccination in a subject. A therapeutic response may include the fact of (1) delaying or preventing the onset of the disease to be treated; (2) slowing down or stopping the progression, aggravation, or deterioration of one or more symptoms of the disease to be treated; (3) bringing about ameliorations of the symptoms of the disease to be treated; (4) reducing the severity or incidence of the disease to be treated; or (5) curing the disease to be treated.
As used herein, "about" preceding a figure means more or less 10% of the value of said figure. As used herein, "subject or patient" refers to a mammal, preferably a human. In the present invention, the terms subject and patient may be used with the same meaning. Examples of non-human mammal include a pet such as a dog, a cat, a domesticated pig, a rabbit, a ferret, a hamster, a mouse, a rat and the like; a primate such as a chimp, a monkey, and the like; an economically important animal such as cattle, a pig, a rabbit, a horse, a sheep, a goat. In one embodiment, the subject is awaiting the receipt of, or is receiving medical care or was/is/will be the object of a medical procedure, or is monitored for the development of a disease. In one embodiment, the subject is an adult (for example a subject above the age of 18). In another embodiment, the subject is a child (for example a subject below the age of 18). In one embodiment, the subject is a male. In another embodiment, the subject is a female.
As used herein, "allogeneic cells" refers to cells isolated from one subject (the donor) and infused in another (the recipient or host).
As used herein, "autologous cells" refers to cells that are isolated and infused back into the same subject (recipient or host).
DETAILED DESCRIPTION Methods
Invariant Foxp3+ regulatory T cells The present invention relates to a method for generating ex vivo invariant Foxp3+ regulatory T cells as defined here above.
In one embodiment, the method for generating ex vivo invariant Foxp3+ regulatory T cells, comprises:
- culturing CD3+Va24+ T cells, preferably CD3+Va24+CD45RA+ T cells, in the presence of an invariant T cell activator and the following agents: i) an cAMP
(Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected from the group of IL-2, IL-7, IL-15 and TSLP (Thymic stromal lymphopoietin), for at least 5 days, thereby obtaining a population of invariant Foxp3+ regulatory T cells ex vivo generated, preferably from invariant naive (CD45RA+) T cells. In one embodiment, the CD3+Va24+ T cells, preferably CD3+Va24+CD45RA+ T cells, are obtained by any technic well known in the art from a blood sample. In one embodiment, the CD3+Va24+ T cells, preferably CD3+Va24+CD45RA+ T cells, are isolated from PBMCs (peripheral blood mononuclear cells) by flow cytometry. In one embodiment, the CD3+Va24+ T cells, preferably CD3+Va24+CD45RA+ T cells, may be isolated from frozen PBMCs.
In one embodiment, the obtainment of isolated CD3+ Va24+ T cells, preferably CD3+ Va24+ CD45RA+ T cells, may be improved by an optional first to a purification step. The CD3+ Va24+ T cells, preferably CD3+ Va24+ CD45RA+ T cells, are stimulated with antigen pulsed tolerogenic DCs (for example ovalbumin pulsed tolerogenic DCs) in the presence of soluble anti-CD28 and anti-CD40 antibodies. In one embodiment, the time of stimulation ranges between 1 hour and 24 hours, preferably between 10 hours and 20 hours, more preferably during about 16 hours. After stimulation, cells are washed, for example with PBS, and stained with anti-CD 154 and anti-CD4 antibodies for sorting. The purified CD3+ Va24+ CD154+ T cells are enriched and may be used for the following activation step.
In one embodiment, the CD3+Va24+ T cells are activated in the presence of an invariant T cell activator. Said invariant T cell activator can be a polyclonal invariant T cell activator or an antigen-specific invariant T cell activator.
In the present invention, the polyclonal invariant T cell activator is a Va24 activator. Examples of Va24 activator include, but are not limited to, anti-Va24 antibody such as 6B11 antibody (Montoya CJ et al. Immunology. 2007 Sep; 122(1): 1-14), or CD1 ligands including CD la ligands, CD lb ligands, CDlc ligands and CD Id ligands, preferably CD Id ligands such as a-galactosylceramide (a-GalCer) and analogs such as for example HS44 (a synthetic amino cyclitolic ceramide analogue in which the sugar head group is a carba cyclitol ring that mimics glucose instead of galactose, and which has the O- glycosidic linkage replaced with an amide group), cc-GalCer analogs of the table 1 herein below:
Table 1 : Structure of a-GalCer analogs
Figure imgf000014_0001
and homodimeric a-galactosylceramide analogs including the following:
Figure imgf000015_0001
wherein R is C02Me, C02H or CH2OH; a-glucuronyl- and a-galacturonyl-ceramides and analogs thereof; iGb3 (Isoglobotriosylceramide); N-glycolyl (NGc) gangliosides such as for example NGcGM3; glycosphingo lipids or phosphoglycero lipids such as phosphatidylinositol, phosphatidylethanolamine, and phosphatidylglyerol presented preferably by CD Id.
In one embodiment, the polyclonal invariant T cell activator is an anti-Va24 antibody, preferably a m6Bl 1 antibody.
In one embodiment, the polyclonal invariant T cell activator is soluble in the culture medium. In another embodiment, the polyclonal invariant T cell activator is coated to the culture plate. In one embodiment, the polyclonal invariant T cell activator is used in the presence of feeder cells, preferably autologous feeder cells.
Feeder cells include, but are not limited to, ACD3 cells (T cell-depleted accessory cells), irradiated PBMCs, irradiated DCs, artificial APCs (antigen presenting cells), Sf9 cells, insect cells, a pool of PBMCs or a pool of B cells from different subjects, KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
Preferably, the feeder cells used in the invention are ACD3 cells that are isolated by negative selection from PBMCs by incubation with anti-CD3 coated beads and then irradiated at 3000 rad. In one embodiment, the ratio T cells / feeder cells ranges from 1:100 to 1:10000, preferably from 1:1000 to 1:5000. Within the scope of the invention, the expression "from 1:100 to 1:10000" includes, without limitation 1:100, 1:200, 1:300, 1:400, 1:500, 1:600, 1:700, 1:800, 1:900, 1:1000, 1:1250, 1:1500, 1: 1750, 1:2000, 1:2250, 1:2500, 1:2750, 1:3000, 1:3250, 1:3500, 1: 3750, 1:4000, 1:4250, 1:4500, 1:4750, 1:5000, 1:5250, 1:5500, 1: 5750, 1:6000, 1:6250, 1:6500, 1: 6750, 1:7000, 1:7250, 1:7500, 1: 7750, 1:8000, 1:8250, 1:8500, 1: 8750, 1:9000, 1:9250, 1:9500, 1:9750 and 1:10000.
In the present invention, the antigen-specific invariant T cell activator is tolerogenic dendritic cells (DCs) expressing CD1, i.e. CD la, CD lb, CDlc and/or CD Id, and pulsed with at least one non peptide lipid antigen. Preferably the tolerogenic DCs express CD Id.
In one embodiment, tolerogenic DCs express on their surface the major histocompatibility (MHC) class la and/or MHC class lb. The MHC class la presentation refers to the "classical" presentation through HLA-A, HLA-B and/or HLA-C molecules whereas the MHC class lb presentation refers to the "non-classical" antigen presentation through HLA-E, HLA-F, HLA-G and/or HLA-H molecules.
In one embodiment, tolerogenic DCs express 50% of MHC class la molecules and 50% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 45% of MHC class la molecules and 55% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 40%> of MHC class la molecules and 60% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 35% of MHC class la molecules and 65% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 30% of MHC class la molecules and 70% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 25% of MHC class la molecules and 75% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 20% of MHC class la molecules and 80% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 15% of MHC class la molecules and 85% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 10% of MHC class la molecules and 90% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 5% of MHC class la molecules and 95% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express only MHC class lb molecules on their surface.
In one embodiment, tolerogenic DCs express 50% of HLA-A, HLA-B and/or HLA-C molecules and 50% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 45% of HLA-A, HLA-B and/or HLA-C molecules and 55% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 40% of HLA-A, HLA-B and/or HLA-C molecules and 60% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 35% of HLA-A, HLA-B and/or HLA-C molecules and 65% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 30% of HLA-A, HLA-B and/or HLA-C molecules and 70% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 25% of HLA-A, HLA-B and/or HLA-C molecules and 75% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 20% of HLA-A, HLA-B and/or HLA-C molecules and 80% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 15% of HLA-A, HLA-B and/or HLA-C molecules and 85% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 10% of HLA-A, HLA-B and/or HLA-C molecules and 90% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 5% of HLA-A, HLA-B and/or HLA-C molecules and 95% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express only HLA-E molecules on their surface.
In one embodiment, the non peptide lipid antigen is a recombinant antigen a- galactosylceramide and analogs such as for example HS44 (a synthetic amino cyclitolic ceramide analogue in which the sugar head group is a carba cyclitol ring that mimics glucose instead of galactose, and which has the O-glycosidic linkage replaced with an amide group), a-GalCer analogs of the table 1 herein above and homodimeric a- galactosylceramide analogs including the following:
Figure imgf000018_0001
wherein R is C02Me, C02H or CH2OH; a-glucuronyl- and a-galacturonyl-ceramides and analogs; iGb3 (Isoglobotriosylceramide); N-glycolyl (NGc) gangliosides such as for example NGcGM3; glycosphingo lipids or phosphoglycerolipids such as phosphatidylinositol, phosphatidylethanolamine, and phosphatidylglyerol presented preferably by CD Id.
In another embodiment, the non peptide lipid antigen is derived from immunogenic apoptotic bodies from cancer cells or derived from tissue lysate. Cancer cells may derive from tumor biopsy or from expansion of circulatory cancer cells.
Immunogenic apoptotic bodies from cancer cells may be obtained for example with anthracyclines including, without limitation, doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin, UVC or γ-radiation treated cancer cells releasing apoptotic bodies or can be directly isolated from anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin; UVC or γ-radiation treated cancer.
Examples of tissue lysate include, but are not limited to, synovial liquid or inflammatory tissue lysate. As used herein, "tolerogenic DCs" refers to DCs capable to induce tolerance. In one embodiment, tolerogenic DCs are capable of secreting more suppressive cytokines such as IL-10 and TGF than proinflammatory cytokines such as IL-12, IL-23 or TNFa. In one embodiment, DCs are defined as tolerogenic when they secrete IL-10 and IL-12 in a ratio IL-10 : IL-12 > 1. Methods for obtaining tolerogenic DCs are well-known in the art. An exemplary method is the generation of tolerogenic DCs from CD14+ monocytes. For example, CD14+ monocytes are cultured in the presence of GM-CSF and IL-4, or in the presence of GM-CSF and IFNa, for the generation of immature DCs.
Methods for inhibiting MHC class la molecules expression or inducing the expression of HLA-E molecules on the surface of tolerogenic DCs are well-known. The inhibition of the TAP transporter (transporter associated with antigen processing) leads to a decreased expression of MHC class la molecules thereby promoting HLA-E molecules expression on the surface of tolerogenic DCs.
Exemplary methods to inhibit the TAP transporter in the endoplasmic reticulum include, but are not limited to, CRISPR-CAS-9 technology, silencing RNA, transfected DCs with the UL-10 viral protein from the CMV (cytomegalovirus) or the use of viral proteins.
Examples of viral proteins able to inhibit the TAP transporter include, but are not limited to, HSV-1 ICP47 protein, varicella- virus UL49.5 protein, cytomegalovirus US6 protein or gammaherpesvirus EBV BNLF2a protein.
Another method is the use of a chemical product to inhibit the expression of MHC class la molecules without changing HLA-E expression on the surface of tolerogenic DCs. Examples of chemical products include, but are not limited to, 5'- methyl-5 '- thioadenosine or leptomycin B. Methods for inducing the expression of CDl, i.e. CD la, CD lb, CDlc and/or CD Id on the surface of the tolerogenic DCs are well-known in the art. For example, sulfatide can be used for the expression of CD la; rifampin and a number of its derivative (e.g., rifabutin) for the expression of CD lb; cholesteryl esters and acylated steryl glycosides for the expression of CDlc and rosiglitazone; retinoic acid; RARa agonist such as AM580, CD437, AM80, BMS961, NRX195183, All-trans-retinoic acid, 9-cis-Retinoic acid, Ch55, TTNPB (4-[(E)-2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)- l-propenyl]benzoic acid), tamibarotene; PPARy agonist such as ciglitazone, darglitazone, edaglitazone, genistein, indomethacin, GW 1929, LG 100754, LT175, CAY10506, nTZDpa, pioglitazone, 15-deoxy-A-12,14-Prostaglandin J2, S26948, telmisartan, tesaglitazar, carnosic acid, troglitazone, bezafibrate, 13(S)- Hydroxyoctadeca-9Z,l lE-dienoic acid, oxidized low-density lipoprotein (oxLDL) for the expression of CD Id. The tolerogenic DCs expressing CDl thus obtained are then pulsed with the non peptide ligand antigen as described here above. In one embodiment, rosiglitazone can be used at a concentration ranging from 0.1 μΜ to 10 μΜ.
In one embodiment, AM580 can be used at a concentration ranging from 1 nM to 10 μΜ. In one embodiment, the cAMP activator added in the culture allows the activation of the cAMP pathway. Examples of cAMP activator include, but are not limited to PGE2 (prostaglandin E2), an EP2 or EP4 agonist, a membrane adenine cyclase activator such as forskolin, or metabotropic glutamate receptors agonists. Examples of PGE2 include, but are not limited to, PGE2 of ref P5640 or P0409 (Sigma- Aldrich), PGE2 of ref 2296 (R&D Systems), PGE2 of ref 2268 (Bio Vision), PGE2 of ref 72192 (Stemcell), PGE2 of ref abl44539 (Abeam), and PGE2 of ref 14010 (Cayman Chemical).
In one embodiment, the cAMP activator, preferably PGE2 is used at a concentration ranging from 0.01 μΜ to 10 μΜ. Within the scope of the invention, the expression "from 0.01 μΜ to 10 μΜ" includes, without limitation 0.02 μΜ, 0.03 μΜ, 0.04 μΜ, 0.05 μΜ, 0.06 μΜ, 0.07 μΜ, 0.08 μΜ, 0.09 μΜ, 0.1 μΜ, 0.2 μΜ, 0.3 μΜ, 0.4 μΜ, 0.5 μΜ, 0.6 μΜ, 0.7 μΜ, 0.8 μΜ, 0.9 μΜ, 1 μΜ, 1.5 μΜ, 2 μΜ, 2.5 μΜ, 3 μΜ, 3.5 μΜ, 4 μΜ, 4.5 μΜ, 5 μΜ, 6 μΜ, 7 μΜ, 8 μΜ, 9 μΜ. In certain embodiments, PGE2 is at a concentration ranging from 0.03 μΜ to 1.5 μΜ.
In one embodiment, the TGF pathway activator added in the culture allows the activation of the TGF pathway. Examples of TGF pathway activators include, but are not limited to, TGFP family (TGFpi, TGFP2, TGFP3), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, and nodal. Examples of TGFP include, but are not limited to, TGFpi of ref T7039 (Sigma- Aldrich), TGFp2 of ref T2815 (Sigma- Aldrich), TGFp3 of ref T5425 (Sigma- Aldrich), human TGFpl of ref P01137 (R&D system), human TGFpl of ref 580702 (Biolegend), TGFpl of ref HZ-1011 (HumanZyme), human TGFpl of ref 14- 8348-62 (Affymetrix eBioscience). In one embodiment, the pathway activator is used at a concentration ranging from 1 ng/ml to 20 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 20 ng/ml" includes, without limitation 2 ng/ml, 2.5 ng/ml, 3 ng/ml, 3.5 ng/ml, 4 ng/ml, 4.5 ng/ml, 5 ng/ml, 5.5 ng/ml, 6 ng/ml, 6.5 ng/ml, 7 ng/ml, 7.5 ng/ml, 8 ng/ml, 8.5 ng/ml, 9 ng/ml, 9.5 ng/ml, 10 ng/ml, 11 ng/ml, 12 ng/ml, 13 ng/ml, 14 ng/ml, 15 ng/ml, 16 ng/ml, 17 ng/ml, 18 ng/ml, 19 ng/ml. In certain embodiments, TGFP is at a concentration ranging from 2.5 ng/ml to 7.5 ng/ml.
In one embodiment, the mTOR inhibitor added in the culture allows the inhibition of the mTOR pathway. Examples of mTOR inhibitor include, but are not limited to, rapamycin (also named sirolimus) and its analogs (termed rapalogs); wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG); curcumin; resveratrol; genistein; 3, 3-diindolylmethane (DIM); LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l- benzopyran-4-one); PP242; PP30; Torinl ; Ku-0063794; WAY-600; WYE-687; WYE- 354; and mTOR and PI3K dual-specificity inhibitors such as GNE477, NVP-BEZ235, PI-103, XL765 and WJD008. Examples of rapamycin include, but are not limited to, rapamycin of ref R0395 (Sigma- Aldrich), rapamycin of ref SI 039 (Selleckchem), rapamycin of ref 1292 (Tocris), rapamycin of ref R-5000 (LC Laboratories), rapamycin of ref tlrl-rap (InvivoGen), rapamycin of ref abl20224 (Abeam), rapamycin of ref R0395 (Sigma-Aldrich). Examples of compounds of the same chemical class than rapamycin used clinically include, but are not limited to, Everolimus (code name RADOOl), Temsirolimus (code name CCI-779, NSC 683864), Zotarolimus (code name ABT-578).
In one embodiment, the mTOR inhibitor, preferably rapamycin, is used at a concentration ranging from 0.1 nM to 50 nM. Within the scope of the invention, the expression "from 0.1 nM to 50 nM" includes, without limitation 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 26 nM, 27 nM, 28 nM, 29 nM, 30 nM, 31 nM, 32 nM, 33 nM, 34 nM, 35 nM, 36 nM, 37 nM, 38 nM, 39 nM, 40 nM, 41 nM, 42 nM, 43 nM, 44 nM, 45 nM, 46 nM, 47 nM, 48 nM, 49 nM. In one embodiment, at least one cytokine selected from IL-2, IL-7, IL-15 and TSLP can be added in the culture.
In one embodiment, IL-2 is used at a concentration ranging from 10 IU/ml to 1000 IU/ml. Within the scope of the invention, the expression "from 10 IU/ml to 1000 IU/ml" includes, without limitation 15 IU/ml, 20 IU/ml, 25 IU/ml, 30 IU/ml, 35 IU/ml, 40 IU/ml, 45 IU/ml, 50 IU/ml, 55 IU/ml, 60 IU/ml, 65 IU/ml, 70 IU/ml, 75 IU/ml, 80 IU/ml, 85 IU/ml, 90 IU/ml, 95 IU/ml, 100 IU/ml, 150 IU/ml, 200 IU/ml, 250 IU/ml, 300 IU/ml, 350 IU/ml, 400 IU/ml, 450 IU/ml, 500 IU/ml, 550 IU/ml, 600 IU/ml, 650 IU/ml, 700 IU/ml, 750 IU/ml, 800 IU/ml, 850 IU/ml, 900 IU/ml, 950 IU/ml. In certain embodiments, IL-2 is used at a concentration ranging from 50 IU/ml to 250 IU/ml.
In one embodiment, IL-7 is used at a concentration ranging from 1 ng/ml to 100 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 100 ng/ml" includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
In one embodiment, IL-15 is used at a concentration ranging from 1 ng/ml to 50 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 50 ng/ml" includes, without limitation 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 9 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml. In certain embodiments, IL-15 is used at a concentration ranging from 10 ng/ml to 30 ng/ml.
In one embodiment, TSLP is used at a concentration ranging from 1 ng/ml to 100 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 100 ng/ml" includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
In one embodiment neutralizing antibodies can be added to the culture to prevent the generation of other populations of regulatory T cells. Examples of neutralizing antibodies include, but are not limited to, anti-IFNy, anti-IL-4, and/or anti-IL12 antibodies.
Examples of anti-IFNy antibodies include, but are not limited to, Affymetrix eBioscience (Ref 14-7318), R&D systems (Ref MAB285), Novus Biologicals (Ref AF- 485-NA).
Examples of anti-IL-4 antibodies include, but are not limited to, R&D Systems (Ref MAB304, MAB204, or MAB204), Affymetrix eBioscience (Ref 14-7048), GeneTex (Ref GTX10755).
Examples of anti-IL-12 antibodies include, but are not limited to, Affymetrix eBioscience (Ref 16-7129 or 16-8126), Biolegend (Ref 508803), R&D systems (Ref MAB219, AF-219, or AB-219).
In one embodiment, the culture medium used in the culture of the invention comprises (i) one or more pH buffering system(s); (ii) inorganic salt(s); (iii) trace element(s); (iv) free amino acid(s); (v) vitamin(s); (vi) hormone(s); (vii) carbon/energy source(s). Examples of inorganic salts include, but are not limited to, calcium bromide, calcium chloride, calcium phosphate, calcium nitrate, calcium nitrite, calcium sulphate, magnesium bromide, magnesium chloride, magnesium sulphate, potassium bicarbonate, potassium bromide, potassium chloride, potassium dihydrogen phosphate, potassium disulphate, di- potassium hydrogen phosphate, potassium nitrate, potassium nitrite, potassium sulphite, potassium sulphate, sodium bicarbonate, sodium bromide, sodium chloride, sodium disulphate, sodium hydrogen carbonate, sodium dihydrogen phosphate, di-sodium hydrogen phosphate, sodium sulphate and a mix thereof.
Examples of trace elements include, but are not limited to cobalt (Co), copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), selenium (Se), zinc (Zn) and the salts thereof.
Examples of free amino acids include, but are not limited to L-alanine, L-arginine, L- asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, taurine, L-threonine, L-tryptophan, L-tyrosine, L-valine and a mix thereof
Examples of vitamins include, but are not limited to biotin (vitamin H); D-calcium- pantothenate; choline chloride; folic acid (vitamin B9); myo-inositol; nicotinamide; pyridoxal (vitamin B6); riboflavin (vitamin B2); thiamine (vitamin Bl); cobalamin (vitamin B 12); acid ascorbic; a-tocopherol (vitamin E) and a mix thereof.
Examples of carbon/energy sources include, but are not limited to D-glucose; pyruvate; lactate; ATP; creatine; creatine phosphate; and a mix thereof. In one embodiment, the culture medium is a commercially available cell culture medium, in particular selected in a group comprising the IMDM (Iscove's Modified Dulbecco's Medium) from GIBCO® or the RPMI 1640 medium from GIBCO®.
In another embodiment, the culture medium is a serum-free culture medium such as the AIM-V medium from GIBCO®, the X-VIVO 10, 15 and 20 media from LONZA. In another embodiment, the culture medium can be further supplemented with additional compound(s), in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
In one embodiment, the culture medium is IMDM. In some particular embodiments, the culture medium comprises IMDM cell culture medium; from 1% (w/w) to 5% (w/w) of foetal bovine serum; from 10 IU/ml to 200 IU/ml of penicillin; from 10 IU/ml to 200 IU/ml of streptomycin; from 0.1 mM to 10 mM of a mixture of non-essential amino acids, in particular amino acids selected in a group comprising alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine; from 0.5 mM to 10 mM of glutamine from 10 mM In one embodiment, the medium is a nTreg polarizing medium. The inventors define a "nTreg polarizing medium" as a medium such as RPMI medium comprising at least one cAMP activator as described hereabove, at least one TGF pathway activator as described here above and at least one mTor inhibitor as described hereabove. In a preferred embodiment, the "nTreg polarizing medium" refers to a RPMI medium comprising TGF , rapamycin and PGE2.
In another embodiment, the medium is an inflammatory medium. The inventors define an "inflammatory medium" as a medium such as IMDM comprising inflammatory cytokines such as for example IL-Ιβ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml).
In one embodiment, the culture for generating the invariant Foxp3+ regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days. Within the scope of the invention, the expression "at least 5 days" includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days.
In one embodiment, a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium. Within the scope of the invention the term "portion" is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium. In certain embodiments, 40% (v/v) to 60%) (v/v) of the volume of the culture medium of the first step is discarded. In certain embodiments, the volume that is discarded is replaced with an identical volume of fresh culture medium. Within the scope of the invention, the expression "fresh culture medium" refers to a culture medium that has not been in contact with any CD3+ T cells.
In one embodiment, the method for generating ex vivo invariant Foxp3+ regulatory T cells comprises: - culturing CD3+Va24+ T cells, preferably CD3+Va24+CD45RA+ T cells, in the presence of ACD3 feeder cells and coated 6B11 mAb and the following agents: i) PGE2, ii) TGFp, iii) rapamycin, and optionally iv) IL-2 and/or IL-15, for at least 5 days, thereby obtaining a population of invariant Foxp3+ regulatory T cells ex vivo generated, preferably from invariant naive (CD45RA+) T cells.
In another embodiment, the method for generating ex vivo invariant Foxp3+ regulatory T cells comprises:
- culturing CD3+Va24+ T cells, preferably CD3+Va24+CD45RA+ T cells, in the presence of tolerogenic DCs expressing CDld (incubation with AM580 during about 24h) and pulsed with aGalactosyleramide and in the presence of ACD3 feeder cells and the following agents: i) PGE2, ii) TGFp, iii) rapamycin, and optionally iv) IL-2 and/or IL-15, for at least 5 days, thereby obtaining a population of invariant Foxp3+ regulatory T cells ex vivo generated, preferably from invariant naive (CD45RA+) T cells.
The present invention also relates to an ex vivo method of generation and expansion of invariant Foxp3+ regulatory T cells, comprising: generating the invariant Foxp3+ regulatory T cells as described here above, expanding the invariant Foxp3+ regulatory T cells generated by contacting them with an invariant T cell activator (preferably either ACD3 feeder cells and coated 6B11 mAb or tolerogenic DCs expressing CDld (incubation with AM580 during about 24h) and pulsed with aGalactosyleramide in the presence of ACD3 feeder cells) in the presence of and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator (preferably PGE2), ii) a TGF (Transforming growth factor beta) pathway activator (preferably TGFP), iii) a mTOR inhibitor (preferably rapamycin), and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (preferably IL-2 and/or IL- 15), for at least 5 days, thereby obtaining an expanded population of invariant Foxp3+ regulatory T cells.
In one embodiment, the invariant Foxp3+ regulatory T cell population generated ex vivo is isolated by flow cytometry based on the following phenotype: CD3+Va24+CD45RO+Foxp3+. In one embodiment, the isolated invariant Foxp3+ regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of a polyclonal T cell activator. Examples of polyclonal T cell activator are listed hereinabove. Alternatively, other examples of polyclonal T cell activators that may be used during expansion include, but are not limited to, mitogen such as PMA/ionomycin, super-antigen, anti-CD3 antibody... Preferably, the anti-CD3 monoclonal antibody is coated. In one embodiment, the polyclonal T cell activator can be used in the presence of feeder cells as described here above.
In another embodiment, the isolated invariant Foxp3+ regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of antigen- specific invariant T cell activator as described here above. In one embodiment, the antigen-specific T cell activator can be used in the presence of feeder cells as described here above.
In one embodiment, the culture for expanding the ex vivo generated invariant Foxp3+ regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days. Within the scope of the invention, the expression "at least 5 days" includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days. In one embodiment, a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium. Within the scope of the invention the term "portion" is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium. In certain embodiments, 40%) (v/v) to 60%) (v/v) of the volume of the culture medium of the first step is discarded. In certain embodiments, the volume that is discarded is replaced with an identical volume of fresh culture medium. Within the scope of the invention, the expression "fresh culture medium" refers to a culture medium that has not been in contact with any CD3+ T cells.
In one embodiment, invariant Foxp3+ regulatory T cells are generated ex vivo by culturing CD3+Va24+CD45RA+ T cells obtained from PBMCs by flow cytometry (5.103 cells/ml) in the presence of autologous ACD3 feeder cells (125 105 cells/ml) and coated 6B11 mAb (2 μg/ml) in the presence of PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1 , IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture. Every 3 days, half of the medium volume is discarded and replaced by fresh medium comprising PGE2 (50 nM), TGF (5 ng/ml), Rapamycin (1 nM), IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Once cells begin to expand, they can be split every 2 or 3 days and cultured in the presence of ACD3 feeder cells and coated 6B11 mAb every 9 days in a medium comprising PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml).
In another embodiment, invariant Foxp3+ regulatory T cells are generated ex vivo by culturing CD3+Va24+CD45RA+ T cells (5.103 cells/ml) obtained from PBMCs by flow cytometry (5.103 cells/ml) in the presence of tolerogenic DCs expressing CD Id (incubation with AM580 during about 24h) and pulsed with aGalactosyleramide and in the presence of ACD3 feeder cells (125 105 cells/ml), PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1 , IL-2 (100 Ul/ml), IL- 15 (10 ng/ml) and TGF (5 ng/ml), are added to the culture. Every 3 days, half of the medium volume is discarded and replaced by fresh medium comprising PGE2 (50 nM), TGF (5 ng/ml), Rapamycin (1 nM), IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Once cells begin to expand, they can be split every 2 or 3 days and restimulated every 9 days with tolerogenic DCs pulsed with aGalactosylceramide in the presence of ACD3 feeder cells and PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml). In this embodiment, tolerogenic DCs were obtained by culturing CD14+ monocytes isolated from PBMCs in the presence of AIMV supplemented with GMCSF (lOO ng/ml), IL-4 (10 ng/ml) and AM580 (100 nM). At day 3 and 6, the medium is discarded and replaced by fresh medium comprising GM-CSF and IL-4. On day 6, the tolerogenic DCs are pulsed for 24 hours in the presence of aGalactosylceramide (100 ng/ml) and AM580 (100 nM). yd Foxp3+ regulatory T cells
The present invention also relates to a method for generating ex vivo γδ Foxp3+ regulatory T cells. In one embodiment, the method for generating ex vivo γδ Foxp3+ regulatory T cells, comprises:
- culturing CD3+TCRy5+ T cells, preferably CD3+ TCRy5+ CD45RA+ T cells, in the presence of an γδ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (Thymic stromal lymphopoietin), for at least 5 days, thereby obtaining a population of γδ Foxp3+ regulatory T cells ex vivo generated, preferably from γδ naive (CD45RA+) T cells. In one embodiment, the CD3+ TCRy5+ T cells, preferably CD3+ TCRy5+ CD45RA+ T cells, are obtained by any technic well known in the art from a blood sample. In one embodiment, the CD3+ TCRy5+ T cells, preferably CD3+ TCRy5+ CD45RA+ T cells, are isolated from PBMCs (peripheral blood mononuclear cells) by flow cytometry. In one embodiment, the CD3+ TCRy5+ T cells, preferably CD3+ TCRy5+ CD45RA+ T cells, may be isolated from frozen PBMCs.
In one embodiment, the obtainment of isolated CD3+ TCRy5+ T cells, preferably CD3+ TCRy5+ CD45RA+ T cells, may be improved by an optional first to a purification step. The CD3+ TCRy5+ T cells, preferably CD3+ TCRy5+ CD45RA+ T cells, are stimulated with antigen pulsed tolerogenic DCs (for example ovalbumin pulsed tolerogenic DCs) in the presence of soluble anti-CD28 and anti-CD40 antibodies. In one embodiment, the time of stimulation ranges between 1 hour and 24 hours, preferably between 10 hours and 20 hours, more preferably during about 16 hours. After stimulation, cells are washed, for example with PBS, and stained with anti-CD 154 and anti-CD4 antibodies for sorting. The purified CD3+ TCRy5+ CD154+ T cells are enriched and may be used for the following activation step.
In one embodiment, the CD3+ TCRy5+ T cells are activated in the presence of an γδ T cell activator. Said γδ T cell activator can be a polyclonal γδ T cell activator or an antigen-specific γδ T cell activator.
In the present invention, the polyclonal γδ T cell activator is a TCRy0 activator. Examples of TCRy0 activator include, but are not limited to, anti-TCR γδ antibody such as purified mouse anti-human TCR γδ Clone B l (ref 555715, BD Biosciences), anti- human TCR γδ Antibody (ref 331209, Biolegend), monoclonal TCR γδ Antibody (ref NBP2-22489 or NBP2-22510, Novus Biologicals), anti-mouse γδ TCR (ref 12-571 1-81 , eBioscience), TCR γδ Antibody (ref MAB7297, R&D Systems), anti-T-Cell Receptor γδ antibody (ref ABIN2372990, antibodies-online), anti-TCR gamma + TCR delta antibody (ref ab25663, Abeam), anti-γδ TCR antibody clone IMMU510 (Beckman Coulter); non peptide phosphoantigens (also called phosphorylated non peptide antigens) including but not limited to isoprenyl pyrophosphate (IPP), (E)-4-hydroxy-3- methyl-but-2-enyl diphosphate (HMB-PP) and analogs thereof (such as bromohydrin diphosphate (BrHPP) and 2-methyl-3-butenyl- l -pyrophosphate (2M3B1PP)); Fi- ATPase; apo lipoprotein A-I; Mycobacterium tuberculosis; UL16-binding protein 4 (ULBP4); CD lc; CD Id tetramers loaded with sulphatide; Endothelial protein C receptor (EPCR), Lipoexapeptides; Phycoerythrin, Histidyl-tRNA synthase and butyrophilin 3A1.
In another embodiment, the polyclonal γδ T cell activator is MHC Class-I related A (MICA). In another embodiment, the polyclonal γδ T cell activator is immunogenic apoptotic bodies from cancer cells or derived from tissue lysate.
Cancer cells may derive from tumor biopsy or from expansion of circulatory cancer cells. Immunogenic apoptotic bodies from cancer cells may be obtained for example with anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin, UVC or γ-radiation treated cancer cells releasing apoptotic bodies or can be directly isolated from anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin; UVC or γ-radiation treated cancer. In one embodiment, the polyclonal γδ T cell activator is an anti-TCRy5 antibody or a non peptide phosphoantigen such as isoprenyl pyrophosphate (IPP).
In one embodiment, the polyclonal γδ T cell activator, preferably the anti-TCR γδ antibody, is soluble in the culture medium. In another embodiment, the polyclonal γδ T cell activator is coated to the culture plate. In one embodiment, the polyclonal γδ T cell activator, preferably the anti-TCR γδ antibody, is used in the presence of feeder cells, preferably autologous feeder cells.
Feeder cells include, but are not limited to, ACD3 cells (T cell-depleted accessory cells), irradiated PBMCs, irradiated DCs, artificial APCs (antigen presenting cells), Sf9 cells, insect cells, a pool of PBMCs or a pool of B cells from different subjects, KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL).
Preferably, the feeder cells used in the invention are ACD3 cells that are isolated by negative selection from PBMCs by incubation with anti-CD3 coated beads and then irradiated at 3000 rad.
In one embodiment, the ratio T cells / feeder cells ranges from about 1 : 100 to about 1 : 10 000, preferably from 1 : 1 000 to 1 :5 000. Within the scope of the invention, the expression "from 1 : 100 to 1 : 10 000" includes, without limitation 1 : 100, 1 :200, 1 :300, 1 :400, 1 :500, 1 :600, 1 :700, 1 :800, 1 :900, 1 : 1 000, 1 : 1 250, 1 :1 500, 1 : 1 750, 1 :2 000, 1 :2 250, 1 :2 500, 1 :2 750, 1 :3 000, 1 :3 250, 1 :3 500, 1 : 3 750, 1 :4 000, 1 :4 250, 1 :4 500, 1 : 4 750, 1 :5 000, 1 :5 250, 1 :5 500, 1 :5 750, 1 :6 000, 1 :6 250, 1 :6 500, 1 :6 750, 1 :7 000, 1 :7 250, 1 :7 500, 1 :7 750, 1 :8 000, 1 :8 250, 1 :8 500, 1 : 8 750, 1 :9 000, 1 :9 250, 1 :9 500, 1 :9 750 and 1 : 10 000. In the present invention, the antigen-specific γδ T cell activator is tolerogenic dendritic cells (DCs).
As used herein, "tolerogenic DCs" refers to DCs capable to induce tolerance. In one embodiment, tolerogenic DCs are capable of secreting more suppressive cytokines such as IL-10 and TGF than proinflammatory cytokines such as IL-12, IL-23 or TNFa. In one embodiment, DCs are defined as tolerogenic when they secrete IL-10 and IL-12 in a ratio IL-10: IL-12 > 1.
In one embodiment, tolerogenic DCs express on their surface the major histocompatibility (MHC) class la and/or MHC class lb. The MHC class la presentation refers to the "classical" presentation through HLA-A, HLA-B and/or HLA-C molecules whereas the MHC class lb presentation refers to the "non-classical" antigen presentation through HLA-E, HLA-F, HLA-G and/or HLA-H molecules.
In one embodiment, tolerogenic DCs express 50% of MHC class la molecules and 50%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 45%> of MHC class la molecules and 55%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 40%> of MHC class la molecules and 60% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 35%> of MHC class la molecules and 65%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 30% of MHC class la molecules and 70% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 25%> of MHC class la molecules and 75%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 20% of MHC class la molecules and 80% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 15%> of MHC class la molecules and 85%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 10% of MHC class la molecules and 90% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 5% of MHC class la molecules and 95% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express only MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 50% of HLA-A, HLA-B and/or HLA-C molecules and 50% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 45% of HLA-A, HLA-B and/or HLA-C molecules and 55% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 40% of HLA-A, HLA-B and/or HLA-C molecules and 60% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 35% of HLA-A, HLA-B and/or HLA-C molecules and 65% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 30% of HLA-A, HLA-B and/or HLA-C molecules and 70% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 25% of HLA-A, HLA-B and/or HLA-C molecules and 75% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 20% of HLA-A, HLA-B and/or HLA-C molecules and 80% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 15% of HLA-A, HLA-B and/or HLA-C molecules and 85% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 10% of HLA-A, HLA-B and/or HLA-C molecules and 90%> of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 5% of HLA-A, HLA-B and/or HLA-C molecules and 95% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express only HLA-E molecules on their surface.
Methods for obtaining tolerogenic DCs are well-known in the art. An exemplary method is the generation of tolerogenic DCs from CD14+ monocytes. For example, CD14+ monocytes are cultured in the presence of GM-CSF and IL-4, or in the presence of GM-CSF and IFNa, for the generation of immature DCs.
Methods for inhibiting MHC class la molecules expression or inducing the expression of HLA-E molecules on the surface of tolerogenic DCs are well-known. The inhibition of the TAP transporter (transporter associated with antigen processing) leads to a decreased expression of MHC class la molecules thereby promoting HLA-E molecules expression on the surface of tolerogenic DCs.
Exemplary methods to inhibit the TAP transporter in the endoplasmic reticulum include, but are not limited to, CRISPR-CAS-9 technology, silencing RNA, transfected DCs with the UL-10 viral protein from the CMV (cytomegalovirus) or the use of viral proteins.
Examples of viral proteins able to inhibit the TAP transporter include, but are not limited to, HSV-1 ICP47 protein, varicella- virus UL49.5 protein, cytomegalovirus US6 protein or gammaherpesvirus EBV BNLF2a protein.
Another method is the use of a chemical product to inhibit the expression of MHC class la molecules without changing HLA-E expression on the surface of tolerogenic DCs. Examples of chemical products include, but are not limited to, 5'- methyl-5 '- thioadenosine or leptomycin B. The tolerogenic DCs are pulsed in the presence of at least one bisphosphonate, preferably aminobiphosphonate, during about 24h. Examples of biphosphonate include, but are not limited to, zoledronic acid (or zoledronate), pamidronic acid, alendronic acid, risedronic acid, ibandronic acid, incadronic acid, etidronic acid, tiludronic acid, a combination thereof, a salt thereof and a hydrate thereof. Preferably, the biphosphanate is zoledronic acid or zoledronate.
In one embodiment, biphosphonate, in particular zoledronic acid, is used at a concentration from 10 nM to 50 μΜ. Within the scope of the invention, the expression "from 10 nM to 50 μΜ" includes, without limitation 50 nM, 100 nM, 250 nM, 500 nM, 750 nM, 1 μΜ, 10 μΜ, 20 μΜ, 30 μΜ, 40 μΜ, 50 μΜ. In one embodiment, the cAMP activator added in the culture allows the activation of the cAMP pathway. Examples of cAMP activator include, but are not limited to, PGE2 (prostaglandin E2), an EP2 or EP4 agonist, a membrane adenine cyclase activator such as forskolin, or metabotropic glutamate receptors agonists. Examples of PGE2 include, but are not limited to, PGE2 of ref P5640 or P0409 (Sigma- Aldrich), PGE2 of ref 2296 (R&D Systems), PGE2 of ref 2268 (Bio Vision), PGE2 of ref 72192 (Stemcell), PGE2 of ref abl44539 (Abeam), and PGE2 of ref 14010 (Cayman Chemical).
In one embodiment, the cAMP activator, preferably PGE2 is used at a concentration ranging from 0.01 μΜ to 10 μΜ. Within the scope of the invention, the expression "from 0.01 μΜ to 10 μΜ" includes, without limitation 0.02 μΜ, 0.03 μΜ, 0.04 μΜ, 0.05 μΜ, 0.06 μΜ, 0.07 μΜ, 0.08 μΜ, 0.09 μΜ, 0.1 μΜ, 0.2 μΜ, 0.3 μΜ, 0.4 μΜ, 0.5 μΜ, 0.6 μΜ, 0.7 μΜ, 0.8 μΜ, 0.9 μΜ, 1 μΜ, 1.5 μΜ, 2 μΜ, 2.5 μΜ, 3 μΜ, 3.5 μΜ, 4 μΜ, 4.5 μΜ, 5 μΜ, 6 μΜ, 7 μΜ, 8 μΜ, 9 μΜ. In certain embodiments, PGE2 is at a concentration ranging from 0.03 μΜ to 1.5 μΜ.
In one embodiment, the TGFP pathway activator added in the culture allows the activation of the TGFP pathway. Examples of TGFP pathway activators include, but are not limited to, TGFP family (TGFpi, TGFP2, TGFP3), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, and nodal. Examples of TGFP include, but are not limited to, TGFpi of ref T7039 (Sigma- Aldrich), TGFp2 of ref T2815 (Sigma- Aldrich), TGFp3 of ref T5425 (Sigma- Aldrich), human TGFpl of ref P01137 (R&D system), human TGFpl of ref 580702 (Biolegend), TGFpl of ref HZ-1011 (HumanZyme), human TGFpl of ref 14- 8348-62 (Affymetrix eBioscience).
In one embodiment, the pathway activator is used at a concentration ranging from 1 ng/ml to 20 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 20 ng/ml" includes, without limitation 2 ng/ml, 2.5 ng/ml, 3 ng/ml, 3.5 ng/ml, 4 ng/ml, 4.5 ng/ml, 5 ng/ml, 5.5 ng/ml, 6 ng/ml, 6.5 ng/ml, 7 ng/ml, 7.5 ng/ml, 8 ng/ml, 8.5 ng/ml, 9 ng/ml, 9.5 ng/ml, 10 ng/ml, 11 ng/ml, 12 ng/ml, 13 ng/ml, 14 ng/ml, 15 ng/ml, 16 ng/ml, 17 ng/ml, 18 ng/ml, 19 ng/ml. In certain embodiments, TGF is at a concentration ranging from 2.5 ng/ml to 7.5 ng/ml. In one embodiment, the mTOR inhibitor added in the culture allows the inhibition of the mTOR pathway. Examples of mTOR inhibitor include, but are not limited to, rapamycin (also named sirolimus) and its analogs (termed rapalogs); wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG); curcumin; resveratrol; genistein; 3, 3-diindolylmethane (DIM); LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l- benzopyran-4-one); PP242; PP30; Torinl ; Ku-0063794; WAY-600; WYE-687; WYE- 354; and mTOR and PI3K dual-specificity inhibitors such as GNE477, NVP-BEZ235, PI-103, XL765 and WJD008. Examples of rapamycin include, but are not limited to, rapamycin of ref R0395 (Sigma- Aldrich), rapamycin of ref SI 039 (Selleckchem), rapamycin of ref 1292 (Tocris), rapamycin of ref R-5000 (LC Laboratories), rapamycin of ref tlrl-rap (InvivoGen), rapamycin of ref abl20224 (Abeam), rapamycin of ref R0395 (Sigma-Aldrich).
Examples of compounds of the same chemical class than rapamycin used clinically include, but are not limited to, Everolimus (code name RADOOl), Temsirolimus (code name CCI-779, NSC 683864), Zotarolimus (code name ABT-578).
In one embodiment, the mTOR inhibitor, preferably rapamycin, is used at a concentration ranging from 0.1 nM to 50 iiM. Within the scope of the invention, the expression "from 0.1 nM to 50 nM" includes, without limitation 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 26 nM, 27 nM, 28 nM, 29 nM, 30 nM, 31 nM, 32 nM, 33 nM, 34 nM, 35 nM, 36 nM, 37 nM, 38 nM, 39 nM, 40 nM, 41 nM, 42 nM, 43 nM, 44 nM, 45 nM, 46 nM, 47 nM, 48 nM, 49 nM.
In one embodiment, at least one cytokine selected from IL-2, IL-7, IL-15 and TSLP can be added in the culture.
In one embodiment, IL-2 is used at a concentration ranging from 10 IU/ml to 1000 IU/ml. Within the scope of the invention, the expression "from 10 IU/ml to 1000 IU/ml" includes, without limitation 15 IU/ml, 20 IU/ml, 25 IU/ml, 30 IU/ml, 35 IU/ml, 40 IU/ml, 45 IU/ml, 50 IU/ml, 55 IU/ml, 60 IU/ml, 65 IU/ml, 70 IU/ml, 75 IU/ml, 80 IU/ml, 85 IU/ml, 90 IU/ml, 95 IU/ml, 100 IU/ml, 150 IU/ml, 200 IU/ml, 250 IU/ml, 300 IU/ml, 350 IU/ml, 400 IU/ml, 450 IU/ml, 500 IU/ml, 550 IU/ml, 600 IU/ml, 650 IU/ml, 700 IU/ml, 750 IU/ml, 800 IU/ml, 850 IU/ml, 900 IU/ml, 950 IU/ml. In certain embodiments, IL-2 is used at a concentration ranging from 50 IU/ml to 250 IU/ml.
In one embodiment, IL-7 is used at a concentration from 1 ng/ml to 100 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 100 ng/ml" includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
In one embodiment, IL-15 is used at a concentration ranging from 1 ng/ml to 50 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 50 ng/ml" includes, without limitation 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 9 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml. In certain embodiments, IL-15 is used at a concentration ranging from 10 ng/ml to 30 ng/ml.
In one embodiment, TSLP is used at a concentration ranging from 1 ng/ml to 100 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 100 ng/ml" includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
In one embodiment neutralizing antibodies can be added to the culture to prevent the generation of other populations of regulatory T cells.
Examples of neutralizing antibodies include, but are not limited to, anti-IFNy, anti-IL-4, and/or anti-IL12 antibodies.
Examples of anti-IFNy antibodies include, but are not limited to, Affymetrix eBioscience (Ref 14-7318), R&D systems (Ref MAB285), Novus Biologicals (Ref AF- 485-NA). Examples of anti-IL-4 antibodies include, but are not limited to, R&D Systems (Ref MAB304, MAB204, or MAB204), Affymetrix eBioscience (Ref 14-7048), GeneTex (Ref GTX10755).
Examples of anti-IL-12 antibodies include, but are not limited to, Affymetrix eBioscience (Ref 16-7129 or 16-8126), Biolegend (Ref 508803), R&D systems (Ref MAB219, AF-219, or AB-219).
In one embodiment, the culture medium used in the culture of the invention comprises (i) one or more pH buffering system(s); (ii) inorganic salt(s); (iii) trace element(s); (iv) free amino acid(s); (v) vitamin(s); (vi) hormone(s); (vii) carbon/energy source(s). Examples of inorganic salts include, but are not limited to, calcium bromide, calcium chloride, calcium phosphate, calcium nitrate, calcium nitrite, calcium sulphate, magnesium bromide, magnesium chloride, magnesium sulphate, potassium bicarbonate, potassium bromide, potassium chloride, potassium dihydrogen phosphate, potassium disulphate, di- potassium hydrogen phosphate, potassium nitrate, potassium nitrite, potassium sulphite, potassium sulphate, sodium bicarbonate, sodium bromide, sodium chloride, sodium disulphate, sodium hydrogen carbonate, sodium dihydrogen phosphate, di-sodium hydrogen phosphate, sodium sulphate and a mix thereof.
Examples of trace elements include, but are not limited to, cobalt (Co), copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), selenium (Se), zinc (Zn) and the salts thereof.
Examples of free amino acids include, but are not limited to, L-alanine, L-arginine, L- asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, taurine, L-threonine, L-tryptophan, L-tyrosine, L-valine and a mix thereof.
Examples of vitamins include, but are not limited to, biotin (vitamin H); D-calcium- pantothenate; choline chloride; folic acid (vitamin B9); myo-inositol; nicotinamide; pyridoxal (vitamin B6); riboflavin (vitamin B2); thiamine (vitamin Bl); cobalamin (vitamin B 12); acid ascorbic; a-tocopherol (vitamin E) and a mix thereof.
Examples of carbon/energy sources include, but are not limited to, D-glucose; pyruvate; lactate; ATP; creatine; creatine phosphate; and a mix thereof. In one embodiment, the culture medium is a commercially available cell culture medium, in particular selected in a group comprising the IMDM (Iscove's Modified Dulbecco's Medium) from GIBCO® or the RPMI 1640 medium from GIBCO®.
In another embodiment, the culture medium is a serum-free culture medium such as the AIM-V medium from GIBCO®, the X-VIVO 10, 15 and 20 media from LONZA. In another embodiment, the culture medium can be further supplemented with additional compound(s), in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
In one embodiment, the culture medium is IMDM. In some particular embodiments, the culture medium comprises IMDM cell culture medium; from 1% (w/w) to 5% (w/w) of foetal bovine serum; from 10 IU/ml to 200 IU/ml of penicillin; from 10 IU/ml to 200 IU/ml of streptomycin; from 0.1 mM to 10 mM of a mixture of non-essential amino acids, in particular amino acids selected in a group comprising alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine; from 0.5 mM to 10 mM of glutamine from 10 mM to 25 mM of HEPES pH 7.6-7.8.
In one embodiment, the medium is a nTreg polarizing medium. The inventors define a "nTreg polarizing medium" as a medium such as RPMI medium comprising at least one cAMP activator as described hereabove, at least one TGF pathway activator as described here above and at least one mTor inhibitor as described hereabove. In a preferred embodiment, the "nTreg polarizing medium" refers to a RPMI medium comprising TGF , rapamycin and PGE2. In another embodiment, the medium is an inflammatory medium. The inventors define an "inflammatory medium" as a medium such as IMDM comprising inflammatory cytokines such as for example IL-Ιβ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml). In one embodiment, the culture for generating the γδ Foxp3+ regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days. Within the scope of the invention, the expression "at least 5 days" includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days. In one embodiment, a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium. Within the scope of the invention the term "portion" is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium. In certain embodiments, 40%) (v/v) to 60%) (v/v) of the volume of the culture medium of step a) is discarded. In certain embodiments, the volume that is discarded is replaced with an identical volume of fresh culture medium. Within the scope of the invention, the expression "fresh culture medium" refers to a culture medium that has not been in contact with any CD3+ T cells.
In one embodiment, the method for generating ex vivo γδ Foxp3+ regulatory T cells, comprises:
- culturing CD3+TCR γδ + T cells, preferably CD3+ TCR γδ + CD45RA+ T cells, in the presence of autologous ACD3 feeder cells and coated anti-TCR γδ antibody and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, thereby obtaining a population of γδ Foxp3+ regulatory T cells ex vivo generated, preferably from γδ naive (CD45RA+) T cells.
In one embodiment, the method for generating ex vivo γδ Foxp3+ regulatory T cells, comprises: - culturing CD3+TCR γδ + T cells, preferably CD3+ TCR γδ + CD45RA+ T cells, in the presence of tolerogenic DC that have been pulsed with zoledronate during about 24h and in the presence of ACD3 feeder cells and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, - thereby obtaining a population of γδ Foxp3+ regulatory T cells ex vivo generated, preferably from γδ naive (CD45RA+) T cells.
The present invention also relates to an ex vivo method of generation and expansion of γδ Foxp3+ regulatory T cells, comprising: generating the γδ Foxp3+ regulatory T cells as described here above, - expanding the γδ Foxp3+ regulatory T cells generated by contacting them in the presence of an γδ T cell activator (preferably either autologous ACD3 feeder cells and coated anti-TCR γδ antibody or tolerogenic DC that have been pulsed with zoledronate during about 24h and in the presence of ACD3 feeder cells) and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator (preferably PGE2), ii) a TGF (Transforming growth factor beta) pathway activator (preferably TGFP), iii) a mTOR inhibitor (preferably rapamycin), and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (preferably IL-2 and/or IL-15), for at least 5 days, thereby obtaining an expanded population of γδ Foxp3+ regulatory T cells. In one embodiment, the γδ Foxp3+ regulatory T cell population generated ex vivo is isolated by flow cytometry based on the following phenotype: CD3+TCR^+CD45RO+Foxp3+. In one embodiment, the isolated γδ Foxp3+ regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of a polyclonal γδ T cell activator. Examples of polyclonal γδ T cell activator are listed hereinabove. Alternatively, other examples of polyclonal γδ T cell activators that may be used during expansion include, but are not limited to, mitogen such as PMA/ionomycin, super- antigen, anti-CD3 antibody... Preferably, the anti-CD3 monoclonal antibody is coated. In one embodiment, the polyclonal γδ T cell activator can be used in the presence of feeder cells as described here above.
In another embodiment, the isolated γδ Foxp3+ regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of an antigen- specific γδ T cell activator as described here above. In one embodiment, the antigen- specific T cell activator can be used in the presence of feeder cells as described here above.
In one embodiment, the culture for expanded the ex vivo generated γδ Foxp3+ regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days. Within the scope of the invention, the expression "at least 5 days" includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days or more.
In one embodiment, a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium. Within the scope of the invention the term "portion" is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium. In certain embodiments, 40%) (v/v) to 60%) (v/v) of the volume of the culture medium of the first step is discarded. In certain embodiments, the volume that is discarded is replaced with an identical volume of fresh culture medium. Within the scope of the invention, the expression "fresh culture medium" refers to a culture medium that has not been in contact with any CD3+ T cells.
In one embodiment, γδ Foxp3+ regulatory T cells are generated ex vivo by culturing CD3+TCRy5+CD45RA+ T cells obtained from PBMCs by flow cytometry (5.103 cells/ml) in the presence of autologous ACD3 feeder cells (125 105 cells/ml) and coated anti-TCRy5 antibody (2 μg/ml) in the presence of PGE2 (1 μΜ), TGFP (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture. Every 3 days, half of the medium volume is discarded and replaced by fresh medium comprising PGE2 (50 nM), TGFP (5 ng/ml), Rapamycin (1 nM), IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Once cells begin to expand, they can be split every 2 or 3 days and cultured in the presence of ACD3 feeder cells and coated anti-TCRy5 antibody every 9 days in a medium comprising PGE2 (1 μΜ), TGFp (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml).
In another embodiment, γδ Foxp3+ regulatory T cells are generated ex vivo by culturing CD3+TCRy5+CD45RA+ T cells (5.103 cells/ml) obtained from PBMCs by flow cytometry (5.103 cells/ml) in the presence of tolerogenic DCs, that have been pulsed with zoledronate during about 24h, and in the presence of ACD3 feeder cells (1.25 105 cells/ml), PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1, IL-2 (100 Ul/ml), IL-15 (10 ng/ml) and TGF (5 ng/ml), are added to the culture. Every 3 days, half of the medium volume is discarded and replaced by fresh medium comprising PGE2 (50 nM), TGF (5 ng/ml), Rapamycin (1 nM), IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Once cells begin to expand, they can be split every 2 or 3 days and restimulated every 9 days with tolerogenic DCs in the presence of ACD3 feeder cells and PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml).
In this embodiment, tolerogenic DCs were obtained by culturing CD14+ monocytes isolated from PBMCs in the presence of AIMV supplemented with GMCSF (100 ng/ml) and IL-4 (10 ng/ml). At day 3 and 6, the medium is discarded and replaced by fresh medium comprising GM-CSF and IL-4. On day 6, the tolerogenic DCs are pulsed for 24 hours in the presence of zoledronate (100 nM).
MHCII restricted CD4+ Foxp3+ regulatory T cells
The present invention also relates to a method for generating ex vivo MHCII restricted CD4+ Foxp3+ regulatory T cells.
In one embodiment, the method for generating ex vivo MHCII restricted CD4+ Foxp3+ regulatory T cells, comprises:
- culturing CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, in the presence of a TCRa activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (Thymic stromal lymphopoietin), for at least 5 days, thereby obtaining a population of MHCII restricted CD4+ Foxp3+ regulatory T cells ex vivo generated, preferably from naive (CD45RA+) T cells.
In one embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are obtained by any technic well known in the art from a blood sample. In one embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25" CD45RA+T cells, are isolated from PBMCs (peripheral blood mononuclear cells) by flow cytometry or by negative selection using a MACS system for example.
In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD62L+.
In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD127+. In another embodiment, CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD27+. In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD62L+ CD127+.
In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD62L+ CD27+. In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD127+ CD27+.
In another embodiment, CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are CD62L+ CD127+ CD27+.
In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are TCRy5\
In another embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, are Va24\
In one embodiment, the CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, may be isolated from frozen PBMCs. In one embodiment, the obtainment of isolated CD3+ CD4+ CD25" T cells, preferably CD3+ CD4+ CD25 CD45RA+ T cells, may be improved by an optional first to a purification step. The CD3+ CD4+ CD25 T cells, preferably CD3+ CD4+ CD25" CD45RA+ T cells, are stimulated with antigen pulsed tolerogenic DCs (for example ovalbumin pulsed tolerogenic DCs) in the presence of soluble anti-CD28 and anti-CD40 antibodies. In one embodiment, the time of stimulation ranges between 1 hour and 24 hours, preferably between 10 hours and 20 hours, more preferably during about 16 hours. After stimulation, cells are washed, for example with PBS, and stained with anti- CD 154 and anti-CD4 antibodies for sorting. The purified CD3+ CD4+ CD25 CD154+ T cells are enriched and may be used for the following activation step. In one embodiment, the CD3+ CD4+ CD25" T cells are activated in the presence of an aPTCR cell activator. Said a TCR cell activator can be a polyclonal a TCR cell activator or an antigen-specific a TCR cell activator. In the present invention, the polyclonal a^TCR cell activator is a TCR αβ activator. Examples of TCR αβ activator include, but are not limited to, anti-TCR αβ antibody such as purified anti-human TCR α/β antibody (ref 306702, Biolegend), Anti-Human alpha beta TCR antibody (ref 11-9986-41, eBioscience), anti- human TCR αβ (ref 563826, BD Biosciences), TCR alpha/beta antibody (ref GTX80083, GeneTex); anti- CD3 antibody such as purified anti-human CD3 antibody (ref 344801, BioLegend), anti-CD3 antibody (ab5690, Abeam), anti-human CD3 purified (ref 14-0038-80, eBioscience), CD3 antibody (ref MA5- 17043, Invitrogen antibodies), CD3 monoclonal antibody (ref ALX-804-822-C100, Enzo Life Sciences), human CD3 antibody (ref 130- 098-162, Miltenyi Biotec); mitogen such as pokeweed mitogen, ionomycin, phorbol myristate acetate (PMA), phytohaemagglutinin (PHA), lipopolysaccharide (LPS), superantigen such as staphylococcal enterotoxins (SPE), retroviral antigens, streptococcal antigens, mycoplasma antigens, mycobacterium antigens, viral antigens (e.g., a superantigen from mouse mammary tumor virus, rabies virus or herpes virus) and endoparasitic antigens (e.g., protozoan or helminth antigens).
In one embodiment, the polyclonal TCRa-β cell activator is an anti-TCRa^ antibody or an anti-CD3 antibody.
In one embodiment, the polyclonal TCRa-β cell activator, preferably the anti-TCRa^ or anti-CD3 antibody, is soluble in the culture medium. In another embodiment, the polyclonal TCR αβ cell activator is coated to the culture plate.
In one embodiment, the polyclonal TCRa-β cell activator is used in the presence of feeder cells, preferably autologous feeder cells.
Feeder cells include, but are not limited to, ACD3 cells (T cell-depleted accessory cells), irradiated PBMCs, irradiated DCs, artificial APCs (antigen presenting cells), Sf9 cells, insect cells, a pool of PBMCs or a pool of B cells from different subjects, KCD40L cells EBV-trans formed B cell lines and EBV-trans formed lymphoblastoid cells (LCL). Preferably, the feeder cells used in the invention are ACD3 cells that are isolated by negative selection from PBMCs by incubation with anti-CD3 coated beads and then irradiated at 3000 rad.
In one embodiment, the ratio T cells / feeder cells ranges from about 1:100 to about 1:10000, preferably from 1:1000 to 1:5000. Within the scope of the invention, the expression "from 1:100 to 1:10000" includes, without limitation 1:100, 1:200, 1:300, 1:400, 1:500, 1:600, 1:700, 1:800, 1:900, 1:1000, 1:1250, 1:1500, 1: 1750, 1:2000, 1:2250, 1:2500, 1:2750, 1:3000, 1:3250, 1:3500, 1: 3750, 1:4000, 1:4250, 1:4500, 1: 4750, 1:5000, 1:5250, 1:5500, 1:5750, 1:6000, 1:6250, 1:6500, 1:6750, 1:7000, 1:7250, 1 :7500, 1 :7750, 1 :8000, 1 :8250, 1 :8500, 1 : 8750, 1 :9000, 1 :9250, 1 :9500, 1:9750 and 1:10000.
In the present invention, the antigen-specific TCRa cell activator is tolerogenic dendritic cells (DCs).
As used herein, "tolerogenic DCs" refers to DCs capable to induce tolerance. In one embodiment, tolerogenic DCs are capable of secreting more suppressive cytokines such as IL-10 and TGF than proinflammatory cytokines such as IL-12, IL-23 or TNFa. In one embodiment, DCs are defined as tolerogenic when they secrete IL-10 and IL-12 in a ratio IL-10: IL-12 > 1.
In one embodiment, tolerogenic DCs express on their surface the major histocompatibility (MHC) class la and/or MHC class lb. The MHC class la presentation refers to the "classical" presentation through HLA-A, HLA-B and/or HLA-C molecules whereas the MHC class lb presentation refers to the "non-classical" antigen presentation through HLA-E, HLA-F, HLA-G and/or HLA-H molecules.
In one embodiment, tolerogenic DCs express 50% of MHC class la molecules and 50% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 45%> of MHC class la molecules and 55%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 40%> of MHC class la molecules and 60% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 35%> of MHC class la molecules and 65%> of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 30% of MHC class la molecules and 70% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 25% of MHC class la molecules and 75% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 20% of MHC class la molecules and 80% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 15% of MHC class la molecules and 85% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 10% of MHC class la molecules and 90% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express 5% of MHC class la molecules and 95% of MHC class lb molecules on their surface. In one embodiment, tolerogenic DCs express only MHC class lb molecules on their surface.
In one embodiment, tolerogenic DCs express 50% of HLA-A, HLA-B and/or HLA-C molecules and 50% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 45% of HLA-A, HLA-B and/or HLA-C molecules and 55% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 40% of HLA-A, HLA-B and/or HLA-C molecules and 60% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 35% of HLA-A, HLA-B and/or HLA-C molecules and 65% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 30% of HLA-A, HLA-B and/or HLA-C molecules and 70% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 25% of HLA-A, HLA-B and/or HLA-C molecules and 75% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 20% of HLA-A, HLA-B and/or HLA-C molecules and 80% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 15% of HLA-A, HLA-B and/or HLA-C molecules and 85% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 10% of HLA-A, HLA-B and/or HLA-C molecules and 90% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express 5% of HLA-A, HLA-B and/or HLA-C molecules and 95% of HLA-E molecules on their surface. In one embodiment, tolerogenic DCs express only HLA-E molecules on their surface. Methods for obtaining tolerogenic DCs are well-known in the art. An exemplary method is the generation of tolerogenic DCs from CD14+ monocytes. For example, CD14+ monocytes are cultured in the presence of GM-CSF and IL-4, or in the presence of GM-CSF and IFNa, for the generation of immature DCs.
Methods for inhibiting MHC class la molecules expression or inducing the expression of HLA-E molecules on the surface of tolerogenic DCs are well-known.
The inhibition of the TAP transporter (transporter associated with antigen processing) leads to a decreased expression of MHC class la molecules thereby promoting HLA-E molecules expression on the surface of tolerogenic DCs.
Exemplary methods to inhibit the TAP transporter in the endoplasmic reticulum include, but are not limited to, CRISPR-CAS-9 technology, silencing RNA, transfected DCs with the UL-10 viral protein from the CMV (cytomegalovirus) or the use of viral proteins.
Examples of viral proteins able to inhibit the TAP transporter include, but are not limited to, HSV-1 ICP47 protein, varicella- virus UL49.5 protein, cytomegalovirus US6 protein or gammaherpesvirus EBV BNLF2a protein.
Another method is the use of a chemical product to inhibit the expression of MHC class la molecules without changing HLA-E expression on the surface of tolerogenic DCs. Examples of chemical products include, but are not limited to, 5'- methyl-5 '- thioadenosine or leptomycin B.
The tolerogenic DCs are pulsed in the presence of at least one self-peptide antigen, modified self-peptide antigen, over-expressed self-peptide antigen or foreign antigen. By "self-peptide antigen" is meant an antigen that is normally expressed in the body from which the regulatory T cells are derived. In another embodiment, self-antigen is comparable to one, or, in another embodiment, indistinct from one normally expressed in a body from which the regulatory T cells are derived, though may not directly correspond to the antigen. In another embodiment, self-antigen refers to an antigen, which when expressed in a body, may result in the education of self-reactive T cells. In one embodiment, self-antigen is expressed in an organ that is the target of an autoimmune disease. In one embodiment, the self-antigen is expressed in a pancreas, thyroid, connective tissue, kidney, lung, digestive system or nervous system. In another embodiment, self-antigen is expressed on pancreatic β cells. Examples of self-peptide antigen, modified self-peptide antigen and over-expressed self-peptide antigen include, but are not limited to, antigenic peptides of insulin, insulin beta, glutamic acid decarboxylase 1 (GAD1), glutamic acid decarboxylase 65 (GAD 65), HSP, thyro globulin, nuclear proteins, acetylcholine receptor, collagen, thyroid stimulating hormone receptor (TSHR), ICA512(IA-2) and ΙΑ-2β (phogrin), carboxypeptidase H, ICA69, ICA12, thyroid peroxidase, native DNA, myelin basic protein, myelin proteolipid protein, acetylcholine receptor components, histocompatibility antigens, antigens involved in graft rejection and altered peptide ligands.
In another embodiment, the self-peptide antigen is derived from immunogenic apoptotic bodies from cancer cells or derived from tissue lysate.
Cancer cells may derive from tumor biopsy or from expansion of circulatory cancer cells.
Immunogenic apoptotic bodies from cancer cells may be obtained for example with anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin, UVC or γ-radiation treated cancer cells releasing apoptotic bodies or can be directly isolated from anthracyclines including doxorubicin, daunorubicin, idarubicin and mitoxanthrone; oxaliplatin; UVC or γ-radiation treated cancer.
Examples of tissue lysate include, but are not limited to, synovial liquid or inflammatory tissue lysate. By "foreign antigen" is meant a molecule or molecules which is/are not endogenous or native to a mammal which is exposed to it. The foreign antigen may elicit an immune response, e.g. a humoral and/or T cell mediated response in the mammal. Generally, the foreign antigen will result in the production of antibodies there against. Examples of foreign antigens include, but are not limited to, proteins (including a modified protein such as a glycoprotein, a mucoprotein, etc.), nucleic acids, carbohydrates, proteoglycans, lipids, mucin molecules, immunogenic therapeutic agents (including proteins such as antibodies, particularly antibodies comprising non-human amino acid residues, e.g. rodent, chimeric/humanized, and primatized antibodies), toxins (optionally conjugated to a targeting molecule such as an antibody, wherein the targeting molecule may also be immunogenic), gene therapy viral vectors (such as retroviruses and adenoviruses), grafts (including antigenic components of the graft to be transplanted into the heart, lung, liver, pancreas, kidney of graft recipient and neural graft components), infectious agents (such as bacteria and virus or other organism, e.g., protists), alloantigens (i.e. an antigen that occurs in some, but not in other members of the same species) such as differences in blood types, human lymphocyte antigens (HLA), platelet antigens, antigens expressed on transplanted organs, blood components, pregnancy (Rh), and hemophilic factors (e.g. Factor VTfl and Factor IX). In one embodiment, the self-peptide antigen or the foreign antigen is soluble.
In one embodiment, the cAMP activator added in the culture allows the activation of the cAMP pathway. Examples of cAMP activator include, but are not limited to PGE2 (prostaglandin E2), an EP2 or EP4 agonist, a membrane adenine cyclase activator such as forskolin, or metabotropic glutamate receptors agonists. Examples of PGE2 include, but are not limited to, PGE2 of ref P5640 or P0409 (Sigma- Aldrich), PGE2 of ref 2296 (R&D Systems), PGE2 of ref 2268 (Bio Vision), PGE2 of ref 72192 (Stemcell), PGE2 of ref abl44539 (Abeam ), and PGE2 of ref 14010 (Cayman Chemical).
In one embodiment, the cAMP activator, preferably PGE2 is used at a concentration ranging from 0.01 μΜ to 10 μΜ. Within the scope of the invention, the expression "from 0.01 μΜ to 10 μΜ" includes, without limitation 0.02 μΜ, 0.03 μΜ, 0.04 μΜ, 0.05 μΜ, 0.06 μΜ, 0.07 μΜ, 0.08 μΜ, 0.09 μΜ, 0.1 μΜ, 0.2 μΜ, 0.3 μΜ, 0.4 μΜ, 0.5 μΜ, 0.6 μΜ, 0.7 μΜ, 0.8 μΜ, 0.9 μΜ, 1 μΜ, 1.5 μΜ, 2 μΜ, 2.5 μΜ, 3 μΜ, 3.5 μΜ, 4 μΜ, 4.5 μΜ, 5 μΜ, 6 μΜ, 7 μΜ, 8 μΜ, 9 μΜ. In certain embodiments, PGE2 is at a concentration ranging from 0.03 μΜ to 1.5 μΜ. In one embodiment, the TGFP pathway activator added in the culture allows the activation of the TGFP pathway. Examples of TGFP pathway activators include, but are not limited to, TGFP family (TGFpi, TGFP2, TGFP3), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, and nodal. Examples of TGFP include, but are not limited to, TGFpi of ref T7039 (Sigma- Aldrich), TGFp2 of ref T2815 (Sigma- Aldrich), TGFp3 of ref T5425 (Sigma- Aldrich), human TGFpl of ref P01137 (R&D system), human TGFpl of ref 580702 (Biolegend), TGFpl of ref HZ-1011 (HumanZyme), human TGFpl of ref 14- 8348-62 (Affymetrix eBioscience). In one embodiment, the pathway activator is used at a concentration ranging from 1 ng/ml to 20 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 20 ng/ml" includes, without limitation 2 ng/ml, 2.5 ng/ml, 3 ng/ml, 3.5 ng/ml, 4 ng/ml, 4.5 ng/ml, 5 ng/ml, 5.5 ng/ml, 6 ng/ml, 6.5 ng/ml, 7 ng/ml, 7.5 ng/ml, 8 ng/ml, 8.5 ng/ml, 9 ng/ml, 9.5 ng/ml, 10 ng/ml, 11 ng/ml, 12 ng/ml, 13 ng/ml, 14 ng/ml, 15 ng/ml, 16 ng/ml, 17 ng/ml, 18 ng/ml, 19 ng/ml. In certain embodiments, TGF is at a concentration ranging from 2.5 ng/ml to 7.5 ng/ml.
In one embodiment, the mTOR inhibitor added in the culture allows the inhibition of the mTOR pathway. Examples of mTOR inhibitor include, but are not limited to, rapamycin (also named sirolimus) and its analogs (termed rapalogs); wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG); curcumin; resveratrol; genistein; 3, 3-diindolylmethane (DIM); LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l- benzopyran-4-one); PP242; PP30; Torinl ; Ku-0063794; WAY-600; WYE-687; WYE- 354; and mTOR and PI3K dual-specificity inhibitors such as GNE477, NVP-BEZ235, PI-103, XL765 and WJD008. Examples of rapamycin include, but are not limited to, rapamycin of ref R0395 (Sigma- Aldrich), rapamycin of ref SI 039 (Selleckchem), rapamycin of ref 1292 (Tocris), rapamycin of ref R-5000 (LC Laboratories), rapamycin of ref tlrl-rap (InvivoGen), rapamycin of ref abl20224 (Abeam), rapamycin of ref R0395 (Sigma-Aldrich). Examples of compounds of the same chemical class than rapamycin used clinically include, but are not limited to, Everolimus (code name RAD001), Temsirolimus (code name CCI-779, NSC 683864), Zotarolimus (code name ABT-578).
In one embodiment, the mTOR inhibitor, preferably rapamycin, is used at a concentration ranging from 0.1 nM to 50 nM. Within the scope of the invention, the expression "from 0.1 nM to 50 nM" includes, without limitation 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 26 nM, 27 nM, 28 nM, 29 nM, 30 nM, 31 nM, 32 nM, 33 nM, 34 nM, 35 nM, 36 nM, 37 nM, 38 nM, 39 nM, 40 nM, 41 nM, 42 nM, 43 nM, 44 nM, 45 nM, 46 nM, 47 nM, 48 nM, 49 nM.
In one embodiment, at least one cytokine selected from IL-2, IL-7, IL-15 and TSLP can be added in the culture.
In one embodiment, IL-2 is used at a concentration ranging from 10 IU/ml to 1000 IU/ml. Within the scope of the invention, the expression "from 10 IU/ml to 1000 IU/ml" includes, without limitation 15 IU/ml, 20 IU/ml, 25 IU/ml, 30 IU/ml, 35 IU/ml, 40 IU/ml, 45 IU/ml, 50 IU/ml, 55 IU/ml, 60 IU/ml, 65 IU/ml, 70 IU/ml, 75 IU/ml, 80 IU/ml, 85 IU/ml, 90 IU/ml, 95 IU/ml, 100 IU/ml, 150 IU/ml, 200 IU/ml, 250 IU/ml, 300 IU/ml, 350 IU/ml, 400 IU/ml, 450 IU/ml, 500 IU/ml, 550 IU/ml, 600 IU/ml, 650 IU/ml, 700 IU/ml, 750 IU/ml, 800 IU/ml, 850 IU/ml, 900 IU/ml, 950 IU/ml. In certain embodiments, IL-2 is used at a concentration ranging from 50 IU/ml to 250 IU/ml.
In one embodiment, IL-7 is used at a concentration ranging from 1 ng/ml to 100 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 100 ng/ml" includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml.
In one embodiment, IL-15 is used at a concentration ranging from 1 ng/ml to 50 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 50 ng/ml" includes, without limitation 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml,
9 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml. In certain embodiments, IL-15 is used at a concentration ranging from
10 ng/ml to 30 ng/ml. In one embodiment, TSLP is used at a concentration from ranging 1 ng/ml to 100 ng/ml. Within the scope of the invention, the expression "from 1 ng/ml to 100 ng/ml" includes, without limitation 1 ng/ml, 5 ng/ml, 10 ng/ml, 15 ng/ml, 20 ng/ml, 25 ng/ml, 30 ng/ml, 35 ng/ml, 40 ng/ml, 45 ng/ml, 50 ng/ml, 55 ng/ml, 60 ng/ml, 65 ng/ml, 70 ng/ml, 75 ng/ml, 80 ng/ml, 85 ng/ml, 90 ng/ml, 95 ng/ml, 100 ng/ml. In one embodiment neutralizing antibodies can be added to the culture to prevent the generation of other populations of regulatory T cells.
Examples of neutralizing antibodies include, but are not limited to, anti-IFNy, anti-IL-4, and/or anti-IL12 antibodies.
Examples of anti-IFNy antibodies include, but are not limited to, Affymetrix eBioscience (Ref 14-7318), R&D systems (Ref MAB285), Novus Biologicals (Ref AF- 485-NA).
Examples of anti-IL-4 antibodies include, but are not limited to, R&D Systems (Ref MAB304, MAB204, or MAB204), Affymetrix eBioscience (Ref 14-7048), GeneTex (Ref GTX10755). Examples of anti-IL-12 antibodies include, but are not limited to, Affymetrix eBioscience (Ref 16-7129 or 16-8126), Biolegend (Ref 508803), R&D systems (Ref MAB219, AF-219, or AB-219).
In one embodiment, the culture medium used in the culture of the invention comprises (i) one or more pH buffering system(s); (ii) inorganic salt(s); (iii) trace element(s); (iv) free amino acid(s); (v) vitamin(s); (vi) hormone(s); (vii) carbon/energy source(s).
Examples of inorganic salts include, but are not limited to, calcium bromide, calcium chloride, calcium phosphate, calcium nitrate, calcium nitrite, calcium sulphate, magnesium bromide, magnesium chloride, magnesium sulphate, potassium bicarbonate, potassium bromide, potassium chloride, potassium dihydrogen phosphate, potassium disulphate, di- potassium hydrogen phosphate, potassium nitrate, potassium nitrite, potassium sulphite, potassium sulphate, sodium bicarbonate, sodium bromide, sodium chloride, sodium disulphate, sodium hydrogen carbonate, sodium dihydrogen phosphate, di-sodium hydrogen phosphate, sodium sulphate and a mix thereof.
Examples of trace elements include, but are not limited to, cobalt (Co), copper (Cu), iron (Fe), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), selenium (Se), zinc (Zn) and the salts thereof. Examples of free amino acids include, but are not limited to, L-alanine, L-arginine, L- asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, taurine, L-threonine, L-tryptophan, L-tyrosine, L-valine and a mix thereof. Examples of vitamins include, but are not limited to, biotin (vitamin H); D-calcium- pantothenate; choline chloride; folic acid (vitamin B9); myo-inositol; nicotinamide; pyridoxal (vitamin B6); riboflavin (vitamin B2); thiamine (vitamin Bl); cobalamin (vitamin B 12); acid ascorbic; a-tocopherol (vitamin E) and a mix thereof.
Examples of carbon/energy sources include, but are not limited to, D-glucose; pyruvate; lactate; ATP; creatine; creatine phosphate; and a mix thereof.
In one embodiment, the culture medium is a commercially available cell culture medium, in particular selected in a group comprising the IMDM (Iscove's Modified Dulbecco's Medium) from GIBCO® or the RPMI 1640 medium from GIBCO®.
In another embodiment, the culture medium is a serum-free culture medium such as the AIM-V medium from GIBCO®, the X-VIVO 10, 15 and 20 media from LONZA.
In another embodiment, the culture medium can be further supplemented with additional compound(s), in particular selected in a group comprising foetal bovine serum, pooled human AB serum, cytokines and growth factors; antibiotic(s), in particular selected in a group comprising penicillin, streptomycin and a mix thereof.
In one embodiment, the culture medium is IMDM.
In some particular embodiments, the culture medium comprises IMDM cell culture medium; from 1% (w/w) to 5% (w/w) of foetal bovine serum; from 10 IU/ml to 200 IU/ml of penicillin; from 10 IU/ml to 200 IU/ml of streptomycin; from 0.1 mM to 10 mM of a mixture of non-essential amino acids, in particular amino acids selected in a group comprising alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine; from 0.5 mM to 10 mM of glutamine from 10 mM to 25 mM of HEPES pH 7.6-7.8.
In one embodiment, the culture for generating the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days. Within the scope of the invention, the expression "at least 5 days" includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days.
In one embodiment, a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium. Within the scope of the invention the term "portion" is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium. In certain embodiments, 40%) (v/v) to 60%) (v/v) of the volume of the culture medium of step a) is discarded. In certain embodiments, the volume that is discarded is replaced with an identical volume of fresh culture medium. Within the scope of the invention, the expression "fresh culture medium" refers to a culture medium that has not been in contact with any CD3+ T cells.
In one embodiment, the medium is a nTreg polarizing medium. The inventors define a "nTreg polarizing medium" as a medium such as RPMI medium comprising at least one cAMP activator as described hereabove, at least one TGF pathway activator as described here above and at least one mTor inhibitor as described hereabove. In a preferred embodiment, the "nTreg polarizing medium" refers to a RPMI medium comprising TGF , rapamycin and PGE2. In another embodiment, the medium is an inflammatory medium. The inventors define an "inflammatory medium" as a medium such as IMDM comprising inflammatory cytokines such as for example IL-Ιβ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml).
In one embodiment, the method for generating ex vivo MHCII restricted CD4+ Foxp3+ regulatory T cells, comprises: culturing CD3+CD4+CD25" T cells in the presence of autologous ACD3 feeder cells and coated anti-CD3 antibody and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, - thereby obtaining a population of MHCII restricted CD4+ Foxp3+ regulatory
T cells ex vivo generated.
In one embodiment, the method for generating ex vivo MHCII restricted CD4+ Foxp3+ regulatory T cells, comprises: culturing CD3+CD4+CD25" T cells in the presence of tolerogenic DC that have been pulsed with at least one self-peptide antigen during about 24h and in the presence of ACD3 feeder cells and in the presence of the following agents: i) PGE2, ii) TGFp, iii) rapamycin and optionally iv) at least one cytokine selected in the group of IL-2 and IL-15, for at least 5 days, thereby obtaining a population of MHCII restricted CD4+ Foxp3+ regulatory T cells.
The present invention also relates to an ex vivo method of generation and expansion of MHCII restricted CD4+ Foxp3+ regulatory T cells, comprising: generating the MHCII restricted CD4+ Foxp3+ regulatory T cells as described here above, expanding the MHCII restricted CD4+ Foxp3+ regulatory T cells generated by contacting them in the presence of an TCRaP cell activator (preferably either autologous ACD3 feeder cells and coated anti-CD3 antibody or tolerogenic DC that have been pulsed with at least one self-peptide antigen during about 24h and in the presence of ACD3 feeder cells) and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator (preferably PGE2), ii) a TGF (Transforming growth factor beta) pathway activator (preferably TGFP), iii) a mTOR inhibitor (preferably rapamycin), and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP (preferably IL-2 and/or IL- 15), for at least 5 days, thereby obtaining an expanded population of MHCII restricted CD4+ Foxp3+ regulatory T cells.
In one embodiment, the MHCII restricted CD4+ Foxp3+ regulatory T cell population generated ex vivo is isolated by flow cytometry based on the following phenotype: CD3+TCRap+CD45RO+Foxp3+.
In one embodiment, the isolated MHCII restricted CD4+ Foxp3+ regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of a polyclonal αβ T cell activator. Examples of polyclonal αβ T cell activator are listed hereinabove. Alternatively, other examples of polyclonal αβ T cell activators that may be used during expansion include, but are not limited to, mitogen such as PMA/ionomycin, super-antigen, anti-CD3 antibody... Preferably, the anti-CD3 monoclonal antibody is coated. In one embodiment, the polyclonal T cell activator can be used in the presence of feeder cells as described here above.
In another embodiment, the isolated MHCII restricted CD4+ Foxp3+ regulatory T cell population thus obtained is then expanded ex vivo by culturing these cells in the presence of antigen-specific TCRaP cell activator as described here above. In one embodiment, the antigen-specific TCRaP cell activator can be used in the presence of feeder cells as described here above.
In one embodiment, the culture for expanding the ex vivo generated MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention is performed during at least 5 days, at least 6 days, at least 7 days, at least 8 days. Within the scope of the invention, the expression "at least 5 days" includes, without limitation 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days or more. In one embodiment, a portion of the culture medium is discarded once, twice, three times, four times or five times during the time course of the generation culture and replaced with the same volume of fresh culture medium. Within the scope of the invention the term "portion" is intended to mean at least 20% (v/v), at least 25% (v/v), at least 30% (v/v), at least 35% (v/v), at least 40% (v/v), at least 45% (v/v), at least 50% (v/v), at least 55% (v/v), at least 60% (v/v), at least 65% (v/v), at least 70% (v/v), at least 75% (v/v) of the volume of the culture medium. In certain embodiments, 40%) (v/v) to 60%) (v/v) of the volume of the culture medium of the first step is discarded. In certain embodiments, the volume that is discarded is replaced with an identical volume of fresh culture medium. Within the scope of the invention, the expression "fresh culture medium" refers to a culture medium that has not been in contact with any CD3+ T cells.
In one embodiment, MHCII restricted CD4+ Foxp3+ regulatory T cells are generated ex vivo by culturing CD3+TCR ap+CD45RA+, preferably CD3+TCR a +CD45RA+CD25", T cells obtained from PBMCs by negative selection (5.103 cells/ml) in the presence of autologous ACD3 feeder cells (125 105 cells/ml) and coated anti-CD3 antibody (2 μg/ml) in the presence of PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture. Every 3 days, half of the medium volume is discarded and replaced by fresh medium comprising PGE2 (50 nM), TGF (5 ng/ml), Rapamycin (1 nM), IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Once cells begin to expand, they can be split every 2 or 3 days and cultured in the presence of ACD3 feeder cells and coated anti-CD3 antibody every 9 days in a medium comprising PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin ( 10 nM) and IL-2 (100 Ul/ml) .
In another embodiment, MHCII restricted CD4+ Foxp3+ regulatory T cells are generated ex vivo by culturing CD3+TCR a +CD45RA+ T cells, preferably CD3+TCR a +CD45RA+CD25", obtained from PBMCs by negative selection (5.103 cells/ml) in the presence of tolerogenic DCs, that have been pulsed with at least one self-peptide antigen during about 24h, and in the presence of ACD3 feeder cells (125 105 cells/ml), PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml) in IMDM-5. On day 1, IL-2 (100 Ul/ml), IL-15 (10 ng/ml) and TGF (5 ng/ml), are added to the culture. Every 3 days, half of the medium volume is discarded and replaced by fresh medium comprising PGE2 (50 nM), TGF (5 ng/ml), Rapamycin (1 nM), IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Once cells begin to expand, they can be split every 2 or 3 days and restimulated every 9 days with tolerogenic DCs in the presence of ACD3 feeder cells and PGE2 (1 μΜ), TGF (5 ng/ml), Rapamycin (10 nM) and IL-2 (100 Ul/ml).
In this embodiment, tolerogenic DCs were obtained by culturing CD14+ monocytes isolated from PBMCs in the presence of AIMV supplemented with GMCSF (100 ng/ml) and IL-4 (10 ng/ml). At day 3 and 6, the medium is discarded and replaced by fresh medium comprising GM-CSF and IL-4. On day 6, the tolerogenic DCs are pulsed for 24 hours in the presence of self-peptide antigen.
Populations Invariant Foxp3+ regulatory T cells
The present invention relates to an isolated population of invariant Foxp3+ regulatory T cells having the following phenotype: CD3+ Va24+ Foxp3+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD 127" CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD45RO+ CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD 127" CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO + CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO + CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO + CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+. In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD 161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD127 CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD45RO+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CD45RO+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD45RO+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD 161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD127" CD161 CD56".
In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD 127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD 127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD45RO+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD 127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CTLA4+ CD45RO+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CTLA4+ CD45RO+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CD45RO+ CD 127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD127" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD 127" CD161". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD127" CD161" CD56". In another embodiment, the invariant Foxp3+ regulatory T cells are CD3+ Va24+ Foxp3+ CD4+ CD25+ CTLA4+ CD45RO+ CD 127" CD161" CD 127". In another embodiment, the invariant Foxp3+ regulatory T cells are Va24+ Jal8+.
In one embodiment, the invariant Foxp3+ regulatory T cells CD3+ Va24+ Foxp3+ CD4+ express the TCR νβΐ chain. In one embodiment, the invariant Foxp3+ regulatory T cells CD3+ Va24+ Foxp3+ CD4+ express the TCR νβ2 chain. In one embodiment, the invariant Foxp3+ regulatory T cells CD3+ Va24+ Foxp3+ CD4+ express the TCR Υβ4 chain. In one embodiment, the invariant Foxp3+ regulatory T cells CD3+ Va24+ Foxp3+ CD4+ express the TCR νβ8 chain. In one embodiment, the invariant Foxp3+ regulatory T cells CD3+ Va24+ Foxp3+ CD4+ express the TCR νβ5.1 chain.
In one embodiment, the invariant Foxp3+ regulatory T cells of the invention are human cells. yd Foxp3+ regulatory T cells
The present invention also relates to γδ Foxp3+ regulatory T cells having the following phenotype: CD3+TCRy5+Foxp3+. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the V52 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the Vy9 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the Vy9V02 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention do not express the νδ2 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention do not express the Vy9 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention do not express the Vy9V02 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the νδ3 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the νδ4 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the νδ5 isotype. In one embodiment, the γδ Foxp3+ regulatory T cells of the invention express the νδ6 isotype. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy0+ CD25+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy0+ IL-1R1". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy0+ IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy0+ IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy0+ IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy0+ CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-1R1". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-1R1" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-6R" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-23R" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-33R" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-1R1" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-6R" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-23R" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL- 33R" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-lRl" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-6R" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-23R" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-33R" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-lR IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-lRl" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-lRl" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-6R" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL- 6R~ IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD45RO+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-lRl" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-6R" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-23R" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-33R" CTLA4+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-lRl" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-6R" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-23R" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-33R" CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-lRl" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-6R" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-23R" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-33R" CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-1R1" IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-1R1" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-1R1" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-6R" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-6R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CD45RO+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL- 1R1". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-lRl" IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-1R1 IL-23R IL-33R- . In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-6R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-6R" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CTLA4+ CD45RO+ CD127+ IL-lRl" IL-6R" IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+. In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-lRl". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-6R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL- 1R1" IL-23R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-6R" IL-33R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1 IL-6R IL-23R". In one embodiment, said population of γδ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ TCRy5+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1 IL-6R IL-23R IL-33R-.
MHCII restricted CD4+ Foxp3+ regulatory T cells
The present invention also relates to a population of MHCII restricted CD4+ Foxp3+ regulatory T cells having the following phenotype: CD3+TCRa,p+Foxp3+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-1R1". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-6R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CTLA4+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-lRl" CTLA4+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-6R" CTLA4+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-23R" CTLA4+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-33R" CTLA4+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-lRl" CD25+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-6R" CD25+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-23R" CD25+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-33R" CD25+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-lRl" CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-6R" CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-23R" CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-33R" CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-lRl" IL-6R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-lRl" IL-23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-lRl" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-6R" IL- 23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-6R" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ IL-23R" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CTLA4+ CD45RO+. In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-lRl" IL-6R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-lRl" IL-23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-lRl" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R" IL- 23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-23R" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-6R" IL-23R" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1 IL-23R IL-33R\ In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-6R" IL-33R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1" IL-6R" IL-23R". In one embodiment, said population of MHCII restricted CD4+ Foxp3+ regulatory T cells has the following phenotype: CD4+ Foxp3+ CD25+ CTLA4+ CD45RO+ CD127+ IL-1R1 IL-6R-IL-23R IL-33R\
FoxP3 In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) at least equivalent to the Foxp3 MFI measured in naive regulatory T cells. As used herein, "na'ive regulatory T cells" refers to T cells having for phenotype Foxp3+CD45RA+CD4+CD25+CD127\
In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) of at least 2000. In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) of at least 2 or 3 fold the Foxp3 MFI measured in na'ive regulatory T cells. In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention express Foxp3 with a median fluorescence intensity (MFI) of at least 2000, 3000, 4000, 5000, 10000, 20000, 30000, 40000, 50000, 60000, 70000.
In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention comprise at least 65% of the CD3+ Va24+ cells expressing Foxp3. The expression "at least 65%" includes, without limitation 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 752%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 82%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and 100%.
As used herein, the term "expression" may refer alternatively to the transcription of a molecule (i.e. expression of the mRNA) or to the translation (i.e. expression of the protein) of a molecule. In one embodiment, detecting the expression may correspond to an intracellular detection. In another embodiment, detecting the expression may correspond to a surface detection, i.e. to the detection of molecules expressed at the cell surface. In another embodiment, detecting the expression may correspond to an extracellular detection, i.e. to the detection of secretion. In another embodiment, detecting the expression may correspond to intracellular, surface and/or extracellular detections. Methods for determining the expression level are well-known from the skilled artisan, and include, without limitation, determining the transcriptome (in an embodiment wherein expression relates to transcription of a molecule) or proteome (in an embodiment wherein expression relates to translation of a cytotoxic molecule) of cells.
In one embodiment of the invention, the expression of the molecules is assessed at the mRNA level. Methods for assessing the transcription level of a molecule are well known in the prior art. Examples of such methods include, but are not limited to, RT- PCR, RT-qPCR, Northern Blot, hybridization techniques such as, for example, use of microarrays, and combination thereof including but not limited to, hybridization of amplicons obtained by RT-PCR, sequencing such as, for example, next-generation DNA sequencing (NGS) or RNA-seq (also known as "Whole Transcriptome Shotgun Sequencing") and the like. In another embodiment of the invention, the expression of the molecules is assessed at the protein level. Methods for determining a protein level in a sample are well-known in the art. Examples of such methods include, but are not limited to, immunohistochemistry, Multiplex methods (Luminex), western blot, enzyme-linked immunosorbent assay (ELISA), sandwich ELISA, fluorescent-linked immunosorbent assay (FLISA), enzyme immunoassay (EIA), radioimmunoassay (RIA), flow cytometry (FACS) and the like. In another embodiment, determining the expression level of at least one molecule corresponds to detecting and/or quantifying binding of a ligand to a molecule. In one embodiment, said ligand is an antibody specific of said molecule, and the method of the invention comprises detecting and/or quantifying a complex formed between said antibody and said molecule. The complex can be detected if the ligand has been for example, but not limited to, covalently coupled with a detectable molecule such as an antibody constant fragment (Fc) or a fluorescent compound (e.g. Cyanine dye, Alexa dye, Quantum dye, etc). The complex can also be detected if the ligand has been tagged with different means well known to the person skilled in the art. For example, but without limitation, a tag used with the invention can be a tag selected from the group comprising or consisting of Hemaglutinin Tag, Poly Arginine Tag, Poly Histidine Tag, Myc Tag, Strep Tag, S-Tag, HAT Tag, 3x Flag Tag, Calmodulin-binding peptide Tag, SBP Tag, Chitin binding domain Tag, GST Tag, Maltose-Binding protein Tag, Fluorescent Protein Tag, T7 Tag, V5 Tag and Xpress Tag. The use of the ligand therefore allows on the one hand the identification and detection of the molecule depending on the ligand used, and on the other hand the quantification of the complex formed.
In one embodiment, determining the expression level of molecules is conducted by flow cytometry, immunofluorescence or image analysis, for example high content analysis. Preferably, the determination of the expression level of molecules is conducted by flow cytometry. In one embodiment, before conducting flow cytometry analysis, cells are fixed and permeabilized, thereby allowing detecting intracellular proteins.
In one embodiment, determining the expression level of a molecule in a cell population comprises determining the percentage of cells of the cell population expressing the molecule (i.e. cells "+" for the molecule). Preferably, said percentage of cells expressing the molecule is measured by FACS.
The terms "expressing (or +)" and "not expressing (or -)" are well known in the art and refer to the expression level of the cell marker of interest, in that the expression level of the cell marker corresponding to "+" is high or intermediate, also referred as "+/-". The cell marker corresponding to "-" is a null expression level of the cell marker or also refers to less than 10 % of a cell population expressing the said cell marker.
The expression level of the cell marker of interest is determined by comparing the Median Fluorescence Intensity (MFI) of the cells from the cell population stained with fiuorescently labeled antibody specific for this marker to the fluorescence intensity (FI) of the cells from the same cell population stained with fiuorescently labeled antibody with an irrelevant specificity but with the same isotype, the same fluorescent probe and originated from the same specie (referred as Isotype control). The cells from the population stained with fiuorescently labeled antibody specific for this marker and that show equivalent MFI or a lower MFI than the cells stained with the isotype controls are not expressing this marker and then are designated (-) or negative. The cells from the population stained with fiuorescently labeled antibody specific for this marker and that show a MFI value superior to the cells stained with the isotype controls are expressing this marker and then are designated (+) or positive.
In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention do not present a regulatory T cells specific demethylated region (TSDR) of the gene Foxp3. In another embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention present a regulatory T cells specific demethylated region (TSDR) of the gene Foxp3. In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention present a percentage of demethylation of the TSDR of the gene FOXP3 superior to at least 30%, 40%>, 50%>. A protocol for measuring promoter demethylation percentage is shown in the Material and Method part of the Examples.
In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention present a percentage of enrichment of acetylated histone in Foxp3 promoter region superior to at least 10%>, 20%>, 30%>, 40%> or 50%>. A protocol for measuring enrichment of acetylated histones in percentage is shown in the Material and Method part of the Examples.
An example of phenotypic characteristics of the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention is shown in Figure 1. Suppressive activity
In one embodiment, the invariant Foxp3+ regulatory T cells of the invention, the γδ Foxp3+ regulatory T cells of the invention and the MHCII restricted CD4+ Foxp3+ regulatory T cells of the invention are capable of suppressive activity similar to the suppressive activity of na'ive CD4+ CD25+ CD45RA+ CD 127" regulatory T cells. Determination of the suppressive activity of a cell population is well known in the art and can be performed by conventional assays such as the standard polyclonal cell-cell contact Treg suppression assay or the autologous MLR suppression assay as described in the Examples.
Population obtained by the method The present invention also relates to invariant Foxp3+ regulatory T cells, γδ Foxp3+ regulatory T cells and MHCII restricted CD4+ Foxp3+ regulatory T cells obtainable or obtained by the ex vivo generation method as described here above. The present invention also relates to invariant Foxp3+ regulatory T cells, γδ Foxp3+ regulatory T cells and MHCII restricted CD4+ Foxp3+ regulatory T cells obtainable or obtained by the ex vivo generation and expansion method as described here above.
In one embodiment, the populations of invariant Foxp3+ regulatory T cells, γδ Foxp3+ regulatory T cells and MHCII restricted CD4+ Foxp3+ regulatory T cells obtained by the generation and expansion method of the invention comprises at least 106, 107, 108, 109, 1010 cells.
Population stable in inflammatory conditions
In one embodiment, the populations of invariant Foxp3+ regulatory T cells, γδ Foxp3+ regulatory T cells and MHCII restricted CD4+ Foxp3+ regulatory T cells obtained by the generation and expansion method of the invention has the property to remain stable when placed in inflammatory conditions.
As used herein, "stable" refers to no secretion or a low secretion of IL-17, i.e. inferior to 200 ng/ml, 100 ng/ml, 50 ng/ml and still capable of suppressive capacity, i.e. inhibiting proliferation of conventional T cells as shown in the Examples.
As used herein, "inflammatory condition" refers to a medium enriched in aromatic acid, preferably in tryptophan, such as for example IMDM, comprising inflammatory cytokines such as for example IL-Ιβ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml), IL- 23 (30 ng/ml), IL-2 (100 Ul/ml). A method for determining if a population of regulatory T cells remains stable in inflammatory condition comprises culturing the regulatory T cells in the inflammatory condition medium as described here above in the presence of anti-CD3 (4 μg/ml), preferably coated, and anti-CD28 (4 μg/ml), preferably in a soluble form. After 36h to 72h of culture, the presence of IL-17 in the culture supernatant is measured. The recognition of IL-17 in the culture supernatant may be carried out by conventional methods known in the art such as, for example, a sandwich ELISA anti-IL-17. Briefly, after coated the plate with a capture anti-IL-17 antibody, the culture supernatant is added to each well with a dilution series. After incubation, a detection anti-IL-17 antibody is added to each well. The ELISA is developed by any colorimetric means known in the art such as, for example, using detection antibody labelled with biotin, a poly-streptavidin HRP amplification system and an o- phenylenediamine dihydro chloride substrate solution. An IL-17 level inferior to 200 ng/ml, 100 ng/ml, 50 ng/ml corresponds to no secretion or low secretion of IL-17.
Vaccination Without wishing to be bound to a theory, the inventors state that the stroma of malignant tumor cells comprises TILs (Tumor-infiltrating lymphocytes) that are highly enriched in regulatory T cells and that exert an immune suppressive activity, in particular on NK cells, which likely accounts on the local cancer immune escape. The at least one inactivated regulatory T cells population may represent an antigenic target to induce an immune response directed against the at least one regulatory T cells population present in the TILs, thereby preventing their immune suppressive activity and allowing the cytotoxic activity of effector cells such as NK cells against the tumor cells. The inventors thus suggest using a vaccine composition comprising as active principle at least one inactivated regulatory T cells population among peripheral regulatory T cells population, γδ regulatory T cells population and invariant regulatory T cells population.
Immunogenic product
One object of the invention is an immunogenic product comprising, consisting essentially of or consisting of at least one inactivated Foxp3+ regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population.
In one embodiment, the immunogenic product comprises, consists essentially of or consists of at least one inactivated ex vivo generated Foxp3+ regulatory T cells population among an ex vivo generated MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated γδ Foxp3+ regulatory T cells population and an ex vivo generated invariant Foxp3+ regulatory T cells population. As used herein, the term "consisting essentially of, with reference to an immunogenic product, pharmaceutical composition, vaccine or medicament, means that the at least one ex vivo generated Foxp3+ regulatory T cells population or antibody of the invention is the only one therapeutic agent or agent with a biologic activity within said immunogenic product, pharmaceutical composition, vaccine or medicament.
In one embodiment, the immunogenic product comprises, consists essentially of or consists of at least one inactivated ex vivo generated and optionally expanded regulatory T cells population among an ex vivo generated and optionally expanded MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated and optionally expanded γδ Foxp3+ regulatory T cells population and an ex vivo generated and optionally expanded invariant Foxp3+ regulatory T cells population.
Pharmaceutical composition
Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of the immunogenic product as described here above and at least one pharmaceutically acceptable excipient.
Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one inactivated Foxp3+ regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient.
Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one inactivated ex vivo generated Foxp3+ regulatory T cells population among an ex vivo generated MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated γδ Foxp3+ regulatory T cells population and an ex vivo generated invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient.
As used herein, the term "excipient" refers to any and all conventional solvents, dispersion media, fillers, solid carriers, aqueous solutions, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. For human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by regulatory offices, such as, for example, FDA Office or EMA. By "pharmaceutically acceptable" is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the subject to which it is administered. Examples of pharmaceutically acceptable excipient include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like or combinations thereof. Vaccine composition
Another object of the invention is a vaccine composition comprising, consisting essentially of or consisting of the immunogenic product as described here above.
Another object of the invention is a vaccine composition comprising, consisting essentially of or consisting of at least one inactivated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population.
Another object of the invention is a vaccine composition comprising, consisting essentially of or consisting of at least one inactivated ex vivo generated regulatory T cells population among an ex vivo generated MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated γδ Foxp3+ regulatory T cells population and an ex vivo generated invariant Foxp3+ regulatory T cells population.
As used herein, "inactivated" T cells refers to T cells that are viable but has reduced or no effector function, i.e. have lost any pathogenic potential. Examples of cell surface markers of inactivated T cells include, but are not limited to, 7-Aminoactinomycin D (7- AAD), calreticulin and heat shock protein 90 (HSP-90). Therefore, inactivated T cells express 7-AAD and/or calreticulin and/or HSP-90. The inactivated regulatory T cells of the invention have lost their suppressive activity but are still immunogenic. An example of T cell effector function assay is, but not limited to, T-cell proliferation assay. T-cell proliferation may be assessed on fixed T cells versus non-fixed T cells. Briefly, the T- cell proliferation assay aims at determining the percentage of living proliferating cells in fixed versus non-fixed T cells by flow cytometry. After staining the T cells with CFSE, anti-CD3 antibody and 7-AAD, the living proliferating cells are defined as the CFSE low fraction in gated CD3+ 7-AAD" cells.
In one embodiment, the regulatory T cells are inactivated by any method well-known in the art. Examples of method for inactivating cells include, but are not limited to, irradiation, preferably with about 2500 to 3000 rads and/or chemical inactivation such as exposure to cisplatin, carboplatin, oxaliplatin, mitomycine C or antracycline. In one embodiment, the vaccine composition of the invention further comprises at least one adjuvant. Examples of adjuvant that can be used in the vaccine composition include, but are not limited to, ISA51; emulsions such as CFA, MF59, montanide, AS03 and AF03; mineral salts such as alum, calcium phosphate, iron salt, zirconium salt, and AS04; TLR ligands such as TLR2 ligands (such as outer-surface protein A or OspA), TLR3 ligands (such as poly I:C), TLR4 ligands (such as MPL and GLA), TLR5 ligands, TLR7/8 ligands (such as imiquimod), TLR9 ligands (such as CpG ODN); polysacharrides such as chitin, chitosan, a-glucans, β-glucans, fructans, mannans, dextrans, lentinans, inulin-based adjuvants (such as gamma inulin); TLR9 and STING ligands such as K3 CpG and cGAMP. As used herein, "adjuvant" refers to an agent that potentiates the immune responses to an antigen and/or modulates it towards the desired immune responses.
In one embodiment, the inactivated Foxp3+ regulatory T cells present in the immunogenic product, pharmaceutical composition or vaccine composition of the invention are human Foxp3+ regulatory T cells. In one embodiment, the inactivated Foxp3+ regulatory T cells present in the immunogenic product, pharmaceutical composition or vaccine composition of the invention are autologous Foxp3+ regulatory T cells. In one embodiment, the inactivated regulatory T cells present in the immunogenic product, pharmaceutical composition or vaccine composition of the invention are allogenic Foxp3+ regulatory T cells.
In another embodiment, the immunogenic product, pharmaceutical composition or vaccine composition of the invention may be personalized for a patient. As used herein, a "personalized" immunogenic product or vaccine composition refers to the use of Foxp3+ regulatory T cells generated and expanded ex vivo with at least one patient specific epitope. In this embodiment, the Foxp3+ regulatory T cells to be used as immunogenic product or in the vaccine composition are generated and expanded ex vivo in the presence of apoptotic bodies of cancer cells obtained from the patient, thereby providing at least one patient specific epitope.
In one embodiment, the immunogenic product, pharmaceutical composition or vaccine composition of the invention comprises, consists essentially of or consists of as active principle at least one inactivated Foxp3+ regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population.
In one embodiment, the immunogenic product, pharmaceutical composition or vaccine composition of the invention comprises, consists essentially of or consists of at least 104, 105, 106, 107, 108, 109, 1010 inactivated Foxp3+ regulatory T cells as active principle.
In one embodiment, the immunogenic product, pharmaceutical composition or vaccine composition of the invention comprise, consist essentially of or consist of about 104, 5xl04, 105, 5xl05, 106, 5xl06, 107, 5xl07, 108, 5xl08, 109, 5xl09, 1010, inactivated Foxp3+ regulatory T cells as active principle. In one embodiment, the Foxp3+ regulatory T cells, the inactivated Foxp3+ regulatory T cells, the immunogenic product, the pharmaceutical composition or the vaccine composition of the invention are/is frozen. In one embodiment, the immunogenic product, pharmaceutical composition or vaccine composition of the invention may be administrated to the subject by subcutaneous, intramuscular, intraperitoneal or intravenous injection, or directly into the tumor.
In one embodiment, the immunogenic product, pharmaceutical composition or vaccine composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times in a year. Example of regime of administration includes, but is not limited to, administration of the immunogenic product or vaccine composition at day 0, 4 weeks after day 0, 8 weeks after day 0, 12 weeks after day 0 and 24 weeks after day O. Treatment of cancer
Another object of the invention is a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of at least one inactivated regulatory T cells population or of the immunogenic product, pharmaceutical composition or vaccine composition of the invention as described here above.
Another object of the invention is a method for eliciting an immune response against at least one Foxp3+ regulatory T cells population present in the TILs of a subject affected with a cancer, comprising administering to the subject a therapeutically effective amount of at least one inactivated Foxp3+ regulatory T cells population or of the immunogenic product, pharmaceutical composition or vaccine composition of the invention as described here above.
Another object of the invention is a method for inducing an immunogenic apoptosis of at least one Foxp3+ regulatory T cells population present in the TILs of a subject affected with a cancer, comprising administering to the subject a therapeutically effective amount of at least one inactivated Foxp3+ regulatory T cells population or of the immunogenic product, pharmaceutical composition or vaccine composition of the invention as described here above. Examples of cancer that can be treated with the immunogenic product, pharmaceutical composition or vaccine composition of the invention include, but are not limited to, adrenocortical carcinoma, anal cancer, bladder cancer, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal, pineal tumors, hypothalamic glioma, breast cancer, carcinoid tumor, carcinoma, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, extrahepatic bile duct cancer, ewings family of tumors (pnet), extracranial germ cell tumor, eye cancer, intraocular melanoma, gallbladder cancer, gastric cancer, germ cell tumor, extragonadal, gestational trophoblastic tumor, head and neck cancer, hypopharyngeal cancer, islet cell carcinoma, laryngeal cancer, leukemia, acute lymphoblastic, leukemia, oral cavity cancer, liver cancer, lung cancer, small cell lymphoma, AIDS-related, lymphoma, central nervous system (primary) lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, non-Hodgkin's disease, malignant mesothelioma, melanoma, merkel cell carcinoma, metastatic squamous carcinoma, multiple myeloma, plasma cell neoplasms, mycosis fungoides, myelodysplasia syndrome, myeloproliferative disorders, nasopharyngeal cancer, neuroblastoma, oropharyngeal cancer, osteosarcoma, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, exocrine, pancreatic cancer, paranasal sinus and nasal cavity cancer, parathyroid cancer, pheochromocytoma cancer, pituitary cancer, plasma cell neoplasm, rhabdomyosarcoma, rectal cancer, renal cell cancer, salivary gland cancer, Sezary syndrome, Kaposi's sarcoma, small intestine cancer, soft tissue sarcoma, thymoma, malignant thyroid cancer, urethral cancer, uterine cancer, sarcoma, unusual cancer of childhood, vaginal cancer, vulvar cancer or Wilms' tumor, benign conditions associated with chemotherapy treatments, such as, lupus, rheumatoid arthritis and skin diseases. In one embodiment, the cancer that can be treated with the immunogenic product, pharmaceutical composition or vaccine composition of the invention include, but is not limited to, breast cancer, prostate cancer, ovarian cancer and glioblastoma.
Another object of the invention is a method for preparing the immunogenic product of the invention, comprising: identifying from a tumor sample or from TILs obtained from the subject the at least one regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population, that is overrepresented, ex vivo generating the at least one overrepresented regulatory T cells population, inactivating the at least one ex vivo generated regulatory T cell population.
In one embodiment, the MHCII restricted CD4+ Foxp3+ regulatory T cells population is considered overrepresented when superior to 5 to 10 % of total cells present in the sample. In one embodiment, there is a positive correlation between the percentage of Foxp3 expression in MHCII restricted CD4+ Foxp3+ regulatory T cells in the sample and a poor clinical outcome for breast cancer.
In one embodiment, the γδ Foxp3+ regulatory T cells population is considered overrepresented when superior to 5 to 10 % of total cells present in the sample. In one embodiment, there is a positive correlation between the percentage of Foxp3 expression in MHCII restricted CD4+ Foxp3+ regulatory T cells in the sample and a poor clinical outcome for breast cancer.
In one embodiment, the invariant Foxp3+ regulatory T cells population is considered overrepresented when superior to 0.01 to 1 % of total cells present in the sample.
Another object of the invention is a method for treating cancer in a subject in need thereof, comprising administrating to the subject the immunogenic product, pharmaceutical composition or vaccine composition of the invention.
Another object of the invention is a method for treating cancer in a subject in need thereof, comprising:
- preparing an immunogenic product as described here above, optionally preparing a pharmaceutical composition or a vaccine composition comprising the immunogenic product, optionally submitting the subject to plasmapheresis, administrating to the subject the immunogenic product, pharmaceutical composition or vaccine composition of the invention.
Immune suppressive functions
Without wishing to be bound by a theory, the inventors suggest that the Foxp3+ regulatory T cells of the invention, which are committed to exert immune suppressive function, may be capable of inhibiting autoreactive pathogenic immune effector cells including CD4+, CD8+, B cells or innate NK cells, which, in turn, are no longer able to exert their cytotoxic properties towards the self-cells.
One object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient. Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one ex vivo generated Foxp3+ regulatory T cells population among an ex vivo generated MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated γδ Foxp3+ regulatory T cells population and an ex vivo generated invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient.
Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of at least one ex vivo generated Foxp3+ regulatory T cells population among an ex vivo generated and expanded MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated and expanded γδ Foxp3+ regulatory T cells population and an ex vivo generated and expanded invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient.
Another object of the invention is a pharmaceutical composition comprising at least one ex vivo generated Foxp3+ regulatory T cells population among an ex vivo generated MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated γδ Foxp3+ regulatory T cells population and an ex vivo generated invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated regulatory T cells population remains stable when placed in inflammatory condition as described here above.
Another object of the invention is a pharmaceutical composition comprising at least one ex vivo generated Foxp3+ regulatory T cells population among an ex vivo generated and expanded MHCII restricted CD4+ Foxp3+ regulatory T cells population, an ex vivo generated and expanded γδ Foxp3+ regulatory T cells population and an ex vivo generated and expanded invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated and expanded regulatory T cells population remains stable when placed in inflammatory condition as described here above.
One object of the invention is the at least one ex vivo generated Foxp3+ regulatory T cells population or the pharmaceutical composition as described here above for use in adoptive therapy.
Another object of the invention is the at least one ex vivo generated Foxp3+ regulatory T cells population or the pharmaceutical composition as described here above for use in treating inflammatory or autoimmune diseases. One object of the invention is the at least one ex vivo generated and expanded Foxp3+ regulatory T cells population or the pharmaceutical composition as described here above for use in adoptive therapy.
Another object of the invention is the at least one ex vivo generated and expanded Foxp3+ regulatory T cells population or the pharmaceutical composition as described here above for use in treating inflammatory or autoimmune diseases.
Examples of inflammatory or autoimmune diseases include, but are not limited to, acute disseminated encephalomyelitis, acute necrotizing haemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing spondylitis, anti-GBM/Anti-TBM nephritis, antiphospho lipid syndrome, autoimmune angioedema, autoimmune aplastic anaemia, autoimmune dysautonomia, autoimmune haemo lytic anaemia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease, autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura, autoimmune thyroid disease, autoimmune urticaria, axonal and neuronal neuropathies, Balo disease, Behcet's disease, bullous pemphigoid, cardiomyopathy, Castleman disease, celiac disease, Chagas disease, chronic fatigue syndrome, chronic inflammatory demyelinating polyneuropathy, chronic recurrent multifocal osteomyelitis, Churg-Strauss syndrome, cicatricial pemphigoid/benign mucosal pemphigoid, Crohn's disease, Cogans' syndrome, cold agglutinin disease Congenital heart block, Coxsackie myocarditis, CREST disease, essential mixed cryoglobulinemia, demyelinating neuropathies, dermatitis herpetiformis, dermatomyositis, Devic's disease, discoid lupus, Dressler's syndrome, endometriosis, eosinophilic esophagitis, eosinophilic fasciitis, erythema nodosum, experimental allergic encephalomyelitis, Evans syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis, giant cell myocarditis, glomerulonephritis, Goodpasture's syndrome, granulomatosis with Polyangiitis (Wegener's syndrome), Graves' disease, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, haemolytic anaemia, Henoch-Schonlein purpura, herpes gestationis, hypogammaglobulinemia, idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura, IgA nephropathy, IgG4-related sclerosing disease, immunoregulatory lipoproteins, inclusion body myositis, interstitial cystitis, juvenile arthritis, juvenile diabetes (Type 1 diabetes), juvenile myositis, Kawasaki syndrome, Lambert-Eaton syndrome, leukocytoclastic vasculitis, lichen planus, lichen sclerosus, ligneous conjunctivitis, linear IgA disease, lupus, Lyme chronic disease, Meniere's disease, microscopic polyangiitis, mixed connective tissue disease, Mooren's ulcer, Mucha- Habermann disease, multiple sclerosis, myasthenia gravis, myositis, narcolepsy, neuromyelitis optica, neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic rheumatism, paediatric autoimmune neuropsychiatric disorders associated with Streptococcus, paraneoplastic cerebellar degeneration, paroxysmal nocturnal hemoglobinuria, Parry Romberg syndrome, Parsonnage-Turner syndrome, pars planitis (peripheral uveitis), pemphigus, peripheral neuropathy, perivenous encephalomyelitis, pernicious anaemia, POEMS syndrome, polyarteritis nodosa, type I, II, and III autoimmune polyglandular syndromes, polymyalgia rheumatic, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, progesterone dermatitis, primary biliary cirrhosis, primary sclerosing cholangitis, psoriasis, psoriatic arthritis, pyoderma gangrenosum, pure red cell aplasia, Raynauds phenomenon, reactive arthritis, reflex sympathetic dystrophy, Reiter's syndrome, Relapsing polychondritis, restless legs syndrome, retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis, sarcoidosis, Schmidt syndrome, scleritis, scleroderma, Sjogren's syndrome, sperm and testicular autoimmunity, Stiff person syndrome, subacute bacterial endocarditis, Susac's syndrome, sympathetic ophthalmia, systemic lupus erythematosus, Takayasu's arteritis, temporal arteritis/Giant cell arteritis, thrombocytopenic purpura, Tolosa-Hunt syndrome, transverse myelitis, type 1 diabetes, ulcerative colitis, undifferentiated connective tissue disease, uveitis, vasculitis, vesiculobullous dermatosis and vitiligo.
Examples of inflammatory or autoimmune diseases include, but are not limited to, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis.
Another object of the invention is the at least one ex vivo generated Foxp3+ regulatory T cells population or the pharmaceutical composition as described here above for use in preventing transplant rejection, graft versus host disease (GVHD).
Another object of the invention is the at least one ex vivo generated and expanded Foxp3+ regulatory T cells population or the pharmaceutical composition as described here above for use in preventing transplant rejection, graft versus host disease (GVHD).
In one embodiment, the pharmaceutical composition of the invention comprises, consists essentially of or consists of at least 104, 105, 106, 107, 108, 109, 1010 ex vivo generated Foxp3+ regulatory T cells as active principle.
In one embodiment, the pharmaceutical composition of the invention comprises, consists essentially of or consists of about 104, 5x104, 105, 5x105, 106, 5x106, 107, 5xl07, 108, 5xl08, 109, 5xl09, 1010 ex vivo generated Foxp3+ regulatory T cells as active principle. In one embodiment, the at least one ex vivo generated Foxp3+ regulatory T cells population or the pharmaceutical the invention are/is frozen.
In one embodiment, the at least one ex vivo generated Foxp3+ regulatory T cells population present in the pharmaceutical composition of the invention is generated from human T cells.
In one embodiment, the at least one ex vivo generated Foxp3+ regulatory T cells population present in the pharmaceutical composition of the invention is autologous.
In one embodiment, the at least one ex vivo generated Foxp3+ regulatory T cells population present in the pharmaceutical composition of the invention is allogenic.
In one embodiment, the pharmaceutical composition of the invention may be administrated to the subject by subcutaneous, intramuscular, intraperitoneal or intravenous injection.
In one embodiment, the pharmaceutical composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times per week.
In another embodiment, the pharmaceutical composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times per month.
In another embodiment, the pharmaceutical composition of the invention may be administrated to the subject at least once, twice, 3 times, 4 times, 5 times per 3 months.
Another object of the invention is a method for treating inflammatory or autoimmune diseases in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of at least one ex vivo generated Foxp3+ regulatory T cells or the pharmaceutical composition as described here above.
Antibodies
It has been shown in the art that T cell vaccination induces regulatory networks that specifically suppress the immunogenic T cells by activating T cells specific for a clono type-specific determinant (anti- idiotypic response). In addition, anti-ergotypic responses directed at activation markers (corresponding to the ergotope) may also partially account for the suppression of the regulatory T cell population targeted.
Another object of the invention is an antibody recognizing the TCR (T cell receptor) of the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention. In one embodiment, the antibody recognizing the TCR of the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention recognizes at least one of the CDR1, CDR2 and CDR3 (complementary determining region 1, 2 and 3) of the TCR.
In another embodiment, the antibody recognizing the TCR of the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention recognizes the CDR3 of the TCR.
Another object of the invention is a pharmaceutical composition comprising, consisting essentially of or consisting of said antibody and at least one pharmaceutically acceptable excipient. Another object of the invention is the use of said antibody for treating cancer in a subject in need thereof.
In one embodiment, the antibodies directed against the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention consist of antibodies produced following immunization of a mammal, including a human, with the immunogenic composition as described here above.
In another embodiment, the antibodies may also be obtained by cloning the relevant DNA material encoding them, starting for example from B cells obtained from the said mammal, including from the said human.
In another embodiment, the antibodies may also be obtained by sequencing the amino acid sequences of the antibodies collected from the said mammal, including from the said human, and then synthesize a DNA molecule encoding the antibody or a portion thereof comprising the CDR thereof, for producing relevant recombinant antibodies directed against the peripheral regulatory T cells of the invention.
Preparing antibodies directed against the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention by immunization with the immunogenic composition of the invention may be easily performed by a skilled in the art, using the common technical knowledge from the state in the art.
Alternatively, the antibodies directed against the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention may be obtained after immortalization of the human B lymphocytes producing them; their cDNA can also be cloned and used further for producing them or their derivatives through recombinant DAN technology.
The term "antibody" herein is used to refer to a molecule having a useful antigen binding specificity. Those skilled in the art will readily appreciate that this term may also cover polypeptides which are fragments of or derivatives of antibodies yet which can show the same or a closely similar functionality. Such antibody fragments or derivatives are intended to be encompassed by the term antibody as used herein. By "antibody" or "antibody molecule" for the purpose of passive immunotherapy, it is intended herein not only whole immunoglobulin molecules but also fragments thereof, such as Fab, F(ab')2, Fv and other fragments thereof that retain the capacity to bind and inactivate the peripheral regulatory T cells. Similarly, the term antibody includes genetically engineered derivatives of antibodies such as single chain Fv molecules (scFv) and domain antibodies (dAbs).
In some embodiments, an antibody directed against the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention consists of a polyclonal antibody.
In some embodiments, an antibody directed against the at least one ex vivo generated Foxp3+ regulatory T cells population of the invention consists of a monoclonal antibody.
The term "monoclonal antibody" is used herein to encompass any isolated Ab's such as conventional monoclonal antibody hybridomas, but also to encompass isolated monospecific antibodies produced by any cell, such as for example a sample of identical human immunoglobulins expressed in a mammalian cell line.
The variable heavy (VH) and variable light (VL) domains of the antibody are involved in antigen recognition, a fact first recognized by early protease digestion experiments. Further confirmation was found by "humanization" of rodent antibodies. Variable domains of rodent origin may be fused to constant domains of human origin such that the resultant antibody retains the antigenic specificity of the rodent parented antibody (Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81, 6851-6855). That antigenic specificity is conferred by variable domains and is independent of the constant domains is known from experiments involving the bacterial expression of antibody fragments, all containing one or more variable domains. These molecules include Fab-like molecules (Better et al (1988) Science 240, 1041); Fv molecules (Skerra et al (1988) Science 240, 1038); single-chain Fv (ScFv) molecules where the V.sub.H and V.sub.L partner domains are linked via a flexible oligopeptide (Bird et al (1988) Science 242, 423; Huston et al (1988) Proc. Natl. Acad. Sci. USA 85, 5879) and single domain antibodies (dabs) comprising isolated V domains (Ward et al (1989) Nature 341, 544). A general review of the techniques involved in the synthesis of antibody fragments which retain their specific binding sites is to be found in Winter & Milstein (1991, Nature 349, 293- 299). The term "ScFv molecules" encompasses molecules wherein the VH and VL partner domains are linked via a flexible oligopeptide. Engineered antibodies, such as ScFv antibodies, can be made using the techniques and approaches described in J. Huston et al, (1988) "Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single chain Fv analogue produced in E. coli", Proc. Natl. Acad. Sci. USA, 85, pp. 5879-5883, and in A. Pluckthun, (1991) "Antibody engineering; Advances from use of E. coli expression systems", Bio/technology 9 (6): 545-51, incorporated herein by reference.
Suitable monoclonal antibodies which are reactive as described herein may be prepared by known techniques, for example those disclosed in "Monoclonal Antibodies; A manual of techniques", H Zola (CRC Press, 1988) and in "Monoclonal Hybridoma Antibodies: Techniques and Application", S G R Hurrell (CRC Press, 1982).
A further embodiment encompasses humanized antibodies where the regions of the murine antibody that contacted the antigen, the Complementarity Determining Regions (CDRs) were transferred to a human antibody framework. Such antibodies are almost completely human and seldom cause any harmful antibody responses when administered to patients. Several chimeric or humanized antibodies have been registered as therapeutic drugs and are now widely used within various indications (Borrebaeck & Carlsson, 2001, Curr. Opin. Pharmacol. 1 : 404-408). It is preferred if the antibody is a humanized antibody. Suitably prepared non- human antibodies can be "humanized" in known ways, for example by inserting the CDR regions of mouse antibodies into the framework of human antibodies. Humanized antibodies can be made using the techniques and approaches described in Verhoeyen et al (1988) Science, 239, 1534-1536, and in Kettleborough et al, (1991) Protein Engineering, 14 (7), 773-783.
In another embodiment, antibodies also encompass completely human antibodies, which may be produced using recombinant technologies. Typically, large libraries comprising billions of different antibodies are used. In contrast to the previous technologies employing chimerization or humanization of e.g. murine antibodies this technology does not rely on immunization of animals to generate the specific antibody. Instead the recombinant libraries comprise a huge number of pre-made antibody variants wherein it is likely that the library will have at least one antibody specific for any antigen.
The frequency of administration may be determined clinically by following the decline of antibody titers in the serum of patients over time, but in any event may be at a frequency of 1 to 52 times per year, and most preferably between 1 and 12 times per year. Quantities of antibody may vary according to the severity of the disease, or half- life of the antibody in the serum, but preferably will be in the range of 1 to 10 mg/kg of patient, and preferably within the range of 1 to 5 mg/kg of patient, and most preferably 1 to 2 mg/kg of patient. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1: Different frequencies and phenotypic characteristics between FOXP3+ and FOXP3" CD3+ T cell populations, as defined by their variable TCR recognition in human peripheral blood (PBMCs) and in TIL isolated from breast tumor.
Figure 2: Analysis of Foxp3+ expression in lymphocytes present in the TILs extracted from luminal A and B breast subtypes. Tumor tissue from patient with luminal-A and luminal B was minced with scalpels and enzymatically digested by overnight incubation in collagenase Type IV. Expression of FOXP3 marker in lymphocytes present in the isolated TIL was determined by flow cytometric analysis. Representation of the Foxp3 expression level by the MFI in the CD3+CD4+TCRaP+ restricted T cells and in the CD3+CD4+TCRy5+ unrestricted T cells.
Figure 3: Positive correlation of Foxp3+ expression in lymphocytes present in the TILs and a poor clinical outcome in breast cancer. Tumor tissue from patient with luminal-A (n=3), luminal B (n=3) and patients with triple-negative breast cancer (TNBC) (n=2) was minced with scalpels and enzymatically digested by overnight incubation in collagenase Type IV. Expression of FOXP3 marker in lymphocytes present in the isolated TIL was determined by flow cytometric analysis. Representation of the percentage of FOXP3 expression in the CD3+CD4+TCRaP+ restricted T cells and in the CD3+CD4+TCRy5+ unrestricted T cells.
Figure 4: Multiparametric flow cytometry analysis of lymphocytes present in the TILs from luminal A and B breast subtypes. Lymphocytes present in the TIL were stained at the cell surface using Abs directed against CD3, CD4, CD25, CD56, CD161. After fixation and permeabilization Foxp3 and CTLA4 were stained intracellularly. Figure 5: Phenotype and functional suppressive capacity of ex vivo generated Ag specific CD3+ TCRy6+ T cells from stimulated naive CD3+ TCRy6+ T cells. Naive CD3+ TCRy5+ T cells were stimulated with zoledronic acid-treated-autologous tDCs, in presence of the nTreg polarizing medium and IL-2 (100 IU/ml) and IL-15 (10 ng/ml). (A) Overlay histogram displaying Foxp3 expression profiles and (B) suppressive capacity of Ag specific CD3+ TCRy5+ T cells expanded for 21 or 42 days.
Figure 6: In vitro induction of tumor- Ag specific CD3+ TCR Va24+ CD 1 -restricted T cells (invTreg) from stimulated naive CD3+ TCR Va24+ T cells with different nTreg polarizing medium. Naive CD3+ TCR Va24+ T cells were stimulated for 21 days with tumor - apoptotic breast tumor cell line pulsed autologous tDC as described in Fig 3 in presence of IL-2 (100 IU/ml) and IL-15 (10 ng/ml). Where indicated, TGF , RAP A and PGE2 were added. (A) Overlay histogram displaying Foxp3 expression profiles of each of the generated invTreg. (B) Frequency and (C) expression level (evaluated by MFI) of Foxp3 in CD3+ T cell culture. Dashed black line represents in (B) and (C) the frequency and the expression level of FOXP3 in naive Treg phenotypically defined by the expression a high level of CD45RA and CD25 and a low level of CD127.
Figure 7: Combination of TGF , RAPA and PGE2 induce the establishment and the expansion of tumor Antigen specific FOXP3+ CD3+ TCR Va24+ CD 1 -restricted T cell cells committed to exclusively exert regulatory activity, with an autologous MLR assay. CD3+ TCR Va24+ CD45RA+ T cells were stimulated with autologous tolerogenic DC pulsed with apoptotic breast tumor cell lines in presence of IL-2, IL-15 and nTreg polarizing medium. After 21 days of in vitro expansion in nTreg polarizing medium, suppressive capacity of ex vivo generated Tumor Ag-specific invariant Foxp3+ Treg was evaluated in the presence of (A) a low or (B) high inflammatory medium. Fresh na'ive Treg were used as control.
Figure 8: Analysis of Foxp3+ expression in human MHCII restricted CD4+ Foxp3+ CD4+ regulatory T cells (Treg) generated ex vivo from polyclonally stimulated naive CD4+ T cells with different nTreg polarizing medium. Naive CD4+ T cells were stimulated for 12 days with plate-bound anti-CD3 (4 μg/ml) in presence of IL-2 (100 IU/ml). Where indicated, TGF (5 ng/ml), RAPA (10 nM) and PGE2 (1 μΜ) were added. (A) Overlay histogram displaying Foxp3 expression profiles of each of the generated pTreg. (B) Frequency and (C) expression level (evaluated by MFI) of Foxp3 in CD4+ T cell culture. Figure 9: Comparative analysis of in vitro suppressive capacity of human Treg generated with different nTreg polarizing medium. Suppressive capacity of ex vivo generated Treg was evaluated (A) in quiescent and (B) in inflammatory context with the standard polyclonal nTreg assay. CFSE-labeled conventional T cells (Tconv) were cocultured with ex vivo generated Treg at different ratio. Percent inhibition of TconvCFSE proliferation by Treg was depicted. Fresh Treg and Tconv were used as control.
Figure 10: Combination of TGF , RAPA and PGE2 induce the establishment and the expansion of cultured Treg committed to exclusively exert regulatory activity. After 21 days of ex vivo generation in nTreg or TH-17 polarizing medium, suppressive capacity of ex vivo generated OVA- specific Treg was evaluated in the presence of a high inflammatory context inducing medium. Fresh Treg were used as control.
Figure 11: IL-17 production by stimulated OVA- ex vivo generated Treg. Specific- Treg (A) induced after the first 21 days of culture in nTreg polarizing medium or (B) expanded for 3 weeks in nTreg or TH-17 polarizing medium were tested for their IL-17- producing capacity upon stimulation with aCD3 Ab and aCD28 Ab for 2 days in IMDM medium containing IL-2, IL-1, IL-6, IL-21, and IL-23 cytokines. IL-17 was detected in supernatant culture by ELISA.
EXAMPLES
The present invention is further illustrated by the following examples. Materials and Methods
Human Blood Sample. Blood samples from healthy individuals originated from Etablissement Francais du Sang (EFS, Paris). Blood cells are collected using standard procedures.
Human tumor sample. Tumor tissue sample originated from patient with luminal A and Luminal B Breast cancer (Institut Jean Godinot, Reims). Cell Purification and Culture.
Peripheral blood mononuclear cells (PBMCs) are isolated by density gradient centrifugation on Ficoll-Hypaque (Pharmacia). PBMCs are used either as fresh cells or stored frozen in liquid nitrogen. T-cell subsets and T cell-depleted accessory cells (ACD3 cells) are isolated from either fresh or frozen PBMCs. T cell-depleted accessory cells (ACD3 cells) are isolated by negative selection from PBMCs by incubation with anti-CD3-coated Dynabeads (Dynal Biotech) and are irradiated at 3000 rad (referred to as ACD3-feeder).
CD4+ T cells are negatively selected with a CD4+ T-cell isolation kit (Miltenyi Biotec, yielding CD4+ T-cell populations at a purity of 96-99%. Subsequently, selected CD4+ T cells are labeled with anti-CD4 (13B8.2)-FITC (Beckman Coulter), anti-CD25(4E3)- APC (Miltenyi Biotec), and anti-CD127(R34.34)-PE (Beckman Coulter) before being sorted into CD4+CD127-/loCD25high (pTregs) and CD4+CD127+CD25neg/dim [conventional helper CD4 T cells (Tconv)] subpopulations using a F ACS Aria III Cell Sorter (Becton Dickinson).
CD14+ monocytes are isolated from PBMCs by positive selection using a MACS system.
CD3+ CD4+ CD127+ CD45RA+ CD25 TCRaP+ MHCII restricted (naive conventional CD4+ T cells) are isolated from PBMCs after magnetic enrichment (MACS system: CD4 microbeads) and FACs sorting. Before the sorting step, enriched CD3+ CD4+ T cells are stained with anti-CD4 (13B8.2)-FITC (Beckman Coulter), anti-CD25(4E3)- APC (Miltenyi Biotec), and anti-CD127(R34.34)-PE (Beckman Coulter), anti-TCR αβ- BV421 (IP26) (Bio legend).
CD3+ CD45RA+ invTCR Va24+ CD 1 -restricted T cells are isolated from PBMCs after magnetic enrichment (MACS system: anti-iNKT microbeads) and FACS sorting. Before the sorting step, enriched CD3+ invTCR Va24+T cells are stained with anti-CD3 (UCHT-1) V450 anti-invariant TCR Va24-JaQ (6B11)-PE (inv TCR Va24-JaQ (Becton Dickinson) and anti-CD45RA (T6D11)-FITC (Miltenyi Biotec). CD3+ CD45RA+ CD27+ TCRy5+ unrestricted T cells are isolated from PBMCs after magnetic enrichment (MACS system: TCRy5+ T cell isolation kit) and FACS sorting. Before the sorting step, enriched CD3+ TCRy5+ T cells are stained with anti-CD3 (UCHT-1) V450, anti- TCR pany5+ PE (IMMU510) (Beckman Coulter), anti-CD27- APC efluor 780 (0323) (ebioscience) and anti-CD45RA (T6D11)-FITC (Miltenyi Biotec).
T cell subsets are cultured either in IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES (IMDM-5 media) in hypoxia 2%. Breast cancer cell line and culture. The human breast cancer cell line MCF-7 was obtained from the American Type Culture Collection (USA). Cells are maintained in Dulbecco's modified Eagle's medium (DMEM; Invitrogen, USA) supplemented with 10% fetal bovine serum (FBS). MCF-7 cells are treated with 5 μg/ml Doxorubicin for 24 h or by γ irradiation (20 Gy). Extent of apoptosis is monitored by flow cytometric analysis (FACS). Cells are extensively washed prior to feeding DCs.
TIL isolation. Tumor tissue was minced with scalpels and enzymatically digested by overnight incubation in collagenase Type IV (2 mg/ mL, Roche Diagnostic GmbH) in DMEM High Glucose medium supplemented with 2 mM glutamine (Gibco), 50 mg/mL gentamycin and 0.25 % Human Serum Albumin, at 37 °C on a rotary shaker. Ex vivo generation of polyclonal functionally committed FOXP3 expressing regulatory T cells.
Ex vivo generation of polyclonal functionally committed FOXP3 expressing CD3+ TCRafi^ MHC11 restricted T cells: On day 0, T cells are seeded at 2.5 105/well in 48- well plates and stimulated with plate-bound anti-CD3 mAb (4 μg/ml) in the presence of ACD3-feeder (1 M). Cells are cultured in IMDM-5 media (IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES) with PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM. On day 2, IL-2 (lOOIU/ml) are added to the culture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml). On day 11, these CD4+ T-cell lines were further expanded by restimulation with plate-bound anti-CD3 Abs (4μ§/ιη1). The restimulations were performed in the presence of ACD3-feeder, PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM and IL-2 (100 Ul/ml). Then every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml). On day 20, the phenotype of the expanded CD4+ T cells was assessed by flow cytometry. 75% of the stimulated naive conventional T cells that became CD45RO+ express FOXP3+.
Ex vivo generation of polyclonal functionally committed FOXP 3 expressing invariant T cells: On day 0, T cells are seeded at 1 x 103/well in 96-well plates and stimulated with plate-bound anti-inv TCR Va24-JaQ (6B11) mAb (2 μ§/ι 1) in the presence of ACD3- feeder (2.5x105). Cells are cultured in IMDM-5 media with PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Every three days, IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) are added to the culture. On day 12, T cells are further expanded by restimulation with plate-bound anti- anti-inv TCR Va24-JaQ (6B11) mAb (2 in the presence of ACD3-feeder, PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM IL- 2 (100 Ul/ml) and IL-15 (10 ng/ml). Then every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). On day 21, cells are analyzed by flow cytometry. 70% of the stimulated CD3+ invTCR Va24+ RA+ T cells that became CD45RO+ express Foxp3+.
Ex vivo generation of polyclonal functionally committed FOXP3 expressing TCRy5+ T cells: On day 0, T cells are seeded at 1 x 103/well in 96-well plates and stimulated with plate-bound anti-TCRy5 mAb (2 μg/ml) in the presence of ACD3-feeder (2.5X105). Cells are cultured in IMDM-5 media (IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES) with PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM, IL- 2 (100 Ul/ml) and IL-15 (10 ng/ml). Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). On day 11, T cells were further expanded by restimulation with plate-bound anti-pan TCR γδ Abs (2μg/ml). The restimulations were performed in the presence of ACD3-feeder, PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM and IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). Then every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml). On day 21 , cells are analyzed by flow cytometry. 65% of the stimulated CD3+ CD45RA+ CD27+ TCRy5+ T cells that became CD45RO+ express Foxp3+.
Ex vivo generation of antigen specific functionally committed FOXP3 expressing T cells:
Ex vivo generation of antigen (Ovalbumun) specific functionally committed Foxp3 expressing CD3+ TCRa^ MHCII restricted T cells: a) In vitro generation of ovalbumin- loaded Tolerogenic DC from CD14+ monocytes (termed tolerogenic monocyte-derived DC (Tol-Mo-DC): monocytes are cultured in 48-well flat-bottom plates containing 0,5 ml of AIMV per well supplemented with 100 ng/ml recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF) and 10 ng/ml human recombinant IL-4 for the generation of immature DC. At day 3, 500 μΐ of the medium containing cytokines was added. On day 6, Tol-Mo-DC are 1) removed from the wells, washed twice with IMDM-5 (IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES, 2) added to wells of a 48-well plate at a concentration of 3 X 105 /ml in IMDM-5 and 3) pulsed in IMDM-5 with specific
Ag (OVA). b) Ex vivo generation and expansion of specific functionally committed FOXP3 expressing CD3+ TCRo$+ MHCII restricted T cells: On day 0, ovalbumin pulsed tDC are 1) washed twice with IMDM-5 and 2) added to wells of a 48-well plate at a concentration of 3 X 105 /ml in IMDM-5 in the presence of 2 X 105 irradiated autologous feeders, PGE2 1 μΜ, and Rapa 10 nM. Purified naive conventional CD4+ T cells (isolated from the previously frozen PBMC by FACS) are added to the pulsed tDC. On day 1 , IL-2 (lOOIU/ml) and TGF (5ng/ml) are added to the coculture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml (T cell cloning medium). On day 12, these T-cells are further expanded by restimulation with ova-pulsed tDC in the presence of ACD3-feeder, PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM, IL-2 (100 Ul/ml). Once T cells begin to expand, they can be split every 2 to 3 days with T cell cloning medium and irradiated feeder. On day 21, cells are analyzed by flow cytometry. 85 % of the stimulated naive conventional CD4+ T cells that became CD45RO+ express Foxp3+. To confirm that the Ova-specific memory CD3+ TCRa,p+ MHCII restricted T cells are committed to exclusively exert regulatory activity, whatever culture condition of stimulation, after 21 days of expansion in nTreg polarizing medium, the ova-specific-pTreg are further cultured for 3 weeks either in nTreg polarizing medium (comprising the combination of IL-2, TGF , PGE2 and rapamycin) or TH-17 polarizing medium (IMDM medium containing IL-2 IL-1 IL-6, IL-21 IL- 23 cytokines). The 21 -day-expanded- Foxp3 expressing CD3+ CD4+ TCRa,p+ MHCII restricted T cells are stimulated with plate-bound anti-CD3 mAb (4 μg/ml) in the presence of ACD3-feeder (1 M) in 48-well plates and every three days, half of the supernatant volume is discarded and replaced with fresh T cell cloning medium or TH-17 polarizing medium for 21 days.
Ex vivo generation of tumor-antigen specific functionally committed FOXP3 expressing CD3+ invTCR Va24+ CD Id- restricted T cells: a) In vitro generation of tumor- loaded Tolerogenic DC from CD14+ monocytes (termed tolerogenic monocyte-derived DC (tDC): monocytes are cultured in 48-well flat-bottom plates containing 0,5 ml of AIMV per well supplemented with 100 ng/ml recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF) and 10 ng/ml human recombinant IL-4 and AM580 (100 nM) for the generation of immature DC expressing CD Id. At day 3, 500 μΐ of the medium containing cytokines are added. At day 5, a portion of tDCs are co-cultured with apoptotic MCF-7 cells at a DC/tumor cell ratio of 1 :2 for 24h in AIMV with GM-CSF (100 ng/niL), IL-4 (10 ng/mL). Another portion of tDC are freezed at 2 x 106 / per vial vial- in 90% FBS -10% DMSO. b) Ex vivo generation and expansion of tumor-antigen specific functionally committed Foxp3 expressing CD3+ invTCR Va24+ CD Id- restricted T cells: On day 0, tumor-antigen pulsed tDC are 1) washed twice with IMDM-5 and 2) added to wells of a 48-well plate at a concentration of 3 X 105 /ml in
IMDM-5 in the presence of 2 X 105 irradiated autologous feeders, PGE2 1 μΜ, and Rapa 10 nM. Purified CD3+ CD45RA+ invTCR Va24+ CD1- restricted T cells (isolated from the previously frozen PBMC by FACS) are added to the pulsed tDC. On day 1, IL-2 (lOOIU/ml), IL-15 (10 ng/ml) and TGF (5ng/ml) are added to the coculture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) (T cell cloning medium). On day 12, these T-cells are further expanded by restimulation with tumor Ag-pulsed tDC in the presence of ACD3-feeder, PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM, IL- 2 (100 Ul/ml) and IL-15 (10 ng/ml). Once T cells begin to expand, they can be split every 2 to 3 days with T cell cloning medium and irradiated feeder. On day 21, cells are analyzed by flow cytometry. 75 % of the stimulated CD3+ CD45RA+ invTCR Va24+ cells that became CD45RO+ express Foxp3+. Ex vivo generation of phospho-antigen specific functionally committed FOXP3 expressing CD3+ TCRyd+ unrestricted T cells: a) In vitro generation of Tolerogenic DC from CD14+ monocytes (termed tolerogenic monocyte-derived DC (Tol-Mo-DC): monocytes are cultured in 48-well flat-bottom plates containing 0,5 ml of AIMV per well supplemented with 100 ng/ml recombinant human granulocyte- macrophage colony- stimulating factor (GM-CSF) and 10 ng/ml human recombinant IL-4 for the generation of immature DC. At day 3, 500 μΐ of the medium containing cytokines was added. On day 6, generated Tol-Mo- DC are removed from the wells, washed twice with IMDM-5 (IMDM supplemented with 5% SVF, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES, freezed or used for the generation and expansion of phospho- antigen specific functionally committed FOXP3 expressing CD3+ TCRy<T unrestricted T cells. b) Ex vivo generation and expansion of phospho-antigen specific functionally committed FOXP3 expressing CD3+ TCRy<T unrestricted T cells: On day 0, tDC are added to wells of a 48-well plate at a concentration of 3 X 105/ml in IMDM-5 in the presence of 2 X 105 irradiated autologous feeders, PGE2 1 μΜ, and Rapa 10 nM and zoledronic acid (100 nM). Purified CD3+ CD45RA+ TCRy<T unrestricted T cells (isolated from the previously frozen PBMC by FACS) are added to the pulsed tDC. On day 1, IL-2 (lOOIU/ml), IL-15 (10 ng/ml) and TGF (5ng/ml) are added to the coculture. Every three days, half of the supernatant volume is discarded and replaced with fresh IMDM-5 with IL-2 (100 Ul/ml) and IL-15 (10 ng/ml) (T cell cloning medium). On day 12, these T-cells are further expanded by restimulation with tDC in the presence of ACD3-feeder, PGE2 1 μΜ, TGF 5 ng/ml, Rapa 10 nM, IL-2 (100 Ul/ml), IL-15 (10 ng/ml) and zoledronic acid (100 nM). Once T cells begin to expand, they can be split every 2 to 3 days with T cell cloning medium and irradiated feeder. On day 21, cells are analyzed by flow cytometry. 75 % of the stimulated CD3+ CD45RA+ TCRy5+ T cells that became CD45RO+ express Foxp3+.
In vitro generation of stimulator cells for MLR assay: monocytes are cultured in 48- well flat-bottom plates containing 0.5 ml of RPMI-5 per well supplemented with 20 ng/ml recombinant human granulocyte-macrophage colony- stimulating factor (GM- CSF) and 20 ng/ml human recombinant IL-4 for the generation of immature DC (iDC). At day 3, 500 μΐ of the medium containing cytokines are added. At day 5, a portion of iDC are co-cultured with apoptotic MCF-7 cells at a DC/tumor cell ratio of 1 :2 for 24h in RPMI 1640 supplemented with GM-CSF (20 ng/mL), IL-4 (20 ng/niL) and 5 % FBS. Another portion of iDC are freezed at 2 x 106 / per vial - in 90% FBS -10% DMSO. When indicated, pulsed DCs are matured with tumor necrosis factor a (TNF-a; 20 ng/mL final) and PGE2 (1 μΜ) for 2 days (mDC). In some experiments, TNF and PGE2 (at the same concentrations), or lipopolysaccharide (LPS; 10-1000 ng/mL; Sigma) are added directly to MLRs. Antigen- loaded DC stimulators are irradiated at 30 Gy. IL- 17 detection by ELISA.
The presence of IL-17 in the culture supernatant is measured by ELISA. The recognition of IL-17 by an anti-IL-17 antibody may be carried out by conventional methods known in the art such as a sandwich ELISA anti-IL-17. The ELISA is developed by any colorimetric means known in the art such as for example using a detection antibody labelled with biotin, a poly-streptavidin HRP amplification system and an o-phenylenediamine dihydrochloride substrate solution.
One example of said method is the following:
coating a plate with the capture antibody, such as for example an anti-IL17 antibody,
- blocking the plate with a blocking buffer (such as, for example, casein 2% in
PBS) during 90 min at 37°C,
incubating the plate during 90 min at 37°C with a dilution series of IL-17 standard, samples or negative controls,
incubating the plate 90 min at 37°C with the detection antibody such as for example a biotinylated anti-IL-17 antibody,
incubating the plate with streptavidin-HRP during 30 min at 37°C and developing the complex with an o-phenylenediamine dihydrochloride (OPD) substrate solution during 30 min. After stopping the enzymatic reaction, the intensity of the resulting color is determined by spectrophotometric methods at 490 nm.
The person skilled in the art considers that an IL-17 level inferior to 200 ng/ml, 100 ng/ml, 50 ng/ml corresponds to no secretion or low secretion of IL-17 after calculation with the standard curve. Flow Cytometry Analysis. mAb labeling. The following conjugated mAbs are used, a) for CD3+ T cells : anti-CD4(SK3)-PerCP-eFluor 710, anti-TCRap(IP26)-APC (ebioscience), anti-CD25 (B1.49.9)-PeCy55, anti-CD 127(R34.34)-APC-AF700 (Beckman Coulter), anti-CD3(UCHTl)- BB515 anti-invariant TCR Va24-JaQ (6B11)- PE, anti-Foxp3 (259D/C7)-PE-CF594 and anti-CD 152 (BNI3)-BV421, anti-CD 161 (DX12) BV605 and anti-CD56(NCAM 16.2) BU395 (Becton Dickinson), anti-TCR αβ- BV421 (IP26) (Bio legend), anti- TCR pan γδ+ PE (IMMU510) (Beckman Coulter) and anti-CD27- APC efluor 780 (0323) (ebioscience). Cells are stained for surface markers (at 4°C in the dark for 30 min) using mixtures of Ab diluted in PBS containing BSA/NaNs (0.5% BSA, 0.01% NaN3) (FACS buffer). Foxp3 and CTLA-4 intracellular staining are performed with FOXP3 staining kit obtained from ebioscience according to the manufacturer's instructions. Appropriate isotype control Abs are used for each staining combination. Samples are acquired on a BD LSR FORTESSA flow cytometer using BD FACSDIVA 8.0.1 software (Becton Dickinson). Results are expressed in percentage (%) or in mean fluorescence intensity (MFI).
CFSE staining. Tconv are stained with 1 μΜ carboxy- fluorescein succinimidyl ester (CFSE) (CellTrace cell proliferation kit; Molecular Probes/Invitrogen) in PBS for 8 min at 37 °C at a concentration of 1 x 107 cells/mL. The labeling are stopped by washing the cell twice with RPMI 1640 culture medium containing 10% FBS. Cells are then resuspended at the desired concentration and subsequently used for proliferation assays.
7-AAD (7-amino-actinomycin D) staining. Apoptosis of stimulated CFSE- labeled or unlabeled nTregs and Tconv was determined using the 7-AAD assay. Briefly, cultured cells are stained with 20 μg/mL nuclear dye 7-AAD (Sigma- Aldrich) for 30 min at 4 °C. FSC/7-AAD dot plots distinguish living (FSChigh/7-AAD") from apoptotic (FSChigh/7- AAD+) cells and apoptotic bodies (FSClow/7- AAD+) and debris ((FSClow/7-AAD"). Living cells are identified as CD3+ 7-AAD" FSC+ cells.
Functional Assays.
T-cell proliferation. T-cell proliferation is assessed CFSE dilution assay in RPMI supplemented with 5% FBS, 100 IU/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1 mM nonessential amino acids, glutamax and 10 mM HEPES (RPMI-5 media) in normoxia. At coculture completion, stimulated CFSE-labeled Tconv are harvested, costained with anti-CD3 mAb and 7-AAD, and the percentage of living proliferating cells (defined as CFSE low fraction) in gated CD3+ 7-AAD- cells is determined by flow cytometry.
Standard polyclonal cell-cell contact Treg suppression assay: CFSE-labeled Tconv (4 x 104 per well), used as responder cells, are cultured with ACD3- feeder (4 χ 104 per well) in the presence or absence of defined amounts of Foxp3 T cells (blood Treg or ex vivo generated T cells) for 4 to 5 d. Cultures are performed in round-bottom plates coated with 0.2 μg/mL anti-CD3 mAb in 200 of complete RPMI medium. Results are expressed as the percentage of proliferating CFSE low T cells or as a percentage of suppression calculated as follows: (100 x [(percentage of Tconv CFSE low cells - percentage of Tconv CFSE low in coculture with nTregs)/percentage of Tconv CSFE low cells.
Autologous MLR suppression assay: CFSE-labeled Tconv CD4 CD25 T cells (5 x 104) are stimulated either with 1 x 104 pulsed iDC in RPMI-5 media or with 5 x 103 pulsed -mDC in IMDM-5 media supplemented with IL-2 (20 IU/ml) IL-lb (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml) and IL-23 (30 ng/ml) in the presence or absence of defined amounts of Foxp3 T cells (blood Treg or ex vivo generated T cells) for 5 to 6 d. When indicated, culture is performed in IMDM-5 media supplemented with IL-2 (20 IU/ml) IL-Ιβ (10 ng/ml), IL-6 (30 ng/ml), IL-21 (50 ng/ml) and IL-23 (30 ng/ml). Results are expressed as the percentage of proliferating CFSE low T cells or as a percentage of suppression calculated as follows: (100 x [(percentage of Tconv CFSE low cells - percentage of Tconv CFSE low in coculture with nTregs)/percentage of Tconv CSFE low cells.
Measurement of D A methylation: Classically, a stable Treg genetic signature consisted of highly demethylated CpG islands within the conserved non-coding sequence 2 (CNS2) of the Treg specific demethylation region (TSDR). DNA methylation analysis of the TSDR region of the gene FOXP3 was evaluated by quantitative PCR after bisulfite treatment of genomic DNA as previously described by Christopher Fuhrman (Fuhrman et al, Divergent Phenotypes of Human Regulatory T Cells Expressing the Receptors TIGIT and CD226, 2015, Journal of immunology). Briefly Nucleotides were isolated with AllPrep DNA/RNA Mini Kit (Qiagen) or DNeasy tissue kit (Qiagen), as appropriate. Bisulfite treatment of genomic DNA was performed on 500 ng DNA with the EZ DNA Methylation Kit (Zymo Research). DNA standards originated from unmethylated bisulfite-converted human EpiTect control DNA (Qiagen) or universally methylated bisulfite-converted human control DNA (Zymo Research). To obtain a large quantity of standard, the TSDR was PCR-amplified using the following reaction: 50 μΐ reaction volume containing 25 μΐ of ZymoTaq PreMix buffer (Zymo Research) and 0.5 μΜ each of the primers FOXP3_TSDRfwd (5'- ATATTTTTAGATAGGGATATGGAGATGATTTGTTTGG-3' SEQ ID NO: 1) and FOXP3_TSDRrev (5'-AATAAACATCACCTACCACATCCACCAACAC-3' - SEQ ID NO: 2). After incubation at 95°C for 10 min, amplification was performed as follows: 50 cycles at 95°C for 30 s, 55°C for 30 s, and 72°C for 1 min. Amplified PCR products were purified with the QIAquick Gel Extraction Kit (Qiagen). The concentration of purified control TSDR DNA was determined with a GE NanoVue spectrophotometer (GE Healthcare Life Sciences). TSDR real-time PCR was performed with probes that targeted methylated or demethylated target sequences. The reaction was performed in 96-well white trays with a Roche LightCycler 480 system (Roche Diagnostics). Each reaction contained 10 μΐ LightCycler 480 Probes Master Mix (Roche), 10 ng of bisulfite converted DNA sample or standards, 1 μΜ of each primer, and 150 nM of each probe with a final reaction value of 20 μΐ. The probes used for amplification were TSDR- Forward 5 '-GGTTTGTATTTGGGTTTTGTTGTTAT AGT-3 ' (SEQ ID NO: 3) and TSDR-Reverse 5 '-CT AT AAAAT AAAAT ATCTACCCTCTTCTCTTCCT-3 ' (SEQ ID NO: 4). The probes for target sequence detection were FAM-labeled methylated probe, FAM-CGGTCGGATGCGTC-MGB-NFQ (SEQ ID NO: 5), or VIC-labeled unmethylated probe, VIC-TGGTGGTTGGATGTGTTG-MGB-NFQ (SEQ ID NO: 6). All samples were tested in triplicate. The protocol for real-time amplification is as follows: after initial denaturation at 95°C for 10 min, the samples were subjected to 50 cycles at 95°C for 15 s and at 61°C for 1 min. Fourteen different ratios of fully methylated and demethylated template were used as real-time standards. A six-order polynomial equation was used to extrapolate the percentage of cells demethylated at the TSDR for each sample.
Measurement of histone acetylation: Histone acetylation analysis of the four different sites of FOXP3 gene was evaluated by ChIP assay, as previously described by Ling Lu (Ling Lu et al, PNAS 2014). Briefly, 50,000 cells of each treated nTreg cell sample were harvested and cross-linked with 1% formaldehyde, and then lysed with 120μί of lysis buffer [50 mM Tris-HCl, pH 8.0, 10 mM EDTA, 1% (wt/vol) SDS, protease inhibitor mix (1 : 100 dilution; Sigma), 1 mM PMSF, 20 mM Na-butyrate]. The chromatin in the lysate was sonicated to 500-800-bp fragments and then diluted with 800μί of RIP A ChIP buffer [10 mM Tris-HCl, pH 7.5, 140 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 1% (vol/vol) Triton X-100, 0.1% (wt/vol) SDS, 0.1% (wt/vol) Na- deoxycholate, protease inhibitor mix (1 : 100 dilution; Sigma), 1 mM PMSF, and 20 mM Na- butyrate]. Dynabeads protein G (ΙΟμί; Invitrogen) was incubated with ^g ofH3K4me3 (Abeam) or H3K9ac (Cell Signaling) or normal rabbit IgG negative control ChlP-grade antibodies for 2 h separately. Then, ΙΟΟμΕ of the sheared chromatin was immunoprecipitated with pretreated antibody-bead complexes and another ΙΟΟμΕ of the sheared chromatin for total input DNA extraction separately. Immunoprecipitated DNA was quantified by real-time PCR with following primers: promoter, 5'-ACC GTA CAG CGT GGT TTT TC-3' (SEQ ID NO: 7) and 5'-CTA CCT CCC TGC CAT CTC CT-3' (SEQ ID NO: 8); CNS1, 5'- CCC AAG CCC TAT GTG TGATT-3' (SEQ ID NO: 9) and 5'-GTG TGT CAG GCC TTG TGC TA-3' (SEQ ID NO: 10) ; CNS2, 5'-GTC CTC TCC ACAACC CAA GA-3' (SEQ ID NO: 11) and 5'-GAC ACC ACG GAG GAA GAG AA -3' (SEQ ID NO: 12); and CNS3, 5'-AGG TGC CGA CCT TTA CTG TG-3' (SEQ ID NO: 13) and 5'- ACA ATA CGG CCT CCT CCT CT-3' (SEQ ID NO: 14). Results a) Presence of FOXP3+ expressing T cells in tumor infiltrating lymphocytes (TILs) isolated from luminal-B breast cancer.
Luminal A and B subtypes are both estrogen-receptor-positive (ER+) and low-grade, with luminal A tumors growing very slowly and luminal B tumors growing more quickly. Luminal A tumors have the best prognosis. Luminal B tumors are associated with a poor clinical outcome. We examined by flow cytometry the phenotype of lymphocytes in the TIL isolated from both luminal subtypes breast cancer and found the presence of Foxp3 expression in CD3+ CD4+ ΤΟίαβ+ MHCII restricted and CD3+ CD4+ TCRy<5+ unrestricted T cells. No Foxp3 was detected in TILs extracted from Luminal A breast tumor (Figure 2). Moreover, a positive correlation is observed between a high percentage of Foxp3 expression in CD3+ CD4+ TCRaP+ MHCII restricted T cells and in CD3+ CD4+ TCRy5+ unrestricted T cells, and a poor clinical outcome in breast cancer (Figure 3). Foxp3 expressing CD3+ CD4+ TCRa,p+ MHCII restricted T cells and Foxp3 expressing CD3+ TCRa,p+ unrestricted T cells represent approximately 20 % of the CD3+ TCRaP T cells and 23 % of the CD3+ TCRy5+ respectively in the studied sample. Foxp3 expressing CD3+ TCRy5+ T cells present a same phenotypic profile as Foxp3+ CD3+ TCRaP+ T cells. These Foxp3+ TCRy5+ T cell population express levels of Foxp3, CD25 and CTLA4 similar to those of Foxp3+ CD3+ TCRaP+ T cells (Figure 4). b) Ex vivo generation and expansion of specific CD3+ TCRy5+ expressing Foxp3 committed to exclusively exert regulatory activity.
As studies suggested that the suppressive potential of antigen- specific Treg was much greater than that of polyclonal Treg, we set up a method to ex vivo generated and expanded antigen specific Foxp3 expressing CD3+ TCRy5+ unrestricted T cells, committed to exclusively exert regulatory activity, whichever culture condition of stimulation is.
Figure 5 shows that naive CD3+ TCRy5+ T cells (CD3+CD45RA+ CD27+ TCRy5+ T cells) stimulated with zoledronic acid-treated-autologous tDCs, in presence of the nTreg polarizing medium comprising the combination of IL-15, IL-2, TGF , PGE2 and rapamycin, express Foxp3 after 21 days expansion and exhibit significant functional suppressive activity, as assessed by the standard polyclonal cell-cell contact Treg suppression assay. Interestingly the 21 -day-expanded FOXP3 expressing CD3+ TCRy5+ T cells maintain their Foxp3 level and their suppressive activity, after a further 21 -day- culture in nTreg polarizing medium. c) Optimal conditions for inducing Foxp3 expression in invariant Tcells
Starting from naive CD3+ invTCR Va24+ T cells isolated from human PBMCs, different nTreg polarizing medium were assessed for their capacity to induce the expression the differentiation of Foxp3+ cells with suppressive function.
Figure 6 shows that cultured naive CD3+ invTCR Va24+ T cells exhibit a variable level of Foxp3 dependent on their culture condition of stimulation. Polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin results in a higher Foxp3 expression over combinations of IL-2, TGF and rapamycin, IL-2 and PGE2, or IL-2 alone. Moreover, the combination of IL-2, TGF , PGE2 and rapamycin results in an optimal intensity of Foxp3 expression in the invTCR Va24+ T cells, as compared to the other combinations.
Furthermore, it is interesting to note that only naive CD3+ invTCR Va24+ T cells stimulated with the polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin express level and intensity of Foxp3 similar or higher to those of blood naive regulatory T cells (CD3+ TCRa CD4+ CD127 /low CD45RA+ CD25+), corresponding to our positive control. d) Tumor Ag-specific CD3+ TCR Va24+ T cells maintain their ability to perform suppressive function in pro -inflammatory conditions Figure 7A shows that tumor Ag-specific memory invTCR Va24+ T cells ex vivo generated and expanded in the presence of the nTreg polarizing medium above described are endowed of a higher suppressive activity than fresh Foxp3 expressing CD3+ CD4+ TCRaP+ MHCII restricted T cells when using an autologous MLR coculture assay. Furthermore, Figure 7B shows that these tumor Ag-specific invTCR Va24+ T cells still maintain their suppressive activity, when the autologous MLR coculture assay are performed in presence of a high inflammatory medium containing IL-2 IL-1 IL-6, IL-21 IL-23 cytokines, while fresh Foxp3 expressing CD3+ CD4+ TCRaP+ MHCII restricted T cells lose their suppressive activity. e) Optimal conditions for inducing Foxp3 expression in naive CD3+ CD4+ TCRa,p+ MHCII restricted T following polyclonal and antigen- specific activation. Starting from naive conventional CD4+ T cells (CD3+ CD4+ CD127+ CD45RA+ CD25 TCRaP+ MHCII restricted) isolated from human PBMCs, different nTreg polarizing medium were assessed for their capacity to induce the differentiation of Foxp3+ cells with suppressive function.
Figure 8 shows that, when ex vivo activated polyclonally with anti-CD3 mAbs, naive conventional CD4+ T cells exhibit a variable level of Foxp3 dependent on their culture condition of stimulation. Polarizing medium comprising the combination of IL-2, TGF and rapamycin or IL-2, TGF , rapamycin and PGE2 results in a higher Foxp3 expression over combinations of IL-2 and PGE2, or IL-2 alone (B). Moreover, the combination of IL-2, TGF , rapamycin and PGE2 results in an optimal intensity of Foxp3 expression in the CD3+ CD4+ TCRa,p+ MHCII restricted T cells, as compared to the other combinations (C).
It is interesting to note that only naive conventional CD4+ T cells, stimulated with the polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin, express level and intensity of Foxp3 similar or higher to those of blood naive regulatory T cells (CD3+ TCRa + CD4+ CD127 /low CD45RA+ CD25+), corresponding to our positive control.
We next evaluated the functional suppressive capacity of the Foxp3 expressing CD3+ CD4+ TCRa,p+ MHCII restricted T cells polyclonally stimulated. Figure 9A shows that CD3+ CD4+ TCRaP+ MHCII restricted T cells, ex vivo generated and expanded for 21 days, using polyclonal stimulation, in the presence of the nTreg polarizing medium comprising the combination of IL-2, TGF , PGE2 and rapamycin, display a higher suppressive activity compared with both those generated in the presence of the nTreg polarizing medium comprising the combination of IL-2, TGF , rapamycin without PGE2 and fresh FOXP3 expressing CD3+ CD4+ TCRaP+ MHCII restricted T cells, when using the standard polyclonal cell-cell contact Treg suppression assay. Furthermore, Figure 9B shows that these 21-day-expanded-FOXP3 expressing CD3+ CD4+ TCRa,p+ MHCII restricted T cells still maintain their suppressive activity, when the functional suppressive assay is performed in presence of a highly- inflammatory medium containing IL-2 IL-1 IL-6, IL-21 IL-23 cytokines, while fresh FOXP3 expressing CD3+ CD4+ TCRa,p+ MHCII restricted T cells lose their suppressive capacity under these culture condition of stimulation.
To confirm that the Ova-specific CD3+ TCRa,p+ MHCII restricted T cells are committed to exclusively exert regulatory activity, whatever culture condition of stimulation, after 21 days of expansion in nTreg polarizing medium, the ova-specific- pTreg are further cultured for 3 weeks either in nTreg or TH-17 polarizing medium (IMDM medium containing IL-2 IL-1 IL-6, IL-21 IL-23 cytokines) and were tested for 1) their functional suppressive capacity in the presence of a high inflammatory context (Figure 10) and 2) for their IL-17-producing capacity when stimulated through CD3 and CD28 as described above (Figure 11). After a further 21 -day-culture either in nTreg or TH-17 polarizing medium, Ova-specific CD3+ TCRa,p+ MHCII restricted T cells not only still retain, in a high inflammatory context, functional suppressive activity (Figure 10), but also produce low level of IL-17 (Figure 11B). By contrast fresh Foxp3 expressing CD3+ TCRa,p+ MHCII restricted T cells lose their suppressive function while producing IL-17 in this inflammatory context.

Claims

1. An immunogenic product comprising at least one inactivated ex vivo generated Foxp3+ regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population.
2. A pharmaceutical composition comprising at least one inactivated ex vivo generated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient.
3. A vaccine composition comprising at least one inactivated ex vivo generated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one adjuvant.
4. A pharmaceutical composition comprising at least one ex vivo generated regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population and at least one pharmaceutically acceptable excipient, wherein said at least one ex vivo generated regulatory T cells population remains stable when placed in inflammatory condition.
5. The immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 1 to 3 for use in treating cancer.
6. A method for preparing the immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 1 to 3, comprising: identifying from a tumor sample obtained from the subject the at least one regulatory T cells population among a MHCII restricted CD4+ Foxp3+ regulatory T cells population, a γδ Foxp3+ regulatory T cells population and an invariant Foxp3+ regulatory T cells population, that is overrepresented, ex vivo generating the at least one overrepresented regulatory T cells population, inactivating the at least one ex vivo generated regulatory T cell population.
7. The pharmaceutical composition according to claim 4 for use in cell therapy.
8. The pharmaceutical composition according to claim 4 for use in treating inflammatory or autoimmune diseases or for preventing transplant rejection or graft versus host disease (GVHD).
9. The immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 1 to 4 wherein the at least one ex vivo generated regulatory T cells population are obtained by a method comprising: for the MHCII restricted CD4+ Foxp3+ regulatory T cells population: culturing CD3+ CD4+ CD25 T cells in the presence of a TCRap cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the γδ Foxp3+ regulatory T cells population: culturing CD3+ TCRy5 + T cells in the presence of a γδ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the invariant Foxp3+ regulatory T cells population: culturing CD3+ Va24+ T cells in the presence of an invariant T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days.
10. The immunogenic product, pharmaceutical composition or vaccine composition according to claim 9, wherein the TCRa-β cell activator is a polyclonal TCRaP cell activator, preferably an anti-CD3 antibody or an anti-TCRaP antibody; the γδ T cell activator is a polyclonal γδ T cell activator, preferably an anti-TCR γδ antibody or a non peptide phosphoantigen; and the invariant T cell activator is a polyclonal invariant T cell activator, preferably a Va24 activator.
11. The immunogenic product, pharmaceutical composition or vaccine composition according to claim 9, wherein the TCRa-β cell activator is an antigen-specific TCRa-β cell activator, preferably tolerogenic dendritic cells (DCs) and pulsed with at least one self-peptide antigen; the γδ T cell activator is an antigen-specific γδ T cell activator, preferably tolerogenic dendritic cells (DCs) and pulsed with at least one bisphosphonate, preferably at least one aminobiphosphonate and the invariant T cell activator is an antigen-specific invariant T cell activator, preferably tolerogenic dendritic cells (DCs) expressing CD1 and pulsed with at least one non peptide lipid antigen.
12. The immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 9 to 11, wherein the cAMP activator is prostaglandin E2 (PGE2), an EP2 or EP4 agonist, a membrane adenine cyclase activator or a metabotropic glutamate receptors agonist.
13. The immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 9 to 11, wherein the TGF pathway activator is TGF , bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin and nodal .
14. The immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 9 to 11, wherein the mTOR inhibitor is rapamycin, rapamycin analogs, wortmannin; theophylline; caffeine; epigallocatechin gallate (EGCG), curcumin, resveratrol; genistein, 3, 3-diindolylmethane (DIM), LY294002 (2-(4-morpholinyl)-8-phenyl-4H-l-benzopyran-4-one), PP242, PP30, Torinl, Ku-0063794, WAY-600, WYE-687, WYE-354, GNE477, NVP-BEZ235, PI- 103, XL765 and WJD008.
The immunogenic product, pharmaceutical composition or vaccine composition according to anyone of claims 9 to 11, wherein the at least one ex vivo generated Foxp3+ regulatory T cells population are expanded by a method comprising:
For the MHCII restricted CD4+ Foxp3+ regulatory T cells population: culturing CD3+ CD4+ CD25 T cells in the presence of a TCRap cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the γδ Foxp3+ regulatory T cells population: culturing CD3+ TCRy5 + T cells in the presence of a γδ T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days; for the invariant Foxp3+ regulatory T cells population: culturing CD3+ Va24+ T cells in the presence of an invariant T cell activator and the following agents: i) an cAMP (Cyclic adenosine monophosphate) activator, ii) a TGF (Transforming growth factor beta) pathway activator, iii) a mTOR inhibitor, and optionally iv) at least one cytokine selected in the group of IL-2, IL-7, IL-15 and TSLP, for at least 5 days.
PCT/EP2017/069824 2016-08-05 2017-08-04 Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells WO2018024895A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP17752074.9A EP3493833A1 (en) 2016-08-05 2017-08-04 Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells
US16/322,188 US20190167791A1 (en) 2016-08-05 2017-08-04 Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662371370P 2016-08-05 2016-08-05
US62/371,370 2016-08-05
EP16196432 2016-10-28
EP16196432.5 2016-10-28

Publications (1)

Publication Number Publication Date
WO2018024895A1 true WO2018024895A1 (en) 2018-02-08

Family

ID=57218756

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/069824 WO2018024895A1 (en) 2016-08-05 2017-08-04 Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells

Country Status (1)

Country Link
WO (1) WO2018024895A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019243461A1 (en) * 2018-06-19 2019-12-26 Fondazione Telethon Production of engineered dendritic cells and uses thereof
WO2021118374A1 (en) * 2019-12-12 2021-06-17 Gdański Uniwersytet Medyczny Intrathecal administration of t regulatory cells in the treatment of multiple sclerosis
US11111493B2 (en) 2018-03-15 2021-09-07 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
WO2022029080A1 (en) * 2020-08-03 2022-02-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Population of treg cells functionally committed to exert a regulatory activity and their use for adoptive therapy
CN114292813A (en) * 2022-03-02 2022-04-08 北京市希波生物医学技术有限责任公司 Culture medium formulations for activation of the global anti-tumor immune system and methods for preparing agonist-activated global immune effector cells
WO2024020531A1 (en) * 2022-07-21 2024-01-25 Tract Therapeutics, Inc. Immune cell expansion and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007143582A2 (en) * 2006-06-05 2007-12-13 Baylor College Of Medicine Reversal of the suppressive function of specific t cells via toll-like receptor 8 signaling
US20160030443A1 (en) * 2013-03-14 2016-02-04 Rongfu Wang Methods and compositions for modulating regulatory t cell function
US20160136204A1 (en) * 2005-03-18 2016-05-19 The Board Of Trustees Of The Leland Stanford Junior University Production and therapeutic uses of th1-like regulatory t cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160136204A1 (en) * 2005-03-18 2016-05-19 The Board Of Trustees Of The Leland Stanford Junior University Production and therapeutic uses of th1-like regulatory t cells
WO2007143582A2 (en) * 2006-06-05 2007-12-13 Baylor College Of Medicine Reversal of the suppressive function of specific t cells via toll-like receptor 8 signaling
US20160030443A1 (en) * 2013-03-14 2016-02-04 Rongfu Wang Methods and compositions for modulating regulatory t cell function

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
A. PLUCKTHUN: "Antibody engineering; Advances from use of E. coli expression systems", BIO/TECHNOLOGY, vol. 9, no. 6, 1991, pages 545 - 51
BETTER ET AL., SCIENCE, vol. 240, 1988, pages 1041
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423
BORREBAECK; CARLSSON, CURR. OPIN. PHARMACOL., vol. 1, 2001, pages 404 - 408
H ZOLA: "Monoclonal Antibodies; A manual of techniques", 1988, CRC PRESS
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879
J. HUSTON ET AL.: "Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single chain Fv analogue produced in E. coli", PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883, XP000872837, DOI: doi:10.1073/pnas.85.16.5879
KETTLEBOROUGH ET AL., PROTEIN ENGINEERING, vol. 14, no. 7, 1991, pages 773 - 783
MONTOYA CJ ET AL., IMMUNOLOGY, vol. 122, no. 1, September 2007 (2007-09-01), pages 1 - 14
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
S G R HURRELL: "Monoclonal Hybridoma Antibodies: Techniques and Application", 1982, CRC PRESS
SKERRA ET AL., SCIENCE, vol. 240, 1988, pages 1038
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
WARD ET AL., NATURE, vol. 341, 1989, pages 544
WINTER; MILSTEIN, NATURE, vol. 349, 1991, pages 293 - 299

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11111493B2 (en) 2018-03-15 2021-09-07 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11421228B2 (en) 2018-03-15 2022-08-23 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11608500B2 (en) 2018-03-15 2023-03-21 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
WO2019243461A1 (en) * 2018-06-19 2019-12-26 Fondazione Telethon Production of engineered dendritic cells and uses thereof
CN112601811A (en) * 2018-06-19 2021-04-02 泰莱托恩基金会 Production of engineered cells and uses thereof
WO2021118374A1 (en) * 2019-12-12 2021-06-17 Gdański Uniwersytet Medyczny Intrathecal administration of t regulatory cells in the treatment of multiple sclerosis
CN114786688A (en) * 2019-12-12 2022-07-22 格但斯克医科大学 Intrathecal administration of regulatory T cells in the treatment of multiple sclerosis
WO2022029080A1 (en) * 2020-08-03 2022-02-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Population of treg cells functionally committed to exert a regulatory activity and their use for adoptive therapy
CN114292813A (en) * 2022-03-02 2022-04-08 北京市希波生物医学技术有限责任公司 Culture medium formulations for activation of the global anti-tumor immune system and methods for preparing agonist-activated global immune effector cells
CN114292813B (en) * 2022-03-02 2022-11-08 北京市希波生物医学技术有限责任公司 Culture medium formulations for activating the global anti-tumor immune system and methods for preparing agonist-activated global immune effector cells
WO2024020531A1 (en) * 2022-07-21 2024-01-25 Tract Therapeutics, Inc. Immune cell expansion and uses thereof

Similar Documents

Publication Publication Date Title
US11365392B2 (en) Ex vivo generation of MHCII restricted CD4+ FOXP3+ regulatory T cells and therapeutic uses thereof
WO2018024895A1 (en) Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells
WO2018024894A1 (en) Ex vivo generation of mhcii restricted cd4+foxp3+ regulatory t cells and therapeutic uses thereof
AU2005220854B2 (en) Regulatory T cells and their use in immunotherapy and suppression of autoimmune responses
US11198851B2 (en) Ex vivo generation of γδ Foxp3+ regulatory T cells and therapeutic uses thereof
US9114100B2 (en) Methods of treatment using ex vivo expansion of cord blood T cells
JP6422344B2 (en) Methods for increasing allogeneic antigen-reactive regulatory T cells
JP2022028826A (en) EX VIVO GENERATION OF γδFOXP3+ REGULATORY T CELLS AND THERAPEUTIC USES THEREOF
JP2015513403A5 (en)
KR20230018376A (en) Treatment and prevention of cancer using virus-specific immune cells expressing chimeric antigen receptors
US20190167791A1 (en) Immunotherapeutic uses of ex vivo generated foxp3+ regulatory t cells
US20130101567A1 (en) Methods to Expand a T Regulatory Cell Master Cell Bank
JP2023512670A (en) Cancer immunotherapy using allogeneic, tumor-specific CD4+ T cell injection transplantation
US20220119766A1 (en) Ex vivo generation of gamma delta foxp3+ regulatory t cells and therapeutic uses thereof
WO2018024893A1 (en) Invariant foxp3+ regulatory t cells and therapeutic uses thereof
Vevis et al. Characterization of antigen-binding and MHC class II-bearing T cells with suppressive activity in response to tolerogenic stimulus
JP7390740B2 (en) Composition for the treatment and/or prevention of tumors
Jin et al. Functional and phenotypic properties of peripheral T cells anergized by autologous CD3+ depleted bone marrow cells
CN117529551A (en) Virus-specific immune cells expressing chimeric antigen receptor
Zanon Stem cell-like properties of memory T cells in human immune reconstitution
Polyclonal A Novel Method Using Blinatumomab for

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: 17752074

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017752074

Country of ref document: EP

Effective date: 20190305