US20020164331A1 - Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations - Google Patents

Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations Download PDF

Info

Publication number
US20020164331A1
US20020164331A1 US09/885,768 US88576801A US2002164331A1 US 20020164331 A1 US20020164331 A1 US 20020164331A1 US 88576801 A US88576801 A US 88576801A US 2002164331 A1 US2002164331 A1 US 2002164331A1
Authority
US
United States
Prior art keywords
cells
antibody
cell
cd1d
preferentially binds
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US09/885,768
Other languages
English (en)
Inventor
Mark Exley
Samuel Wilson
Steven Balk
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dana Farber Cancer Institute Inc
Beth Israel Deaconess Medical Center Inc
Original Assignee
Dana Farber Cancer Institute Inc
Beth Israel Deaconess Medical Center Inc
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 Dana Farber Cancer Institute Inc, Beth Israel Deaconess Medical Center Inc filed Critical Dana Farber Cancer Institute Inc
Priority to US09/885,768 priority Critical patent/US20020164331A1/en
Assigned to BETH ISRAEL DEACONESS MEDICAL CENTER reassignment BETH ISRAEL DEACONESS MEDICAL CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EXLEY, MARK A., BALK, STEVEN P.
Assigned to DANA-FARBER CANCER INSTITUTE, INC. reassignment DANA-FARBER CANCER INSTITUTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WILSON, SAMUEL B.
Publication of US20020164331A1 publication Critical patent/US20020164331A1/en
Priority to US11/541,958 priority patent/US8138314B2/en
Assigned to DANA-FARBER CANCER INSTITUTE, THE GENERAL HOSPITAL CORPORATION D/B/A MASSACHUSETTS GENERAL HOSPITAL reassignment DANA-FARBER CANCER INSTITUTE CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNMENT PREVIOUSLY RECORDED ON REEL 012442 FRAME 0307. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNEES ARE "THE GENERAL HOSPITAL CORPORATION AND DANA-FARBER CANCER INSTITUTE," NOT "DANA-FARBER CANCER INSTITUTE". Assignors: WILSON, SAMUEL B.
Priority to US13/423,683 priority patent/US20120258040A1/en
Priority to US14/495,502 priority patent/US20150299320A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • 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
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • 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
    • A61K39/4615Dendritic cells
    • 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
    • A61K39/4622Antigen presenting cells
    • 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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/04Drugs for genital or sexual disorders; Contraceptives for inducing labour or abortion; Uterotonics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, 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
    • 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
    • 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
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • 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
    • A61K2239/26Universal/off- the- shelf cellular immunotherapy; Allogenic cells or means to avoid rejection
    • 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
    • A61K2239/58Prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Modulation of the immune system is desirable to treat a variety of diseases and disorders including, but not limited to, autoimmune diseases, infections, allergies, asthma, inflammatory conditions, spontaneous abortion, pregnancy, graft versus host disease, and cancers.
  • T cells are lymphocytes that participate in multiple cell-mediated immune reactions, such as the recognition and destruction of infected or cancerous cells. Subsets of T cells, such as suppressor, cytotoxic, and helper T cells, mediate different immunologic functions. Suppressor T cells are responsible for turning the immune response off after an infection is cleared. Cytotoxic or “natural killer” T cells destroy infected or cancerous cells. Helper T cells produce cytokines that modulate the activity of cytotoxic T cells and/or antibody-producing B cells.
  • helper T cells secrete interleukin-1 (IL-1), IL-2, gamma interferon (INF- ⁇ ), and IL-2 which enhance cell-mediated responses such as cytotoxic T cell activity and inhibit both Th2 helper T cell activity and humoral immunity mediated by soluble antibodies.
  • IL-1 interleukin-1
  • IL-2 IL-2
  • INF- ⁇ gamma interferon
  • IL-2 IL-2
  • IL-2 gamma interferon
  • T cells may also participate in immune deviation responses, such as the suppression of an ongoing immune response which may involve the secretion of TGF- ⁇ or IL-10 cytokines (Sonoda et al, J. Ex. Med. 190:1215-1255, 1999; Streilein et al., Hum. Immunol. 52:138-143, 1997; Hong et al, J. Ex. Med. 190, 1197-1200,1999; Streilein et al., J. Immunol. 158:3557-3560, 1997).
  • TCR T cell receptor
  • the chains of the most common T cell receptors are called ⁇ and ⁇ .
  • the genes for the ⁇ , ⁇ , ⁇ , and ⁇ chains of the T cell receptors have organizations similar to that of antibody genes: there are libraries of V, D, and J regions from which members are joined to form entire genes.
  • invariant T cells In contrast to most T cell subpopulations, which have diverse sequences for their TCR- ⁇ chain, invariant T cells have a highly conserved invariant TCR- ⁇ chain, V ⁇ 24-J ⁇ Q in humans and V ⁇ 14-J ⁇ 281 in mice, that pairs preferentially with human V ⁇ 11 or murine V ⁇ 8. These cells are either CD4 + CD8 ⁇ or CD4 ⁇ CD8 ⁇ .
  • This invariant TCR is presumed to enable invariant T cells to recognize endogenous or pathogen-derived lipid antigens presented by nonpolymorphic MHC class I-like proteins, called CD1 family members.
  • CD1a Humans have four CD1 proteins (CD1a, CD1b, CD1c, and CD1d), but mice have only a duplicated CD1d gene that is highly homologous to human CD1d.
  • Human CD1d is expressed at high levels by thymocytes, at lower levels by B cells and monocytes, and by some cells outside of the lymphoid and myeloid lineages.
  • invariant T cells are distinguished by expression of several cell surface proteins otherwise found largely on natural killer (NK) cells, including CD161 (NKR-P1A) in humans, and a cell surface C-type lectin, NKR-P1C (NK1), in mice.
  • NK natural killer
  • This T cell subpopulation referred to here as “invariant NK T cells,” represents a major fraction of the mature T cells in thymus, the major T cell subpopulation in murine liver, and up to 5% of splenic T cells in some mouse strains.
  • Murine and human invariant T cells produce large amounts of the immunoregulatory cytokines IL-4 (a Th2 effector) and IFN- ⁇ (a Th1 effector) in vivo in response to an anti-CD3 antibody or CD1d. These cytokines allow the cells to participate in both Th2 and Th1 responses.
  • the role of invariant T cells in augmenting the Th2 response is further supported by the presence of defects in invariant T cells in a number of human and murine models of autoimmune diseases, including type 1 diabetes.
  • alterations in the balance between Th1 and Th2 responses induced by invariant T cells may play a role in the development of autoimmune diseases.
  • invariant NK T cells In addition, data from human patients shows fewer invariant NK T cells and reduced Th1-like responses in patients with advanced cancer.
  • the anti-tumor response of activated invariant T cells could be partially mediated by their CD1d specific cytotoxicity and NK/LAK cell-like toxicity.
  • Other regulatory functions of invariant T cells possibly through cytokine production or interactions with antigen presenting cells (APCs), may also play important roles in anti-tumor immune responses.
  • APCs antigen presenting cells
  • Invariant T cells may also have a role in the pathogenesis of spontaneous abortion. Stimulation of decidual invariant T cells in mice by administration of a ligand for invariant T cells provoked abortion in pregnant mice. The perforin-dependent killing and production of IFN- ⁇ and tumor necrosis factor- ⁇ by the invariant T cells were required for this induction of abortion.
  • CD1d-reactive noninvariant T cells In contrast to human peripheral blood in which invariant T cells are the major CD1d-reactive subpopulation, human and mouse bone marrow and human liver have T cell populations dominated by CD1d-reactive noninvariant T cells using diverse TCRs which can also produce a large amount of IL-4 and IFN- ⁇ . These CD1d-reactive noninvariant T cells can be either NK or non-NK T cells, and they function similarly to CD1d-reactive invariant T cells.
  • the CD1d-reactive noninvariant T cells in bone marrow may have a role in suppressing graft versus host disease, and both populations may enhance graft versus leukemia responses.
  • these T cells may protect against infections, such as Hepatitis C infections, but may also cause damage due to their Th1 response.
  • CD1d-reactive NK T cells are critical for immune tolerance to antigens in the anterior chamber of the eye, an immune privileged site (Sonoda et al., supra). Such mechanisms may also be important in the maintenance of peripheral tolerance.
  • ⁇ -galactosylceramide ( ⁇ -GalCer) lipid which was isolated from marine sponge in a screen for anti-tumor activity, is a CD1d-presented antigen.
  • ⁇ -GalCer is an example of an agent which can be used to expand human CD1d-reactive invariant T cells from umbilical cord or peripheral blood samples that are first enriched for invariant T cells by purification using an anti-V ⁇ 24 antibody.
  • the enriched V ⁇ 24 + cells are cocultured in the presence of ⁇ -GalCer and purified antigen-presenting cells (APCs).
  • APCs antigen-presenting cells
  • the invention features antibodies which recognize and expand T cells having specific TCR sequences. These antibodies may be used for diagnosing, preventing, stabilizing, and treating a variety of diseases including cancers and autoimmune diseases.
  • the invention features a purified antibody that preferentially binds a T cell antigen receptor (TCR).
  • TCR T cell antigen receptor
  • This antibody preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of the TCR; or preferentially binds or modulates (e.g., increases or decreases) the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • the invention features a combination of purified antibodies (e.g., a mixture of 2, 3, 4, or 5 antibodies) that together preferentially bind a T cell antigen receptor (TCR).
  • these antibodies preferentially bind a CDR3-loop or an ⁇ - ⁇ junction of the TCR; or preferentially modulate the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • a desirable antibody combination include a mixture of one of the following pairs of antibodies: (i) an anti-V ⁇ 24 antibody and an anti-CD161 antibody; (ii) an anti-V ⁇ 24 antibody and an anti-CD94 antibody; (iii) an anti-V ⁇ 11 antibody and an anti-CD161 antibody; or (iv) an anti-V ⁇ 11 antibody and an anti-CD94 antibody.
  • these antibody combinations preferentially bind or preferentially modulate the expansion or activation of CD1d-reactive T cells.
  • the antibody or antibody combination preferentially binds an invariant T cell.
  • NK T cells that are bound by the antibody or antibody combination are CD1d-reactive T cells, invariant T cells, CD1d-reactive noninvariant T cells, or J ⁇ Q + T cells.
  • the antibody or antibody combination preferentially binds the antigen binding site of a TCR.
  • the NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells are invariant T cells.
  • the CDR3-loop, ⁇ - ⁇ junction, or antigen binding site of a TCR is expressed on a NK T cell, CD1d-reactive T cell, or J ⁇ Q + T cell.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR expressed on a NK T cell, CD1d-reactive T cell, or J ⁇ Q + T cell, and the antibody or antibody combination preferentially modulates the expansion or activation of the bound T cell.
  • a T cell expressing a TCR that is bound by the antibody or antibody combination is expanded in the presence of the antibody.
  • the binding of the CDR3-loop, ⁇ - ⁇ junction, or antigen binding site of a TCR expressed on a T cell is sufficient to quantify the T cells, the CDR3-loops, ⁇ - ⁇ junctions, or the antigen binding sites.
  • a second antibody is used to distinguish different T cell subpopulations bound by the antibody of the invention.
  • the antibody is a bifunctional antibody.
  • the antibody is a polyclonal or monoclonal antibody.
  • the antibody is covalently linked to a toxin, therapeutically active compound, enzyme, cytokine, radiolabel, fluorescent label, magnetic label, or affinity tag.
  • Desirable antibodies have a constant region found in a mammal other than the mouse, such as a human. Desirably, the antibody is humanized.
  • the invention provides a fragment or derivative of an antibody, wherein the antibody preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of a TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • Desirable fragments include ScFv, Fab, and F(ab′) 2 fragments.
  • the invention provides a bifunctional antibody including (a) an antibody of the invention or a fragment of the antibody, and (b) a second antibody or fragment of an antibody that binds a T cell expressing the TCR or that binds a NK T cell, CD1d-reactive T cell, or J ⁇ Q + T cell that is bound by the antibody or fragment of the invention.
  • the second antibody is also an antibody of the invention.
  • the second antibody is an anti-CD3, anti-CD161, anti-CD28, or anti-CD94 antibody.
  • the invention features a bifunctional antibody that preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of the TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • the antibody is a bifunctional anti-CD3 and anti-CD161 antibody, anti-V ⁇ 24 and anti-CD161 antibody; anti-V ⁇ 24 and anti-CD94 antibody; anti-V ⁇ 11 and anti-CD161 antibody; anti-V ⁇ 11 and anti-CD94 antibody; anti-CD3 and anti-CD94 antibody, anti-CD3 and anti-CD28; or anti-V ⁇ 24 and anti-V ⁇ 11 antibody.
  • the bifunctional antibody preferentially binds, expands, or activates CD1d-reactive T cells.
  • the invention provides a stable hybridoma that produces an antibody of the invention.
  • the antibodies of the invention may be used to purify T cell subpopulations.
  • the invention also features a purified T cell subpopulation.
  • the T cells in the subpopulation are specifically bound by an antibody or antibody combination of the invention.
  • the antibody or antibody combination specifically binds the CDR3-loop, ⁇ - ⁇ junction, or the antigen binding site of a TCR expressed on the subpopulation of T cells.
  • the T cells in the subpopulation are NK T cells.
  • Desirable NK T cells include CD1d-reactive T cells, invariant T cells, CD1d-reactive noninvariant T cells, and J ⁇ Q + T cells.
  • the T cells are CD1d-reactive T cells.
  • the T cells are J ⁇ Q + T cells.
  • the NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells are invariant T cells.
  • the invention also provides methods for generating an antibody of the invention that preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of a TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • One such method includes (a) coupling a cyclic peptide to a carrier, (b) immunizing an animal with the coupled peptide, and (c) isolating an antibody that preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of a TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • step (b) is repeated.
  • the animal is a CD1 or invariant T cell deficient mammal or bird.
  • the invention provides another method of generating an antibody of the invention.
  • This method includes (a) immunizing an animal (e.g., a mammal or a bird) with NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells, and (b) isolating an antibody that preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of a TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • step (a) is repeated.
  • invariant T cells are administered to a CD1 or invariant T cell deficient animal.
  • the invention features a method of generating an antibody of the invention.
  • This method involves (a) coupling a cyclic peptide to a carrier, (b) immunizing an animal (e.g., a mammal or a bird) with the coupled peptide, (c) immunizing the animal with NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells, and (d) isolating an antibody that preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of a TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • step (b) or (c) is repeated.
  • invariant T cells are administered to a CD1 or invariant T cell deficient animal.
  • the invention provides a method of generating an antibody of the invention by (a) immunizing an animal (e.g., a mammal or a bird) with NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells, (b) coupling a cyclic peptide to a carrier, and (c) immunizing the animal with the coupled peptide, and (d) isolating an antibody that preferentially binds a CDR3-loop or an ⁇ - ⁇ junction of a TCR; or preferentially binds or modulates the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • step (a) or step (c) is repeated.
  • invariant T cells are administered to a CD1 or invariant T cell deficient animal.
  • the animal e.g., mammal or bird
  • the animal is a CD1 or invariant T cell deficient animal, a CD1d knockout mouse, an animal in which invariant T cells have been removed, an animal lacking part of the TCR- ⁇ chain, or an animal lacking part of the TCR- ⁇ chain.
  • the animal is tolerized to NK T cells, CD1d-reactive T cells, J ⁇ Q + T cells, invariant T cells, or the invariant TCR.
  • the animal lacks all or part of the V ⁇ 14 or J ⁇ 281 molecule. It is also contemplated that another animal with a reduced amount or lacking NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be used.
  • the antibodies, bifunctional antibodies, fragments of antibodies, and derivatives of antibodies of the present invention have a variety of diagnostic, imaging, and therapeutic applications.
  • the invention provides a method of measuring the amount of NK TCRs or the amount of NK T cells in a sample by contacting the sample with an antibody or antibody combination of the invention.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the TCRs.
  • the antibody is a bifunctional antibody that binds both CD3 and CD161 expressed on the same T cell.
  • the invention features a method of measuring the amount of CD1d-reactive TCRs or the amount of CD1d-reactive T cells in a sample.
  • This method involves contacting the sample with an antibody or antibody combination of the invention.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an ⁇ - ⁇ junction, or an antigen binding site of the TCRs.
  • the antibody is a bifunctional anti-V ⁇ 24 and anti-CD161 antibody; anti-V ⁇ 24 and anti-CD94 antibody; anti-V ⁇ 11 and anti-CD161 antibody; anti-V ⁇ 11 and anti-CD94 antibody; anti-CD3 and anti-CD94 antibody, anti-CD3 and anti-CD28; or anti-V ⁇ 24 and anti-V ⁇ 11 antibody.
  • the invention features a method of measuring the amount of J ⁇ Q + TCRs or the amount of J ⁇ Q + T cells in a sample by contacting the sample with an antibody or antibody combination of the invention.
  • the antibody preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the TCRs.
  • the sample is from a subject involved in a clinical trial of a therapy or undergoing a therapy for a condition selected from the group consisting of autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • a condition selected from the group consisting of autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • the amount of the TCRs or the T cells is used to determine the desirable therapy for the treatment or prevention of a condition selected from the group consisting of autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • a condition selected from the group consisting of autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • the amount of the TCRs or the T cells is used to diagnose the recovery or remission from or the efficacy of any therapy for a condition selected from the group consisting of autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, pregnancy, and cancer.
  • the invention provides a method of visualizing the NK TCRs or the NK T cells in a sample, the method comprising contacting the sample with an antibody or antibody combination of the invention.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the TCRs.
  • the antibody is a bifunctional antibody that binds both CD3 and CD161 expressed on the same T cell.
  • the invention features a method of visualizing the CD1d-reactive TCRs or the CD1d-reactive T cells in a sample by contacting the sample with an antibody or antibody combination of the invention.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the TCRs.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an ⁇ - ⁇ junction, or an antigen binding site of the TCRs.
  • the antibody is a bifunctional anti-V ⁇ 24 and anti-CD161 antibody; anti-V ⁇ 24 and anti-CD94 antibody; anti-V ⁇ 11 and anti-CD161 antibody; anti-V ⁇ 11 and anti-CD94 antibody; anti-CD3 and anti-CD94 antibody, anti-CD3 and anti-CD28; or anti-V ⁇ 24 and anti-V ⁇ 11 antibody.
  • the invention provides a method of visualizing the J ⁇ Q + TCRs or the J ⁇ Q + T cells in a sample.
  • This method includes contacting the sample with an antibody or antibody combination of the invention.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the TCRs.
  • the sample is from a transgenic animal or is an autopsy tissue.
  • the antibody is covalently bound to a fluorescent label, radiolabel, or magnetic label (e.g., a magnetic bead).
  • the invention provides a method of identifying a subject at risk for a condition selected from the group consisting of auto immune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • This method includes (a) quantitating the amount of a subpopulation of T cells in a sample from the subject, and (b) comparing the amount of the T cells in the sample to the amount of the T cells found in subjects diagnosed with the condition or subjects not diagnosed with the condition.
  • the amount of the T cells in the sample is compared to the amount of the T cells found in both subjects diagnosed with the condition and subjects not diagnosed with the condition.
  • the method further includes comparing the amount of another T cell type in the sample with the amount of the another T cell type found in subjects diagnosed with the condition or subjects not diagnosed with the condition.
  • the amount of the another T cell type in the sample is compared to the amount of the another T cell type found in both subjects diagnosed with the condition and subjects not diagnosed with the condition.
  • the amount of the subpopulation of T cells in a sample is determined by contacting the sample with an antibody or antibody combination of the invention, such as an antibody or antibody combination that preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the T cells; or that preferentially binds NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • an antibody or antibody combination of the invention such as an antibody or antibody combination that preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the T cells; or that preferentially binds NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • the invention provides a method of diagnosing or staging a subject with a condition selected from the group consisting of autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • This method includes (a) quantitating the amount of a subpopulation of T cells in a sample from the subject, and (b) comparing the amount of the T cells in the sample to the amount of the T cells found in subjects diagnosed with the condition or subjects not diagnosed with the condition.
  • the amount of the T cells in the sample is compared to the amount of the T cells found in both subjects diagnosed with the condition and subjects not diagnosed with the condition.
  • the method further includes comparing the amount of another T cell type in the sample with the amount of the other T cell type found in subjects diagnosed with the condition or subjects not diagnosed with the condition.
  • the amount of the other T cell type in the sample is compared to the amount of the other T cell type found in both subjects diagnosed with the condition and subjects not diagnosed with the condition.
  • the amount of the subpopulation of T cells in a sample is determined by contacting the sample with an antibody or antibody combination of the invention, such as an antibody or antibody combination that preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the T cells; or that preferentially binds NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • an antibody or antibody combination of the invention such as an antibody or antibody combination that preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of the T cells; or that preferentially binds NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • the sample is peripheral blood, lymphatic fluid, ascitic fluid, umbilical cord blood, urine, fecal matter, bone marrow, bile, or a biopsy sample. It is also contemplated that any other sample from a mammal may be used, such as other blood or tissue samples.
  • the invention features a method of treating or preventing an autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, or cancer in an animal.
  • This method includes administering to the animal an antibody or antibody combination of the invention.
  • the antibody or antibody combination is administered to the animal intraarticularly, intralesionally, orally, intramuscularly, intravenously, subcutaneously, or intraperitoneally.
  • the antibody or antibody combination is administered with a pharmaceutically suitable carrier.
  • a cytokine is also administered to the animal.
  • a cell type or other agent which works in concert with the antibodies is also administered to the animal.
  • the invention provides a method of inhibiting T cell pathogenesis in an animal by administering to the animal an antibody or antibody combination of the invention.
  • the administration of the antibody or antibody combination is sufficient to inhibit a T cell expressing a TCR bound by the antibody or antibody combination, a NK T cell, a CD1d-reactive T cell, or a J ⁇ Q + T cell.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR of a NK T cell, a CD1d-reactive T cell, or a J ⁇ Q + T cell.
  • the antibody or antibody combination inhibits the expansion or activation of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • the antibody or antibody combination is covalently linked to a toxin, a radiolabel, or a molecule which targets host defensive or catabolic processes toward the cells.
  • the antibody or antibody combination is administered to the animal orally, intramuscularly, intravenously, intraarticularly, intralesionally, subcutaneously, intraperitoneally, intralesionally, or by any other route sufficient to provide a dose adequate to inhibit the T cell.
  • the antibody or antibody combination is administered with a pharmaceutically suitable carrier.
  • one or more cytokines are also administered to the animal.
  • the T cell pathogenesis is a response of a T cell to a viral infection, such as a Hepatitis infection, picornarirus infection, polio infection, or coxsacchie infection.
  • the invention provides a method of increasing the size of a subpopulation of T cells by contacting a sample having T cells with an antibody or antibody combination of the invention.
  • the invention features a method of increasing the size of a subpopulation of T cells.
  • This method includes (a) contacting a sample comprising the T cells with an antibody or antibody combination of the invention under conditions that allow complex formation between the T cells and the antibody or antibody combination, (b) isolating the complex, and (c) contacting the T cells in the complex or recovered from the complex with an antibody or antibody combination of the invention under conditions that allow the contacting to increase the number of the T cells.
  • the invention provides a method of increasing the size of a subpopulation of T cells.
  • This method includes (a) contacting a sample comprising the T cells with an antibody or antibody combination of the invention under conditions that allow complex formation between the T cells and the antibody or antibody combination, (b) isolating the complex, and (c) contacting the T cells in the complex or recovered from the complex with an antigen and antigen presenting cells under conditions that allow the contacting to increase the number of the T cells.
  • the antigen is ⁇ -galactosylceramide, a lipid or glycosyl-phosphatidylinositol antigen from an infectious pathogen, an antigen from a cancerous cell, a self-lipid, or any other antigen from endogenous or non-physiological sources.
  • the invention features a method of increasing the size of a subpopulation of T cells.
  • This method includes (a) purifying the T cells from a sample comprising the T cells and (b) contacting the purified cells with an antibody or antibody combination of the invention under conditions that allow the contacting to increase the number of the T cells.
  • the method further includes contacting the sample or the complex with one or more cytokines or other cells that work in concert with the antibody or antibody combination.
  • the invention provides a method of increasing the size of a subpopulation of T cells in an animal.
  • This method includes (a) obtaining a sample comprising the T cells from the animal, (b) contacting the T cells with an antibody or antibody combination of the invention under conditions that allow the contacting to increase the number of the T cells, and (c) administering the contacted T cells to the animal.
  • the invention provides a method of increasing the size of a subpopulation of T cells in an animal.
  • This method includes (a) obtaining a sample comprising the T cells from the animal, (b) purifying the T cells, (c) contacting the T cells with an antibody or antibody combination of the invention under conditions that allow the contacting to increase the number the T cells, and (d) administering the contacted T cells to the animal.
  • the purification of the T cells includes contacting the sample with an antibody or antibody combination of the invention.
  • the purification of the T cells includes contacting the sample with an anti-V ⁇ 24, CD4, CD8, CD56, CD161, or V ⁇ 11 antibody. It is also contemplated that another antibody or combination of antibodies that binds a T cell may be used to purify the cells.
  • the invention provides a method of increasing the size of a subpopulation of T cells in an animal.
  • This method includes (a) obtaining a sample comprising the T cells from the animal, (b) contacting the T cells with an antibody or antibody combination of the invention under conditions that allow complex formation between the T cells and the antibody, (c) isolating the complex, and (d) contacting the T cells in the complex or recovered from the complex with an antibody or antibody combination of the invention under conditions that allow the contacting to increase the number of the T cells, and (e) administering the contacted T cells to the animal.
  • the invention features a method of increasing the size of a subpopulation of T cells in an animal.
  • This method includes (a) obtaining a sample comprising the T cells from the animal, (b) contacting the T cells with an antibody or antibody combination of the invention under conditions that allow complex formation between the T cells and the antibody or antibody combination, (c) isolating the complex, and (d) contacting the T cells in the complex or recovered from the complex with an antigen and antigen presenting cells under conditions that allow the contacting to increase the number of the T cells, and (e) administering the contacted T cells to the animal.
  • the antigen is ⁇ -galactosylceramide, a lipid or glycosyl-phosphatidylinositol antigen from an infectious pathogen, an antigen from a cancerous cell, a self-lipid, or any other antigen from endogenous or non-physiological sources.
  • the methods also include administering one or more cytokines to the animal.
  • the cytokine is administered to the animal before, during, or after the contacted T cells are administered to the animal.
  • the cytokine is administered intramuscularly, intravenously, intraarticularly, intralesionally, subcutaneously, or by any other route sufficient to provide a dose adequate to modulate the activity of a T cell.
  • the sample or the T cells are contacted with one or more cytokines.
  • the methods of these aspects are used in the treatment or prevention of an autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, or cancer in the animal.
  • the invention provides a method of purifying a subpopulation of T cells from a sample by contacting the sample with an antibody or antibody combination of the invention.
  • the invention provides a method of purifying a subpopulation of T cells from a sample.
  • This method includes (a) contacting the sample with an antibody or antibody combination of the invention under conditions that allow complex formation between the T cells and the antibody, and (b) isolating the complex.
  • the sample is also contacted with an anti-V ⁇ 24, CD4, CD8, CD56, CD161, or V ⁇ 11 antibody.
  • the sample is contacted with any other antibody that binds a related T cell subset.
  • the method also includes recovering the T cells from the complex.
  • the antibody is covalently linked to a fluorescent label, and the complex is isolated based on the fluorescence signal of the complex.
  • the antibody is covalently linked to a magnetic label, and the complex is isolated based on the magnetism of the complex.
  • the cytokine that is contacted with the sample, T cells, or complex or the cytokine that is administered to the animal is selected from the group consisting of IL-2, IL-4, IL-7, IL-10, IL-12, IL-13, IL-15, IL-18, IFN- ⁇ / ⁇ , IFN- ⁇ , and GM-CSF. It is also contemplated that any other cytokine or combination of cytokines may be used. Desirably, the cytokine alters the ratio of Th1/Th2/immune deviation response by the contacted T cells. Desirably, the subject or the animal is a human.
  • the viral infection relevant to the methods of the invention is a Hepatitis infection, picornarirus infection, polio infection, HIV infection, or coxsacchie infection.
  • the autoimmune disease is type 1 diabetes.
  • the antibody or antibody combination preferentially binds an invariant T cell.
  • the T cell subpopulation, T cells, or T cell expressing the TCR are NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • Desirable NK T cells are CD1d-reactive T cells, invariant T cells, CD1d-reactive noninvariant T cells, or J ⁇ Q + T cells.
  • Desirable CD1d-reactive T cells or J ⁇ Q + T cells are invariant T cells.
  • Desirable J ⁇ Q + T cells are V ⁇ 24 + J ⁇ Q + T cells.
  • the antibody or antibody combination preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR expressed on the T cell and preferentially binds or modulates the expansion or activation of the bound T cell.
  • the antibody or antibody combination preferentially binds or modulates the expansion or activation of only one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR bound by an antibody or antibody combination of the invention is expressed by a NK T cell, CD1d-reactive T cell, J ⁇ Q + T cell, or invariant T cell.
  • an antibody that binds or modulates the expansion or activation a T cell subpopulation of interest e.g., NK T cells, CD1d-reactive T cells, J ⁇ Q + T cells, or invariant T cells
  • preferentially binds the antigen binding site of the TCR of the T cell subpopulation of interest preferentially binds the antigen binding site of the TCR of the T cell subpopulation of interest.
  • the invariant T cells relevant to any of the aspects of the invention may not be CD1d-reactive.
  • Desirable labeled antibodies include antibodies bound to biotin, FITC, PE, or a magnetic bead.
  • each of the aspects of the invention apply equally to the antibodies, bifunctional antibodies, fragments of antibodies, and derivatives of antibodies of the invention.
  • Each of the aspects of the invention also apply equally to a combination of antibodies that together preferentially bind a CDR3-loop or an ⁇ - ⁇ junction of the TCR.
  • the antibodies in an antibody combination of the invention are simultaneously or sequentially contacted with NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells or with a sample containing NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • the antibodies in an antibody combination of the invention are simultaneously or sequentially administered to an animal for the treatment or prevention of a disease or condition.
  • Each aspect of the invention also applies to any antigen-specific oligoclonally expanded T cell subpopulation encountered in animals (e.g., humans, other mammals, and birds) in response to a given antigenic challenge.
  • These antigen-specific oligoclonally expanded T cell subpopulations include those T cells oligoclonally expanded in response to an immunodominant component of the antigen in multiple individuals.
  • an antigen presenting cell APC
  • a soluble or immobilized form of an antigen-presenting molecule may be used to present an antigen to a T cell or a sample containing a T cell of interest under conditions that allow the activation or expansion of the T cell without the presence of an APC.
  • CDR3 loop is meant the amino acids in the junction that is generated by rearrangement between a V segment and a J segment of a TCR- ⁇ chain or by rearrangement between a V segment, a D segment, and a J segment of a TCR- ⁇ chain. Identification of the CDR3 loop is simplified by the presence of a conserved cysteine at the end of the V segment. The first amino acid after this cysteine is the first residue of the CDR3 loop.
  • the sequence of a CDR3 loop can be readily identified based on a sequence alignment of the amino acid sequence of a TCR of interest with one or more sequences of other TCRs.
  • a Kabat table which contains an alignment of the amino acid sequence of numerous T cell receptors may be used to identify the CDR3 loop in a TCR- ⁇ chain or - ⁇ chain of interest (Johnson and Wu, Nuc. Acid. Res. 29(1):205-206, 2001; http://immuno.bme.nwu.edu).
  • antibody that preferentially binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR is meant an antibody which recognizes and binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR or which recognizes and binds a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR expressed on a T cell, but which does not substantially recognize and bind other molecules in a sample, e.g., a biological sample, which naturally includes other protein or cells.
  • the signal in the ELISA assay described in Example 1 for the binding of the antibody to a CDR3-loop, an antigen binding site, or an ⁇ - ⁇ junction of a TCR expressed on a T cell is desirably at least 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, or 500 times greater than that for the binding to a control cell that is not a T cell or to a T cell that expresses TCRs with CDR3-loops, ⁇ - ⁇ junctions, and antigen binding sites with amino acid sequences that are less than 99, 95, 90, 85, 80, 70, 60, 50, 40, or 20% identical to the corresponding sequence of the CDR3-loop, ⁇ - ⁇ junction, or antigen binding site preferentially bound by the antibody.
  • Humanized or other species forms of the antibody may be generated using standard techniques.
  • the antibody is “purified,” meaning it has been separated from other components that naturally accompany it.
  • the antibody is substantially pure when it is at least 50%, by weight, free from proteins, antibodies, and naturally-occurring organic molecules with which it is naturally associated.
  • the antibody is at least 75%, more desirably, at least 90%, and most desirably, at least 99%, by weight, pure.
  • a substantially pure antibody that preferentially binds a CDR3-loop, ⁇ - ⁇ junction, or an antigen binding site of a TCR may be obtained, for example, by using a method of the present invention to immunize a mammal for the generation of the antibody, by construction of hybridoma secreting the antibody, by chemically synthesizing the antibody, or by separation of the antibody from natural sources. Purity can be assayed by any appropriate method, as described below for the isolation of antibodies.
  • ⁇ - ⁇ junction of a TCR is meant the interface between the ⁇ and ⁇ chains of a TCR.
  • the interface includes noncovalent interactions between the variable domains of the ⁇ and ⁇ chains.
  • the three-dimensional structure of the ⁇ - ⁇ junction or a model of the three-dimensional structure of the ⁇ - ⁇ junction is used in the design of polypeptides or small molecules for immunizing a mammal to generate an antibody to the ⁇ - ⁇ junction of the TCR.
  • a small molecule may be designed that has a three-dimensional structure similar to that of an exposed region of the interface.
  • a polypeptide may be designed that has a three-dimensional structure similar to that of a portion or all of the ⁇ - ⁇ junction of a TCR.
  • a modeled or actual structure of a TCR of interest may be used to obtain a modeled or actual structure of the ⁇ - ⁇ junction of the TCR which can then be mimicked by a designed polypeptide (e.g., a polypeptide of approximately 100 amino acids). Covalent bonds or linkers may be added between the domains of the designed polypeptide so that a single chain polypeptide is generated.
  • a designed polypeptide e.g., a polypeptide of approximately 100 amino acids.
  • Covalent bonds or linkers may be added between the domains of the designed polypeptide so that a single chain polypeptide is generated.
  • any standard modeling program such as MolScript, may be used.
  • antigen binding site of a TCR is meant the region of a TCR that binds an antigen. This region includes part of the exposed surface of the variable region of the TCR.
  • the antigen binding site contains the CDR3-loop of the a chain, the CDR3-loop of the ⁇ chain, a predictable portion of the CDR1-loops, a predictable portion of the CDR2-loops, and some nearby structural surfaces.
  • the three-dimensional structure of the antigen binding site or a model of the three-dimensional structure of the antigen binding site is used in the design of polypeptides or small molecules for immunizing a mammal to generate an antibody to the antigen binding site of the TCR.
  • Several three-dimensional structures of antigen binding sites of TCRs have been reported (in the presence or absence of an antigen) and may be used by one skilled in the art to model the antigen binding site of a TCR expressed by a NK, CD1d-reactive, J ⁇ Q + , or invariant T cell. Additionally, a modeled or actual structure of CD1d could be used to determine how other compounds would interact with CD1d-reactive T cells.
  • a small molecule or polypeptide may be designed that has a three-dimensional structure similar to that of a portion or all of the antigen binding site.
  • a modeled or actual structure of a TCR of interest may be used to obtain a modeled or actual structure of the antigen binding site of the TCR which can then be mimicked by a designed polypeptide (e.g., a polypeptide of approximately 100 amino acids). Covalent bonds or linkers may be added between the domains of the designed polypeptide so that a single chain polypeptide is generated.
  • any standard modeling program such as MolScript, may be used.
  • the human CD1d-reactive invariant T cell antigen receptor recognizes CD1d, but not the closely related CD1a, CD1b, or CD1c family members (Exley et al., J. Exp. Med. 186(1):109-120, 1997).
  • fragment is meant a polypeptide having a region of consecutive amino acids that is identical to the corresponding region of an antibody of the invention.
  • the fragment has the ability to bind, activate, and/or expand T cells ex vivo or in vivo, as determined using the assays described herein.
  • the number, activity, or purity of the expanded cells is at least 20, 40, 60, 80, or 90% of that produced by an antibody of the invention, as measured using the assays provided herein.
  • the binding of the fragment to the CDR3-loop, ⁇ - ⁇ junction, or antigen binding site of a TCR is at least 20, 40, 60, 80, or 90% of that of an antibody of the invention.
  • derivative is meant an antibody or fragment of the invention that is modified chemically or through gene fusion technology or chemical synthesis so that it is covalently linked to a toxin, therapeutically active compound, enzyme, cytokine, radiolabel, fluorescent label, or affinity tag.
  • the covalently linked group can be attached to the amino terminus, carboxy terminus, between the amino and carboxy termini, or to a side chain of an amino acid in the antibody or fragment.
  • affinity tag is meant a peptide, protein, or compound that binds another peptide, protein, or compound. In a desirable embodiment, the affinity tag is used for purification or immobilization of the derivative.
  • the affinity tag or toxin is used to target the antibody or fragment to a specific cell, tissue, or organ system in vivo.
  • the fluorescent or radiolabel is used for imaging of the derivative.
  • the therapeutically active compound or radiolabel is used for the treatment or prevention of a disease or disorder.
  • the derivative or fragment of an antibody of the invention has increased stability or increased solubility compared to the antibody. It is also contemplated that the antibody, fragment, or derivative of the invention may be bound non-covalently to another antibody covalently linked to a toxin, therapeutically active compound, enzyme, cytokine, radiolabel, fluorescent label, magnetic label, or affinity tag.
  • humanized is meant alteration of the amino acid sequence of an antibody so that fewer antibodies and/or immune responses are elicited against the humanized antibody when it is administered to a human.
  • an antibody of the invention may be converted to that species format.
  • bifunctional antibody is meant an antibody that includes an antibody or a fragment of an antibody covalently linked to another antibody or another fragment of an antibody. Desirably, both antibodies or fragments bind to different epitopes expressed on the same T cell. Desirably, at least one antibody included in the bifunctional antibody is an antibody of the invention. Desirably, the antibody binds CD3, CD161, or both CD3 and CD161. In other desirable embodiments, the antibody binds one or more of the following: V ⁇ 24, CD94, V ⁇ 11, and anti-CD28.
  • cyclic peptides is meant a non-linear peptide having an amino acid sequence at least 60%, desirably 80%, more desirably 90%, and most desirably 100% identical to a region in the CDR3 loop of a TCR.
  • the amino acid sequence of the peptide includes CVVSDRGSTLGRLADCG (SEQ ID NO 1) from the CDR3-loop of the human invariant TCR- ⁇ or a region of at least 5, 8, 10 , or 15 consecutive amino acids of SEQ ID NO 1.
  • the amino acid sequence of the peptide is identical to that of SEQ ID NO 1.
  • the peptide may be cyclized by the formation of a covalent bond between the N-terminal amino group of the peptide or the side-chain of residue and the C-terminal carboxyl group or the side-chain of a residue.
  • a peptide lactam may be formed by the cyclization between the N-terminal amino group or an amino group of an amino acid side-chain and the C-terminal carboxyl group or a carboxyl or amide containing side-chain, such as that of glutamic, aspartic acid, glutamine, or asparagine.
  • cyclizations include the formation of a thioether by the reaction of a thiol group in a cysteine side-chain with the N-terminal amino group, C-terminal carboxyl group, or the side-chain of another amino acid.
  • a disulfide bond may also be formed between two cysteine residues.
  • a peptide having an amino acid sequence at least 60%, desirably 80%, more desirably 90%, and most desirably 100% identical to a region of another loop or exposed surface of the alpha or beta chain of the invariant TCR or another TCR may be used for the generation of antibodies to another TCR.
  • Desirable loops include the CDR3-loop of the ⁇ or ⁇ chain of a NK T cell, CD1d-reactive T cell, or J ⁇ Q + T cell.
  • CD1d or invariant T cell deficient mammal is meant a mammal that when compared to other mammals of the same species has a reduced amount of or lacks functional CD1d molecules, invariant T cells, TCR- ⁇ chains, or TCR- ⁇ chains.
  • Desirable examples of such mammals include a CD1d knockout mouse (Sonoda et al., 1999, supra), a mammal not tolerized to the invariant TCR, a mammal in which invariant T cells have been removed, a mammal lacking part of the TCR- ⁇ chain (Cui et al., Science 278:1623, 1997), mammal lacking part of the VB8 molecule, or animals deleted of invariant, NK T cells, or related subpopulations by antibodies, cytokines, or repeated antigenic stimulation.
  • a CD1d knockout mouse Nonoda et al., 1999, supra
  • a mammal not tolerized to the invariant TCR a mammal in which invariant T cells have been removed
  • a mammal lacking part of the TCR- ⁇ chain Cui et al., Science 278:1623, 1997)
  • mammal lacking part of the VB8 molecule or animals deleted of invariant
  • immunizing is meant administering to an animal (e.g., a mammal or a bird) either peptides coupled to carriers, invariant T cells, or both using standard procedures.
  • Desirable routes of administration include intraperitoneally, intramuscular, intradermal, and subcutaneous. The dose and frequency of administration can be determined using standard procedures.
  • isolated antibodies purifying the antibody from antiserum, ascites fluid, or hybridomas supernatant.
  • the antibodies can be purified by one skilled in the art using standard techniques such as those described by Ausubel et al. (Current Protocols in Molecular Biology, volume 2, p. 11.13.1-11.13.3, John Wiley & Sons, 1995).
  • the antibody is desirably at least 2, 5, or 10 times as pure as the starting material, as measured using polyacrylamide gel electrophoresis, column chromatography, optical density, HPLC analysis, or western analysis to detect a reduction in the amount of contaminating proteins or ELISA to detect an increase in specific activity for binding to markers for a particular T cell subpopulation.
  • contacting is meant incubating a sample with an antibody or cytokine.
  • the antibody or cytokine can be in a soluble form, or it can be immobilized.
  • the immobilized antibody or cytokine is tightly bound or covalently linked to a bead or plate.
  • the contact occurs ex vivo or in vivo.
  • the T cells are NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • treating or preventing is meant administering an antibody of the present invention or T cells that have been incubated with the antibody to a mammal, as described herein.
  • T cell pathogenesis is meant a disease or disorder that is caused or exacerbated by an activity of T cells, such as their cytokine production or cytotoxicity.
  • autoimmune disease is meant a disease in which an immune system response is generated against self epitopes.
  • Some examples of autoimmune diseases include insulin dependent diabetes mellitus, rheumatoid arthritis, pemphigus vulgaris, multiple sclerosis, and myasthenia gravis.
  • increasing size of a subpopulation of T cells is meant stimulating the expansion of these cells by incubating them with an antibody or incubating them with an antigen and antigen presenting cells.
  • the number of T cells belonging to the subpopulation that are present after this incubation is at least 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, 70, 90, 150, or 500 fold greater than the number of these cells present after the corresponding control incubation in the absence of the antibody or the antigen and antigen presenting cells.
  • the number of T cells belonging to the subpopulation that are present after this incubation is at least 2, 5, 10, 20, or 50 fold greater than the number of these cells present after the corresponding incubation in the presence of 0.1-2 ⁇ g/ml phytohemagglutinin (PHA). It is also contemplated that the percentage may remain the same but the actual numbers of the relevant subset may increase if the total number of T cells increases.
  • the change in the percentage of cells that belong to the subpopulation of T cells is at least 2, 5, 10, 20, or 50 fold greater than corresponding change in the percentage of cells that belong to the subpopulation of T cells in a control sample that has not be incubated with the antibody.
  • the T cells are NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells is meant inducing or inhibiting the expansion or activation of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • the induction of the expansion of these T cell subpopulations may be measured as described for determining the increase in the size of the subpopulation of T cells.
  • the inhibition of the expansion of these T cell subpopulations may be determined by comparing the number of T cells belonging to the subpopulation after incubation with an antibody of the invention compared to a control incubation without the antibody.
  • the number of T cells belonging to the subpopulation present after incubation with the antibody is 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, or 100 fold less than the number of these T cells present after the corresponding control incubation.
  • This inhibition of T cell expansion may be useful in the prevention or treatment of T cell pathogenesis.
  • the induction or inhibition of the activation of a T cell subpopulation may be assayed using standard procedures to measure the cytokine production or cytotoxicity of the T cell subpopulation.
  • the increase or decrease in the cytokine production or cytotoxicity is at least 5, 10, 20, 20, 40, 50, 70, 90, or 100% of the activity of the control T cell subpopulation incubated in the absence of the antibody.
  • the change in the size or activity of at least one T cell subpopulation selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells is least 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, 70, 90, 150, or 500 times greater that the corresponding change in cells other than NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • the antibody preferentially binds or modulates the expansion or activation of only one of the T cell subpopulations selected from the group of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells.
  • anti-V ⁇ 24, CD4, CD8, CD56, CD161, CD94, CD28, or V ⁇ 11 antibody is meant an antibody that recognizes and binds V ⁇ 24, CD4, CD8, CD56, CD161, CD94, CD28, or V ⁇ 11 molecules or cells expressing one of these molecules, but which does not substantially recognize and bind other molecules in a sample, e.g., a biological sample, which naturally includes other protein or cells.
  • obtaining a sample comprising T cells is meant removing a sample that has T cells from a mammal or acquiring a sample that has these cells and is produced by a mammal.
  • the sample is peripheral blood.
  • the sample is a bodily fluid, such a urine, bile, or a bodily tissue.
  • the sample is a bone marrow or an umbilical cord sample.
  • purifying the T cells is meant isolating the cells from a sample that naturally contains other cells.
  • the T cells can be purified using an antibody of the invention or an anti-V ⁇ 24, CD4, CD8, CD56, CD161, or V ⁇ 11 antibody in standard procedures. Desirable methods of purification include fluorescence-activated cell sorting (FACS), immunoprecipitation, immunoaffinity chromatography, magnetic bead immunoaffinity purification, and cell panning with a plate-bound antibody.
  • FACS fluorescence-activated cell sorting
  • the purified T cells are at least 2, 5, 10, 50, 100, 500, or 900 times as pure as the original sample, as measured using ELISA or FACS analysis to detect binding of the purified T cells to markers for the T cell subpopulation in which they belong.
  • purified T cell subpopulation is meant a substantially pure T cell subpopulation that is isolated from a sample that naturally contains other cells.
  • the purified T cell subpopulation is enriched for at least one of the following: NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • the T cell subpopulation is enriched for only one of the following: NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • the purified T cell subpopulation is more pure than the purity of the T cell subpopulation found in nature.
  • the purified T cell subpopulation is at least 1, 5, 15, 30, 50, 75, 90, or 99%, by number, free from cells with which it is naturally associated.
  • these other cells that are associated with the T cell subpopulation differ from the T cells belonging to the subpopulation by not expressing a cell-surface molecule, not binding a ligand, or not having an activity of the T cell subpopulation.
  • complex an antibody-bound T cell in which the binding of the antibody to the T cell is sufficient to enable the isolation or separation of the complex from a sample.
  • FIG. 1 is a schematic illustration of the relationship between NK T cells, CD1d-reactive invariant T cells, CD1d-reactive noninvariant T cells, and J ⁇ Q + T cells.
  • FIG. 2A is a histogram of the staining of a control T-cell clone with the anti-CDR3-loop monoclonal antibodies (3A6 or 6B11) or a 6B11 isotype (IgG1) matched control.
  • FIG. 2B is a histogram of the corresponding staining of an invariant NK T cell clone.
  • FIGS. 3A and 3B are dot plots showing the FACS analysis of the recognition of V ⁇ 24 + T cells by the 6B11 monoclonal antibody.
  • FACS purified V ⁇ 24 + T cells were expanded ex viva for 2 weeks with PHA and analyzed using V ⁇ 24-PE and V ⁇ 11-FITC (FIG. 3A) or V ⁇ 24-PE and 6B11-FITC monoclonal antibodies (FIG. 3B).
  • FIGS. 4A and 4B are graphs showing the expansion of V ⁇ 24 sorted cells that were cultured for three weeks with plate bound 6B11 anti-CDR3-loop monoclonal antibody or PHA, respectively.
  • FIGS. 5A and 5B are graphs showing the FACS analysis of invariant NK T cells in a patient before and during IL-12 treatment, respectively.
  • FIGS. 6 A- 6 C are graphs showing IL-4 production by bulk peripheral blood mononuclear cells, V ⁇ 24 + , and polyclonal (‘poly.’) invariant T cells, respectively. These cells were expanded with ⁇ -GalCer and then stimulated with either buffer, plate bound 6B11 antibody, co-cultured with Hela/CD1d transfectants, or PHA. The amount of secreted IL-4 was determined using standard ELISA analysis.
  • FIGS. 7 A- 7 C are graphs showing the comparable expansion of phenotypically and functionally identical invariant NK T cells from the same healthy donor sample by 6B11 and ⁇ -GalCer.
  • V ⁇ 24 ‘MoFlo’ sorted cells were expanded with ⁇ -GalCer pulsed autologous dendritic cells, 6B11 monoclonal antibody, or mitogen (PHA) for an approximately four week expansion with IL-2 alone.
  • FIG. 8A is a graph showing the recognition of bulk T cells from a peripheral blood sample by the 3A6 antibody.
  • FIG. 8D is a graph of the forward (FSC) and side (SSC) scatter of cells showing the gating for the lymphoid cells that were analyzed using the 6B11, 3A6, or control antibody.
  • FIGS. 8B, 8C, and 8 E- 8 I are graphs showing the recognition of invariant NK T cell clones and lines by the 3A6, 6B11, or isotype control antibodies. These invariant NK T cell clones and lines were purified using the anti-V ⁇ 24 monoclonal antibody followed by expansion using alpha-GalCer. For FIGS.
  • FIG. 9 is a graph showing the FACS analysis of invariant NK T cell lines from whole peripheral blood mononuclear cells (PBMC) of a healthy donor. These cells were sorted with 6B11-FITC by MoFlo FACS and expanded for about six weeks prior to 2-color FACS analysis. After expansion, 21% of the cells were V ⁇ 24 + , compared to an original ⁇ 0.1%. These results are also summarized in FIGS. 12 - 17 .
  • FIGS. 10 A- 10 H are graphs showing the FACs analysis of invariant NK T cell lines from 6B11 antibody magnetic bead sorting.
  • Invariant NK T cells from whole PBMC of a healthy donor were sorted with 6B11 by Dynal or Miltenyi magnetic bead immunoaffinity purification from whole PBMC and expanded for about six weeks prior to 2-color FACS analysis. These results are also summarized in FIGS. 12 - 17 .
  • FIGS. 11 A- 11 F are graphs showing the FACs analysis of invariant NK T cell lines from 6B11 antibody magnetic bead sorting.
  • Invariant NK T cells from whole PBMC of a healthy donor were sorted with 6B11 by Dynal magnetic bead immunoaffinity purification and treated as shown from whole PBMC and expanded for about six weeks prior to 2-color FACS analysis. These results are also summarized in FIGS. 12 - 17 .
  • FIG. 12 is a table showing the percent of T cells that stain with the indicated antibodies after a single round four to eight week expansion in T cell medium.
  • the “PBMC” column represents PBMC cells that were not sorted prior to expansion.
  • the “Dynal @ 40” column represents PBMC cells that were purified using 6B11-Dynal beads prior to expansion.
  • the “Milenyi @ 20” column represents PBMC cells that were sorted using 6B11 (20 ⁇ g/mL coating)-Miltenyi beads prior to expansion. For comparison, the corresponding results for unsorted whole leukopak 21 (LKP 21) cells are listed.
  • a standard T cell medium such as RPMI-1640 or Bio Whittaker medium with 10% FBS and 100 U/mL human recombinant IL-2 was used.
  • FIG. 13 is a table showing the percent of T cells that stain with the indicated antibodies after purification using 6B11-Dynal beads and a single round four to eight week expansion in T cell medium. For comparison, the corresponding results for unsorted whole leukopak 10 (LKP 10) cells are listed. As indicated in the table, some of the T cell expansions were conducted in the presence of T cell medium (“TCM”) containing IL-7 at 10 ng/ml, IL-5 at 10 ng/ml, dexamethasone (“Dex”) at 1 ⁇ M, and/or 6B11 antibody at 40 ⁇ g/mL. The figure numbers listed to the right some of the rows in the table indicate figures illustrate the results from the T cell expansions.
  • TCM T cell medium
  • FIG. 14 is a table showing the percent of T cells that stain with the indicated antibodies after a single round four to eight week expansion in T cell medium. For comparison, the corresponding results for unsorted whole leukopak 14 (LKP 14) cells are listed. The results for cells that were sorted with 6B11-Dynal beads prior to expansion are compared to unsorted control cells. As indicated in the table, some of the T cell expansions were conducted in the presence of 6B11 antibody as above, PHA (1 ⁇ g/mL), and/or equivalent numbers of autologous APCs.
  • FIG. 15 is a table showing the percent of T cells that stain with the indicated antibodies after a single round four to eight week expansion in T cell medium. The results for cells that were sorted with 6B11-Dynal beads prior to expansion are compared to unsorted control cells (LKP 13). Some of the T cell expansions were conducted in the presence of 6B11 antibody, PHA, allogenic APCs, and/or autologous APCs, as described above.
  • FIG. 16 is a table showing the percent of T cells that stain with the indicated antibodies after purification using 6B11-Miltenyi magnetic beads and a single round four to eight week expansion in T cell medium compared to unsorted control cells (LKP 2). These T cell expansions were conducted in the presence of autologous APCs, as described above. During one of the T cell expansions, 10 ng/ml IL-7 and IL-15 were also present.
  • FIG. 17 is another table showing the percent of T cells that stain with the indicated antibodies after purification using 6B11-Miltenyi magnetic beads and a single round four to eight week expansion in T cell medium. These T cell expansions were conducted in the presence of autologous APCs and PHA and are compared to unsorted control cells (LKP 11, 12). During the T cell expansions, 6B11 antibody, and/or V ⁇ 24 antibody were also present as described above and as indicated in the table. During one of the T cell expansions, 10 ng/ml IL-7 and IL-15 were also present. The figure numbers listed to the left some of the rows in the table indicate figures illustrate the results from the T cell expansions.
  • FIG. 18 is graph showing the FACs analysis of a T cell line purified using the biotinylated 6B11 antibody and streptavidin conjugated magnetic beads.
  • FIGS. 20A and 20B are graphs showing the two color flow cytometry analysis with V ⁇ 24-PE and V ⁇ 11-FITC monoclonal antibody of V ⁇ 24 sorted cells after a three week expansion with PHA (FIG. 20A) or ⁇ -GalCer and autologous PBMC feeders (FIG. 20B).
  • FIGS. 20C and 20D are bar graphs showing the production of IL-4 and IFN- ⁇ , respectively, by an invariant NK T cell line stimulated PHA or stimulated with mock transfected or CD1d transfected C1R human EBV-transformed B cells in the presence of either an anti-CD1d monoclonal antibody (51.1, 10 ⁇ g/ml) or an isotype control antibody.
  • the results in FIGS. 20 A- 20 D are representative of multiple healthy donors, as shown below.
  • FIGS. 21 A- 21 C are graphs showing the decreased expansion of invariant NK T cells from prostate cancer patients versus healthy donors.
  • ⁇ -GalCer expanded invariant NK T cells from a healthy donor FIG. 21A
  • advanced prostate cancer patient FIG. 21B
  • prostate cancer patient in remission FIG. 21 C
  • FIG. 21D is a bar graph of the percentage of V ⁇ 24 + V ⁇ 11 + cells (mean and standard deviation) after ⁇ -GalCer expansion from a series ( ⁇ 6) of donors.
  • FIGS. 22 A- 22 E are graphs showing the loss of IFN- ⁇ responses by invariant NK T cells from prostate cancer patients.
  • FIGS. 22A and 22B show the IL-4 and IFN- ⁇ production, respectively, from a prostate cancer patient versus a healthy donor derived invariant NK T cell line.
  • FIGS. 22C and 22D are graphs summarizing the cytokine production results by CD1d stimulated invariant NK T cells and PHA stimulated bulk T cells, respectively from a series of advanced prostate cancer patients (•) and healthy donors ( ⁇ ).
  • 22E is a graph of IFN- ⁇ /IL-4 ratios (mean plus standard deviation) for invariant NK T cells (top) or conventional bulk T cells (bottom) from healthy donors and advanced prostate cancer patients.
  • “Cancer+IL-12” are samples from advanced prostate cancer patients treated in vitro with IL-12.
  • FIG. 23 is a table illustrating the use of the 6B11 antibody to determine the frequence of CD1d-restricted T cells in HIV patients.
  • FIG. 24 is a table illustrating the use of the 6B11 antibody and the anti-V ⁇ 24 antibody to determine the frequence of CD1d-restricted T cells in diabetic patients.
  • FIG. 25A is a table illustrating the genes that are differentially expressed between NK T cell clones ME10 and GW4. Genes populating the six expression clusters for the eleven gene functional categories are listed. Each gene is identified by GenBank accession number (or TIGR identifier for HT designations), followed by a common name and the specific cluster into which it fell (row, column).
  • FIG. 25B is a schematic illustration of a model for identified transcripts whose discordant expression is in accord with observed cell phenotypes. Pictured in cartoon format are genes whose transcripts have clearly defined cellular roles and whose discordant expression between GW4 and ME10 correlated with the established phenotypic differences.
  • the genes are related by either being downstream of PI-3 kinase, particularly several genes that regulate cell survival or are directly required for calcium flux and calcium-regulated gene transcription. Average copies of mRNA molecule/million for genes that were significantly altered by anti-CD3 treatment for GW4 (resting, activated) and ME10 (resting, activated) respectively were: Itk, (49, 115) and (25, 19); GATA3, (13, 26) and (8, 11); Jun-B, (15, 118) and (14, 30); Jun-D, (107, 291) and (68, 130); NFAT4, (33, 35) and (64, 26); STAT4, (36, 29) and (76, 36); 14-3-3, (24, 45) and (83, 41); Bcl-XL, (11, 50) and (6, 7); and IAP, (14, 211) and (10, 41). Selected genes whose expression was constitutive but discordantly expressed for GW4 vs. MW10 (GW4,ME10) were: NKR
  • FIGS. 26 A- 26 D are graphs showing the transcriptional induction and release of cytokines and cytolytic enzymes by activated V ⁇ 24J ⁇ Q T cells.
  • FIGS. 26A and 26B compare the changes of expression on genes for cytokines and chemokines between the V ⁇ 24J ⁇ Q T cell clone GW4 (IL-4 + ) and the ME10 (IL-4-null) clone. For each individual transcript the anti-CD3 induced hybridization intensity is shown.
  • RNA was isolated, amplified and hybridized to genechips displaying probes for 250 genes of immunological interest (Affymetrix, San Jose, Calif.). This chip is custom designed for quantitative analysis by increasing the number of address features for the detection of each specific transcript.
  • FIG. 26C is a graph showing the amount of cytokines produced by supernatants from several V ⁇ 24J ⁇ Q T cell clones, based on quantitative ELISA analysis.
  • FIG. 26D is a graph showing the expression of CD40L.
  • the T cell clone BW5 was activated for four hours with PMA/ionomnycin stimulation and analyzed by flow cytometry.
  • FIGS. 27A and 27B show the CD1d restriction of V ⁇ 24J ⁇ Q T cell activation.
  • FIG. 27A shows the restriction of cytokine release and proliferation. T cell clones were co-cultured with C1R transfectants (expressing CD1a, CD1b, CD1c, or CD1d) or plate-bound anti-CD3. Secreted IL-4, IFN- ⁇ , or proliferation was assayed as described herein.
  • FIG. 27B shows the restriction of cytolysis. The panel of C1R transfectants was screened for cytolysis by V ⁇ 24J ⁇ Q T cell clones in standard four hour 51 Cr-release assays.
  • FIG. 28A is a graph of the CD1d expression on myeloid dendritic cells, based on flow cytometric analysis of cultured dendritic cells.
  • FIG. 28B is a picture of the immunoblot analysis of dendritic cells and control C1R transfectant cells (expressing CD1a or CD1d).
  • FIGS. 28 C- 28 J are pictures of the immunohistochemical staining of serial sections of a representative reactive lymph nodes biopsy (total of 10 biopsies).
  • FIG. 28C is a picture of hematoxylin-eosin staining at low power
  • FIG. 28D is a picture of anti-CD3 staining.
  • FIGS. 28E and 28F The box outlines the region through which serial sections were taken for staining with the various markers shown at higher magnification views in FIGS. 28E and 28F; (FIG. 28E) S100; (FIG. 28F) CD1d; (FIG. 28G) CD1a; (FIG. 28H) CD34.
  • Staining of sinus histiocytes (FIGS. 28I and 28K), and parafollicular regions (FIGS. 28L and 28M) for: hemato28ylin-eosin, (FIGS. 28 I and 28 L); CD68, a macrophage marker, (FIGS. 28 J and 28 M); and CD1d, (FIGS. 28K and 28N) are shown.
  • FIGS. 29 A- 29 D are graphs characterizing the cytolysis of myeloid dendritic cells by V ⁇ 24J ⁇ Q T cells.
  • FIG. 29A is graph showing the allogenic lysis of immature and mature dendritic cells by V ⁇ 24J ⁇ Q T cell clone GW4 in chromium release assays.
  • FIGS. 29B and 29C are graphs of the autologous and allogenic cytolysis of dendritic cells by V ⁇ 24J ⁇ Q T cell clones.
  • FIG. 29D is a graph of the abrogation of cytolysis by calcium chelation and inhibition by addition of the anti-CD1d monoclonal antibody, 42.1.
  • Clones BW3 and BW5 were co-cultured with subject BW DC in the presence or absence of EGTA and MgCl 2 (4 mM each) or F(ab′) 2 fragments from monoclonal antibody 42.1 or control IgG at 100 ⁇ g/ml.
  • FIG. 29E is a bar graph showing the secretion of IL-4 and IFN- ⁇ after co-culture of V ⁇ 24J ⁇ Q T cell clones OY3 and BW5 with autologous DC.
  • FIG. 30 is a schematic illustration of a model demonstrating the interaction of CD1d-restricted T cells with myeloid dendritic cells.
  • Activation of invariant V ⁇ 24J ⁇ Q T cells results in the secretion of cytokines and chemokines important for myeloid dendritic cell recruitment and activation.
  • important cell surface co-stimulatory molecules are also expressed.
  • CD1d is upregulated and activates CD1d-restricted T cells.
  • activated V ⁇ 24J ⁇ Q T cells upregulate perforin, granzyme B, and Granulysin. The CD1d-dependent secretion of these molecules then results in the lysis of myeloid dendritic cells.
  • TCRs T cell antigen receptors
  • the invention also provides purified antibodies to TCRs of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells which are useful for diagnostic, imaging, and therapeutic applications for diseases or conditions that are affected by the number and/or activity of T cells or specific subpopulations of T cells.
  • diseases or conditions include autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, and cancer.
  • NK T cells CD1d-reactive T cells
  • J ⁇ Q + T cells may be performed alone or in conjunction with the administration of other therapeutics, such as cytokines or tumor vaccines.
  • the antigen receptor of almost every individual clone of B and T lymphocytes is the product of a unique random somatic rearrangement. These rearrangements cause even identical twins to have millions of receptors that differ from one another.
  • the CD1d-selective and -reactive invariant TCR utilized by the invention represents a uniquely universal target. Because the sequence of the CDR3 loop of invariant TCR- ⁇ is identical in all individuals, this region is used to generate novel monoclonal and polyclonal antibodies that recognize the human invariant TCR- ⁇ chain. The monoclonal antibodies can preferentially identify and expand invariant T cells ex vivo from normal donors.
  • NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells may contribute to the Th1 protective immune responses against intracellular pathogens.
  • EMCV-D encephalomyocarditis virus
  • NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells are capable of augmenting Th1, Th2, or immune deviation responses depending on stimulus conditions
  • the response of the NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells can be modulated by either the in vivo administration of a cytokine before, during, or after the infusion of the ex vivo expanded NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells or by conducting the ex vivo expansion of the cells in the presence of a cytokine, or a combination of both methods.
  • the cytokine may be used to bias the T cells away from Th1 and Th2 responses and towards immune deviation responses which may contribute to the maintenance of pregnancy.
  • Immune deviation responses include the suppression of an ongoing immune response, such as a response at a immune-privileged site.
  • TGF- ⁇ and IL-10 are examples of cytokines that may participate in immune deviation responses (Sonoda et al., supra).
  • the antibodies of the present invention may be administered to a mammal, such as a human, to increase the number of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells in vivo for the prevention or treatment of autoimmune diseases, infectious diseases, allergies, asthma, inflammatory conditions, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, or cancer.
  • autoimmune diseases infectious diseases, allergies, asthma, inflammatory conditions, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, or cancer.
  • reduced levels of certain T cells such as invariant NK T cells were found in prostate cancer, multiple sclerosis, HIV, and type 1 diabetes patients (Examples 9, 11, and 12).
  • NK T cell function may be a general finding in advanced cancer.
  • antibodies that bind and inhibit the expansion or an activity of NK T cells, CD1d-reactive T cells, and J ⁇ Q + T cells, such as cytokine production or cytotoxicity may be used to inhibit T cell pathogenesis in a mammal.
  • an allergy may be caused or exacerbated by the Th2 response of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells, and thus, inhibiting the expansion or Th2 response of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be useful in the prevention or treatment of this condition.
  • reducing the Th1 response of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may improve, stabilize, or prevent autoimmune diseases or prevent spontaneous abortions.
  • the Th1 response induced by infectious agents, such as Hepatitis viruses can also cause damage which may be minimized by the inhibition of these cells.
  • antibodies that may inhibit these cells include monovalent or Fab molecules or antibodies that are conjugated to a toxin or radiolabel that damages the cells upon binding of the antibody to the cells.
  • the antibodies may also bind to an TCR expressed on the cells and prevent the binding of a ligand to the TCR.
  • the antibodies of the invention may also be used for imaging applications.
  • the antibodies can be coupled to a fluorescent or radiolabel for use in visualizing NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells in vivo or in samples of biopsies, blood, and other bodily material or fluids.
  • the antibodies may also be used to purify NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells from these samples.
  • the antibodies of the present invention may also bind to other NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells that are not invariant T cells and that are more abundant than invariant T cells in some locations, such as the bone marrow and liver.
  • an antibody of the present invention to purify invariant T cells from a peripheral blood sample, in which approximately 0.1% of the cells are invariant T cells, has enabled the isolation of cells that are 90-95% invariant T cells. Even higher purity may be achieved by using multiple rounds of purification or using multiple antibodies per round.
  • an antigen and antigen presenting cells Any antigen (e.g., a protein, peptide, lipid, carbohydrate, nucleic acid, infectious agent, or small molecule) for the T cells of interest may be used.
  • antigens for CD1d-reactive T cells include lipid or glycosyl-phosphatidylinositol antigens from an infectious pathogen and ⁇ -GalCer.
  • Other possible antigens include peptide and protein antigens, such as those presented by a class I or class II MHC molecule.
  • an antigen may be presented to a T cell of interest in the absence of APCs.
  • a soluble, liposome-associated, or immobilized form of an antigen-presenting molecule e.g., CD1d, class I MHC, or class II MHC
  • an antigen e.g., a lipid or a peptide
  • the antigen-presenting molecule that is used may be a naturally-occurring molecule or may be a chemically or genetically modified molecule.
  • the molecule may be expressed as a fusion protein, such as a GST, maltose-binding protein, hexa-histidine, or Fc region fusion protein.
  • a fusion protein such as a GST, maltose-binding protein, hexa-histidine, or Fc region fusion protein.
  • An antigen-presenting molecule expressed as a fusion protein containing a membrane-binding domain, transmembrane domain, or hydrophobic region may be mixed with lipid molecules to form a liposome or micelle containing the fusion protein.
  • This liposome or micelle having an antigen-presenting cell on its surface mimics the ability of an APC to present an antigen to a T cell.
  • an antigen-presenting molecule may be immobilized on a solid support, such as any rigid or semi-rigid surface.
  • the support can be any porous or non-porous water insoluble material, including, without limitation, membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, strips, plates, rods, polymers, particles, microparticles, and capillaries. If desired, the support can have a variety of surface forms, such as wells, trenches, pins, channels and pores, to which the proteins are bound.
  • An antigen-presenting molecule that is either in solution, immobilized on a solid support, or contained in a liposome or micelle may be contacted with an antigen for the T cell subpopulation of interest under conditions that allow the antigen-presenting molecule to bind the antigen. This antigen may then interact with T cells of interest, resulting in the expansion and/or activation of the T cells.
  • the current methods include either flow cytometry with less specific antibodies or a PCR-based method, which involves synthesizing and amplifying cDNA corresponding to the mRNA encoding the TCR- ⁇ chain and comparing this amplified cDNA to that from an invariant T cell clone and a control T cell clone, as described in Example 6.
  • the flow cytometry method has the advantage of determining the total number of invariant T cells; while the PCR-based method only determines the relative frequency of invariant T cells in a sample.
  • flow cytometry allows the determination of which markers are expressed on the invariant T cells.
  • the flow cytometry method is also a simpler and faster method which may be performed in about an hour compared to about a day for the PCR method and which does not require skill in molecular biology techniques.
  • the flow cytometry method using the antibodies of the present invention may be more desirable in a clinical setting. Similar methods may also be used to quantitate other NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells using the antibodies of the present invention.
  • the antibodies can be used in diagnosing a subject that has or is at risk for a disease. As described in Examples 6, 9, and 10, the antibodies can be used to quantitate the number of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells and to isolate these cells for the measurement of their cytokine production and cytotoxicity activities. These parameters can be compared between the subject and subjects who have and/or do not have the condition to diagnose the risk for, presence of, severity of, stage of, recovery from, or remission of a condition.
  • CD1d knockout mice (Sonoda et al., supra) which lack invariant TCR + T cells were used as the host for the production of anti-CDR3-loop antibodies. Since these mice lack the invariant TCR- ⁇ chain they are able to recognize peptides containing the human CDR3 loop sequence as foreign, and thus, generate antibodies against this epitope.
  • a peptide (CVVSDRGSTLGRLADCG, SEQ. ID No. 1) with an amino acid sequence corresponding to a portion of the CDR3 loop of the human invariant TCR- ⁇ chain was used.
  • This peptide was cyclized by reaction of the N-terminal amino group with the C-terminal carboxyl group.
  • the cyclic peptide was coupled to a keyhole limpet hemocyanin carrier and administered to CD1d KO mice, using standard techniques.
  • the peptide coupled to bovine serum albumin was administered as booster injections.
  • Invariant TCR + T cells were later administered to some mice to further stimulate the immune system.
  • Serum from the mice was tested for antibodies that bound the peptide coupled to ovalbumin, using a standard ELISA assay.
  • Hybridomas were generated from seropositive mice spleens using standard techniques.
  • ELISA positive hybridoma wells were further tested by FACS to compare binding of the antibodies to V ⁇ 24J ⁇ Q clones versus negative controls.
  • Cells producing antibodies that bound the V ⁇ 24J ⁇ Q clones but not the negative controls were cloned and fused to immune spleen cells using standard techniques. A group of hybridomas secreting monoclonal antibodies that specifically recognized the peptide were then identified using the ELISA assay.
  • Two antibodies (3A6 and 6B11) were identified that preferentially recognize human invariant T cells (FIG. 2).
  • 6B11 is an IgG1
  • 3A6 is an IgM.
  • PHA was used to expand bulk V ⁇ 24 + T cells purified by FACS. These cells were then analyzed with V ⁇ 24, V ⁇ 11 and 6B11 monoclonal antibodies ( FIG. 3).
  • the results demonstrate that 6B11 recognizes a small fraction of the V ⁇ 24 + T cells, similar to the V ⁇ 11 monoclonal antibody.
  • a single cell sorting from multiple donors was performed and resulted in the establishment of clones from 6B11 + T cells. 6B11 was also shown to induce cytokine responses identical to CD1d + targets.
  • This method for the production of antibodies reactive with a CDR3 loop may be also performed for any TCR of interest.
  • the sequence of the CDR3 loop of a TCR- ⁇ chain or - ⁇ chain of interest may be determined based on a sequence alignment with other TCR- ⁇ chains or - ⁇ chains or based on a modeled or experimentally determined three-dimensional structure of the TCR- ⁇ chain or - ⁇ chain of interest (Johnson and Wu, Nuc. Acid. Res. 29(1):205-206, 2001; http://immuno.bme.nwu.edu).
  • a cyclic peptide containing the sequence of this CDR3 loop may be used as the antigenic peptide for the production of antibodies reactive with the CDR3 loop, as described above.
  • This cyclic peptide may be administered to an animal (e.g., a laboratory animal or an animal of veterinary interest) in one or more doses.
  • the T cells of interest may also be administered to the animal.
  • mammals that may be used for the production of antibodies include mice, rats, rabbits, pigs, goats, sheep, horses, and cattle.
  • birds that may be used include chickens and turkeys.
  • the host animals may be wild-type animals, or they may be animals that have a reduced level or that lack the T cell subpopulation to which the antibody is being generated.
  • Other animals that may be used include animals that naturally, through genetic modification, or depletion lack the T cells. Any other animal that is capable of producing antibodies may also be used for the production of antibodies of the invention.
  • the resulting antibodies may be used for diagnostic or clinical applications involving other animals of the same genus or species as the host animal used for antibody production. Additionally, the antibodies of the invention may be used in applications involving animals of a different genus as the host animal. For example, antibodies that are produced in chickens may be used for the treatment or prevention of disease in other chickens or in other birds.
  • the CDR3 loop of a T cell subpopulation that is associated with that disease or condition may be used (see, for example, Table 1).
  • T cell subpopulations that are present at increased or decreased levels or that have increased or decreased activity in subjects with a disease or condition relative to the corresponding T cell subpopulation in control subjects without the disease or condition may be used for the generation of antibodies of the invention.
  • T cells may be isolated from a sample (such as a biopsy sample) obtained from a subject with a disease or condition.
  • the identity of T cell subpopulations in the sample may be determined using standard techniques, such as by FACS analysis using antibodies reactive with various T cell markers or by sequencing of the TCR- ⁇ chains or - ⁇ chains.
  • the CDR3 loop from an isolated T cell subpopulation may be used for the production of antibodies. These antibodies may be administered to subjects with the disease or condition (including the original subject from whom the T cell subpopulation was isolated as well as other subjects with the condition) to modulate the number and/or activity of relevant T cells in vivo. The antibodies may also be used for ex vivo expansion of the relevant T cells followed by readministration of the expanded T cells to the subject.
  • Antibodies of the invention may also be generated to any antigen-specific oligoclonally expanded T cell subpopulation encountered in animals (e.g., humans, other mammals, and birds) in response to given antigenic challenges. These T cell subsets include those T cells oligoclonally expanded in response to an immunodominant component of the antigen in multiple individuals. For example, an antigen of interest may be administered to an animal, and the oligoclonally expanded T cell subsets that are generated by this administration may be isolated and used for the production of antibodies as described herein.
  • TABLE 1 Citations for T cell receptors that may be used to generate antibodies for clinical applications involving the indicated disease or condition Disease/Condition Citation Multiple Sclerosis Wucherpfennig et al., J Exp.
  • FIG. 4 demonstrates that plate bound 6B11 monoclonal antibody can be used to achieve selective expansion of invariant T cells.
  • Analysis of more than 20 clones and 15 lines derived from 15 healthy donors using 6B11 in this way has produced entirely consistent results: 100% of these were V ⁇ 24 + V ⁇ 11 + CD161 + , highly CD1d-reactive, high IL-4 producers, and modest IFN- ⁇ producers (Table 2).
  • This result further demonstrate the specificity of these monoclonal antibodies and their use as a more definitive and single color alternative that does not require autologous APCs, which would be undesirable in the setting of a clinical trial.
  • the monoclonal antibody 6B11 + was also used alone (2 donors, 10 clones) or paired with the anti-V ⁇ 24 antibody (9 donors, 87 clones) to clone additional invariant T cells by single cell sorting. All the clones were confirmed to be invariant CD1d-restricted T cells. All of these clones also contained the invariant TCR- ⁇ chain, based on sequence analysis of the TCR- ⁇ chain. Additionally, all of the clones were CD1d restricted (Exley et al., J. Exp. Med. 186:109, 1997). In some donors there are very small numbers of 6B11 + and CD3 + T cells.
  • the two donors for whom only the 6B11 antibody was used for T cell purification did not have this population.
  • One of these donors had 0.01% V ⁇ 24 + V ⁇ 11 + cells in their peripheral blood.
  • the generation of 6/6 V ⁇ 24 + V ⁇ 11 + CD1d-restricted T cells from this individual highlights the specificity of 6B11.
  • the probability of 6/6 V ⁇ 24 + V ⁇ 11 + clones based on chance is (10 ⁇ 4 ) 6 .
  • the 6B11 antibody also stimulates secretion of IL-4 from CD1d-restricted T cell lines.
  • the 6B11 antibody has recognized invariant NK T cell clones and lines from all the donors that have been tested, including over 100 subjects from various ethnicities and age groups (see, for example, FIGS. 8 B- 8 I). This antibody has also recognized invariant NK T cells in both male and female subjects, including subjects diagnosed with cancer and subjects not diagnosed with cancer. These results demonstrate that the ability of the 6B11 antibody to recognize invariant NK T cells in not limited to a particular population of patients.
  • a positive and/or negative selection step may be performed.
  • antibodies that may be used in a positive selection step include the 6B11, 3A6, anti-V ⁇ 24, anti-V ⁇ 11, and anti-CD161 antibodies.
  • T cells of interest may also be enriched using a negative selection step that removes some of the undesired T cells or that removes other contaminants from a sample.
  • a negative selection step may be performed prior to, concurrent with, or subsequent to a positive selection step.
  • PBMC or another tissue source is blocked with autologous serum at 4° C.
  • An antibody reactive with invariant T cells e.g., 6B11 or 3A6 antibody
  • the antibody used in this method may be directly labeled with a conjugate such as FITC or biotin, or the antibody may be indirectly labeled with second reagent (e.g., anti-IgG-Fluorochrome or streptavidin-Fluorochrome).
  • second reagent e.g., anti-IgG-Fluorochrome or streptavidin-Fluorochrome
  • One or more other antibodies labeled with different fluorophores may also be added (FIGS. 7 A- 7 C and 9 ).
  • the solution of cells and antibodies is then washed using PBS or any other physiological buffer to remove unbound antibody.
  • the settings of the FACS cell sorter may be adjusted using either a positive (e.g., general anti-T cell antibody) or negative control antibody.
  • the sample of interest may be analyzed at a rate of approximately 100 million cells per hour using a high speed FACS sorter (e.g., ‘MoFlo’ FACS sorter).
  • the purified T cells may be expanded in T cell media [e.g., 10% Fetal Bovine or human autologous or AB pooled serum in RPMI-1640 or equivalent media supplemented with buffer (e.g.
  • a mitogen such as PHA, an anti-CD3 antibody, the 6B11 antibody, the 3A6 antibody, and/or ⁇ -GalCer antigenic stimulus CD3 monoclonal antibody (Exley et al. 1997 and 1998, supra; Wilson et al., Nature 391:177, 1998).
  • the antibody may be in solution or plate bound. Irradiated autologous feeders and IL-2 (10-100 U/ml) may also be added immediately or added at any time point during expansion.
  • IL-7 ⁇ 1-10 ng/ml
  • IL-15 ⁇ 1-10 ng/ml
  • Dexamethasone or a related selective immune suppressive agent may preferentially enhance the growth of invariant NK T cells while inhibiting growth of conventional T cells (e.g. Milner et al., J. Immunol. 163(5):2522-25229, 1999).
  • the media is replaced about twice a week with fresh media.
  • the T cells may be phenotypically analyzed by FACS. This analysis may be performed by staining with 6B11, anti-V ⁇ 24, anti-V ⁇ 11, anti-CD4, anti-CD8 ⁇ , or anti-CD161 antibodies.
  • PBMC or another tissue source is blocked with 1-10% autologous serum by mixing at 4° C. in PBS or other physiological buffer.
  • One or more antibodies that are reactive with invariant T cells e.g., 6B11 or 3A6 antibody
  • 6B11 or 3A6 antibody is added at a ratio of about 1-10 ⁇ g monoclonal antibody/10 7-8 PBMC is incubated for 20 minutes at 4° C.
  • the antibody used in this method may be directly labeled with a conjugate such as FITC or biotin or the antibody may be indirectly labeled with second reagent (e.g., anti-IgG-Fluorochrome or streptavidin-Fluorochrome).
  • One or more other antibodies labeled with different fluorophores may also be added (FIGS. 7 A- 7 C and 9 ).
  • the solution of cells and antibodies is then washed using PBS or any other physiological buffer.
  • Magnetic beads such as ⁇ 10-100 ⁇ l Miltenyi MACS Microbeads Anti-mouse IgG #484 or MACS Steptavidin Microbeads #481 per 10 7-8 PBMC; 10 5-7 Dynal Anti-IgG #M450 beads per 10 7-8 PBMC are added and the solution is mix at 4° C.
  • the beads are isolated using magnets and then washed according to the manufacturers' recommendations (e.g., using Miltenyi MS type separation columns such as product number 130-042-201 and MACS multistand, or Dynal beads directly in tubes on Dynal or comparable magnets).
  • the washed, purified cells are placed into multi-wells with T cell media [10% Fetal Bovine or human autologous or AB pooled serum in RPMI-1640 or equivalent media supplemented with buffer (e.g. Hepes), glutamine, and antibiotic (e.g. gentamicin)].
  • the cells may be expanded using a monoclonal antibody that is directly bound to the beads or in solution.
  • the cells may be expanded using media that is supplemented with a mitogen such as PHA, an anti-CD3 antibody, the 6B11 antibody, the 3A6 antibody, or ⁇ -GalCer antigenic stimulus CD3 monoclonal antibody.
  • a mitogen such as PHA, an anti-CD3 antibody, the 6B11 antibody, the 3A6 antibody, or ⁇ -GalCer antigenic stimulus CD3 monoclonal antibody.
  • the antibody may be in solution or bound to a plate, bead, or other solid surface.
  • various other reagents such as IL-7, IL-15, and dexamethasone may improve product yield and/or purity.
  • the expanded T cells may be phenotypically analyzed as described above.
  • FIGS. 10 A- 17 illustrate the ability of these magnetic bead purification methods to generate a large number of purified invariant T cells.
  • the 6B11 antibody was bound to either (1) Dynal superparamagnetic, polystyrene beads with affinity purified goat anti-mouse IgG covalently bound to the surface (Dynabeads® M-450 goat anti-mouse IgG, Product Number 110.05) or (2) Miltenyi MACS colloidal super-paramagnetic microbeads conjugated to goat anti-mouse IgG (Product Number 484-02, Miltenyi Biotec) according to the manufacturer's instructions (Dynabeads® M-450 goat anti-mouse IgG package insert; MACS goat anti-mouse IgG microbead instructions).
  • PBMC cells were enriched for invariant T cells by purification using 6B11-Dynal or 6B11-Miltenyi beads.
  • the purified cells were than expanded for four to eight weeks under various conditions.
  • the T cells were expanded in the presence of one or more of the following as indicated in FIGS. 12 - 17 : PHA, the 6B11 antibody, IL-5, IL-7, allogenic APC, and autologous APC.
  • the “PBMC” column represents PBMC cells that were not sorted prior to expansion.
  • the “Dynal @ 40” column represents PBMC cells that were purified using 6B11-Dynal beads prior to expansion.
  • the “Miltenyi @ 20′′ column represents PBMC cells that were sorted using 6B11-Miltenyi beads prior to expansion.
  • Invariant T cells were also purified using biotinylated 6B11.
  • ficoll purified PBLs were incubated with 6B11-biotin. The cells were then washed to remove unbound antibody and incubated with streptavidin conjugated to Miltenyi microspheres.
  • the 6B11 + cells were isolated by magnetic bead selection according to the manufacturer's specifications.
  • a total of 6 lines were generated using the 6B11 antibody in combination with IL-2 and IL-7. Three of the six lines were expanded and had a surface phenotype consistent with invariant V ⁇ 24 + CD1d-restricted T cells. The lines varied from 76%-99% invariant after expansion.
  • a FACS profile of the pure line is shown in FIG. 18.
  • NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells producing high levels of IL-4 will bias toward Th2 immune responses, while high IL-12 (induced from APCs by activated CD1d-reactive T cells) and/or IFN- ⁇ (produced by the T cells themselves) relative to IL-4 will drive (or at least be a marker of invariant T cells that will drive) immune responses against tumors or infectious pathogens.
  • NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be biased towards Th1-like NK or lymphokine-activated killer (LAK)-like cytotoxicity of invariant T cells, as well as, IFN- ⁇ production to potentially augment their in vivo anti-tumor or anti-pathogen activity.
  • LAK lymphokine-activated killer
  • invariant T cells can be re-polarized to the Th1 phenotype by culturing in 0.3 nM IL-12 (Exley et al., supra (1998)) during ex vivo expansion with an antibody of the present invention.
  • IL-15, IL-18, and type 1 INFs are also known to enhance Th1 polarization of human T cells.
  • cytokines or combinations of cytokines that may bias NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells towards Th1, Th2, or immune deviation responses include IL-2, IL-4, IL-7, IL-8, IL-10, IL-12, IL-13, IL-15, IL-18, IFN- ⁇ / ⁇ , IFN- ⁇ , and GM-CSF.
  • the Th2 response may be desirable for the prevention or treatment of an autoimmune disease.
  • cytokines may be used to bias NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells towards immune deviation responses instead of Th1 or Th2 responses.
  • Anti-IL-4 or anti-IL-12 antibodies which bind the cytokines produced by the T cells during expansion may also favor the immune deviation or Th2 response of these T cells. These cytokines may present during the entire time period for T cell expansion or may be present for only a portion of the time period for T cell expansion, such as during the last few days of T cell expansion.
  • the T cells may be functionally tested for secretion of L-4, IL-10, GM-CSF, and IFN- ⁇ .
  • the regulation of cytotoxicity against CD1d + (CIR, CD1d), ‘NK’ targets (JY, K562, 721.221, and YAC-1), or LAK targets by cytokine supplementation may also be determined (Exley et al., supra (1997); Exley et al., J. supra, (1998)).
  • one or more of the cytokines may be administered in vivo to patients before, during, or after re-introduction of the ex vivo expanded NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells to bias the re-introduced cells towards Th1, Th2, or immune deviation responses.
  • the monoclonal antibodies of the present invention may be used in a sensitive and specific method to determine whether a therapy for the treatment or prevention of a disease increases the number and/or preferentially binds or modulates the function of invariant T cells.
  • the method described below may be used to more accurately determine the effect of IL-12 treatment, or other treatments, on invariant T cells. Effects of cumulative doses on invariant T cells may be determined by performing this analysis after each dose.
  • Patients can serve as their own controls in these experiments. Approximately 20 ml of peripheral blood can be obtained from patients before treatment is initiated and subsequently during treatment. A small aliquot of these cells (approximately 10 6 cells) can be used for synthesis of cDNA, corresponding to mRNA encoding the TCR- ⁇ chain, and PCR amplification using V ⁇ 24 and C ⁇ primers to determine the frequency of cells expressing the invariant TCR- ⁇ chain. The fraction of invariant TCR in total population may be determined by calibration with an invariant T cell clone diluted in series into a control T cell clone.
  • flow cytometry may be used to determine the frequency and number of invariant T cells. Because of the low numbers involved, particulary in cancer patients, minimal numbers of conditions and large numbers of analyzed events (100,000) are used for each sample.
  • the anti-CDR3-loop monoclonal antibodies 6B11 and/or 3A6, which have been successfully conjugated to fluorescein using standard methods, may be used in a simple 1- or 2-color FACS analysis. Additionally, multi-color FACS may be used for simultaneous analysis of the invariant TCR- ⁇ chain using one or more monoclonal antibodies of the present invention and antibodies for other invariant T cell markers (V ⁇ 24, CD4, CD8, CD56, CD161, V ⁇ 11).
  • results for patient samples before and after treatment may be compared to determine the effect of the treatment on invariant T cell numbers.
  • Cytokine production and cytotoxicity by the activated cells may be assessed, as described in Example 5.
  • the results may be normalized for the number of invariant T cells, allowing direct comparisons for patients before and after treatment.
  • one round of expansion may be required for quantitation.
  • the patient sample may be directly subjected to monoclonal antibody expansion or the sample may be enriched for invariant T cells by high speed FACS sorting or magnetic bead immunoaffinity purification prior to expansion, using standard procedures.
  • FACS sorting one-color sorting using anti-V ⁇ ,24, 6B11, or 3A6 or 2-color sorting using 6B11 or 3A6 and anti-V ⁇ 24, V ⁇ 11, or CD161 may be performed.
  • one or more negative or positive selections may be conducted. For example, a negative depletion may be used to remove monocytes and/or B cells. Then a positive selection could be performed using the V ⁇ 24 monoclonal antibody and/or an anti-CDR3-loop monoclonal antibody.
  • the cells may be cultured with a plate bound or soluble anti-CDR3-loop monoclonal antibody (6B11 or 3A6) or with antigen pulsed autologous or allogenic APCs.
  • a plate bound or soluble anti-CDR3-loop monoclonal antibody (6B11 or 3A6) or with antigen pulsed autologous or allogenic APCs.
  • single round expansions of T cells are widely representative of initial proportions, the cells may alternatively be cultured for approximately one week prior to expansion to rest activated cells which may respond poorly to expansion compared to resting cells.
  • changes in invariant T cell numbers and/or responses may be correlated to key immune parameters including serum IFN- ⁇ , TNFs, and IL-15; expression of IL-12 and other Th1 cytokine and chemokine receptors; and changes in T cell, myeloid, and other cell markers (Fas, FasL, Lymnphotoxin- ⁇ , CD3, CD4, CD8, LFA-1, CD80, CD86, CD161).
  • key immune parameters including serum IFN- ⁇ , TNFs, and IL-15; expression of IL-12 and other Th1 cytokine and chemokine receptors; and changes in T cell, myeloid, and other cell markers (Fas, FasL, Lymnphotoxin- ⁇ , CD3, CD4, CD8, LFA-1, CD80, CD86, CD161).
  • This method for determining the change in the number or activity of invariant T cells may also be applied to NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells. If a pre-enrichment step is required to quantitate these T cells, any antibody that binds these cells, including antibodies of the present invention, may be used in the initial purification step or a negative selection may be used to remove cells that are not T cells or cells that are not NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • Invariant T cells from a peripheral blood sample (20 ml or from the product of leukopheresis) from a mammal may be enriched prior to ex vivo expansion using FACS sorting or immunoaffinity purification or expanded directly using an antibody of the present invention, as described in the previous examples.
  • the cells may be expanded in the presence of a cytokine to bias them towards Th1, Th2, or immune deviation responses.
  • IL-2, IL-7, or a mitogen may be added to stimulate cell expansion.
  • a secondary ex vivo expansion possibly after a second enrichment step, may be conducted under conditions used for the primary expansion to increase both cell number and purity.
  • the cells may be assayed for purity and cytokine production and cytotoxicity as described above. It should be noted that we and others have been able to establish long term human invariant T cell lines and clones from normal donors by multiple rounds of stimulation; thus, similar results are expect from other patients. Desirably at least 10 6 , more desirably at least 10 8 , and most desirably at least 10 9 invariant T cells are obtained after expansion. Desirably, the invariant T cells are at least 60%, 80, or 90% pure, based on the presence of V ⁇ 24, V ⁇ 11, and CD161 invariant T cell markers, and maintain the production of IFN ⁇ and cytotoxicity against CD1d + and ‘NK’ targets.
  • the number of starting cells may be increased by using larger blood samples or through leukopheresis.
  • an initial enrichment of invariant T cells may be performed by positive selection using the V ⁇ 24 or 6B11 monoclonal antibody conjugated to beads, as described above.
  • the invariant T cells may also be purified by FACS or antibody conjugated to beads prior to reinfusion.
  • the therapeutic potential of cellular reinfusion of expanded polyclonal invariant T cell lines may be compared to that of expanded invariant T cell clones, or pools thereof.
  • ex vivo expanded invariant T cells be limited to a particular mode of administration, dosage, or frequency of dosing; the present mode contemplates all modes of administration, including intramuscular, intravenous, intraarticular, intralesional, subcutaneous, or any other route sufficient to provide a dose adequate to prevent or treat an autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, host versus graft disease, spontaneous abortion, pregnancy, or cancer.
  • the cells are re-introduced into the mammal from which the blood sample was taken. It is also contemplated that the cells may be administered to a different mammal.
  • the cells may be administered to the mammal in a single dose or multiple doses. When multiple doses are administered, the doses may be separated from one another by, for example, one week to one month. As mentioned in Example 5, one or more cytokines may also be administered before, during, or after administration of the cells to bias them towards Th1, Th2, or immune deviation responses. It is to be understood that for any particular subject, specific dosage regimes should be adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions. Additionally, the T cells may be re-introduced as resting or activated cells, depending on the application. Resting cells would require in vivo activation.
  • NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be expanded ex vivo as described in Example 7. This method may be modified by expanding NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells from bone marrow, liver, umbilical cord, or other samples. Desirably, the sample contains a greater percentage of CD1d-reactive noninvariant T cells than peripheral blood. Additionally, costimulation (using CD1d, lipids, other antigens, or antibodies) or inhibition of costimulation may be used to expand a desired subpopulation of T cells. For example, stimulation of CD28 may favor the expansion of CD1d reactive noninvariant T cells over invariant T cells.
  • Soluble anti-CD161 antibodies that block CD161 stimulation may also favor the expansion of noninvariant T cells which are less dependent on CD161 stimulation than invariant T cells.
  • the amount of plate bound antibody may also be reduced which may increase the dependence on costimulation for expansion of the T cells.
  • the presence of different cytokines or combinations of cytokines may be used to favor the expansion of a desired subpopulation of T cells.
  • the NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be incubated with IL-4, IL-7, or IL-12 during expansion.
  • the expanded NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be re-introduced into mammals as described in Example 7.
  • prostate cancer patents have fewer invariant NK T cells than healthy patients.
  • the antibodies of the invention that increase the number or activity of invariant NK T cells may be used as therapeutics for the prevention, stabilization, or treatment of prostate cancer.
  • ex vivo expanded invariant NK T cells may be administered to subjects to prevent, stabilize, or treat prostate cancer.
  • Invariant NK T cells may also be present at reduced levels in subjects with other types of cancer; thus, these methods may be used to treat other forms of cancer.
  • Peripheral blood (10-20 ml) was drawn in heparin containing tubes from healthy donors and prostate cancer patients who had given their informed consent.
  • PBMCs were isolated using Ficoll-Paque (Amersham Pharmacia, Uppsala, Sweden).
  • V ⁇ 24 positive T cells were stained with an anti-V ⁇ 24 monoclonal antibody (C15) (Coulter, Miami, Fla.) followed by a goat anti-mouse IgG (H + L) FITC conjugate (KPL, Gaithersburg, Md.) and were sorted by high speed FACS (Modular Flow FACS, Cytomation, Boulder, Colo.) (Dellabona et al., J.Exp.Med. 180:1171, 1994).
  • Autologous PBMCs were irradiated (5000 rads) and used as APCs.
  • the FACS purified V ⁇ 24 + cells were initially co-cultured in 96 well flat bottomed plates (approximately 20,000 per well) with equal numbers of autologous irradiated PBMCs in the presence of ⁇ -GalCer (50 ng/ml, KRN7000, Kirin Brewery Co., Gunma, J ⁇ pan) and recombinant IL-2 (100 U/ml) (National Cancer Institute, Bethesda, Md.). Cultures were then gradually expanded into 24 well plates using the same medium. In some cultures, human recombinant IL-12 (1 ng/ml) (Genetics Institute, Cambridge, Mass.) was added during the last week of culture.
  • Antibodies used were anti-V ⁇ 24 PE, anti-V ⁇ 11 FITC, anti-CD8 ⁇ PE (Immunotech), anti-CD3 Cychrome, anti-CD161 PE, and anti-CD4 Cychrome (PharMingen, La Jolla, Calif.).
  • FACS buffer phosphate buffered saline with 1% fetal bovine serum and 0.1% NaN 3
  • Non-specific antibody binding was blocked by pre-incubating cells with 10% human serum for 15 minutes on ice. Antibodies were then added to cell suspensions and incubated for 20 minutes on ice. Cells were washed twice with FACS buffer and analyzed using a FACScan (Becton Dickinson) with Cell Quest Software.
  • Invariant NK T cells in the peripheral blood of healthy donors and cancer patients were quantitated by 2-color flow cytometry with a V ⁇ 24 monoclonal antibody and a V ⁇ 11 monoclonal antibody or the 6B11 monoclonal antibody against the invariant V ⁇ 24-J ⁇ Q TCR.
  • Invariant NK T cell lines, generated by ⁇ -GalCer expansion of V ⁇ 24 + T cells from healthy donors, were reactive with V ⁇ 24, V ⁇ 11, and 6B11 monoclonal antibodies (FIGS. 19A and 19D). The observation that V ⁇ 11 was expressed by virtually all of the ⁇ -GalCer expanded V ⁇ 24 + T cells from this and multiple other healthy donors further indicated that this V ⁇ was necessary to generate ⁇ -GalCer reactive invariant NK T cells.
  • V ⁇ 24 + V ⁇ 11 + double positive T cells were found in the peripheral blood of healthy donors in numbers that were comparable to V ⁇ 24 + 6B11 + cells, consistent with a large fraction of V ⁇ 24 + V ⁇ 11 + cells being invariant NK T cells (FIGS. 19B and 19E).
  • the average fraction of V ⁇ 24 + 6B11 + cells in a series of healthy donors was 0.11%.
  • Smaller numbers of V ⁇ 24 + V ⁇ 11 + T cells were found in the peripheral blood of advanced prostate cancer patients (Tables 4 and 5).
  • there were no detectable V ⁇ 24 + 6B11 + cells in 5 of 6 patients examined (FIGS. 19C and 19F).
  • invariant NK T cells Due to the small numbers of invariant NK T cells in peripheral blood, one round of ex vivo expansion was carried out to further assess the frequency and function of these cells.
  • Invariant NK T cells from healthy donors and advanced prostate cancer patients were isolated from peripheral blood through an initial FACS purification with a V ⁇ 24 specific monoclonal antibody, followed by selective expansion in vitro for three to four weeks with ⁇ -GalCer and autologous irradiated PBMCs as a source of APCs. T cells were then analyzed by dual staining with V ⁇ 24 and V ⁇ 11 or 6B11 monoclonal antibodies.
  • V ⁇ 24 + T cells stimulated in vitro with a T cell mitogen, PHA, yielded only a minor population of V ⁇ 24 + V ⁇ 11 + T cells that varied in number with different donors (FIG. 20A).
  • stimulation of the purified V ⁇ 24 + T cells from healthy donors with ⁇ -GalCer and autologous irradiated PBMCs yielded a major V ⁇ 24 + V ⁇ 11 + population (FIG. 20B, 94.4% of total cells).
  • the ⁇ -GalCer expanded V ⁇ 24 + V ⁇ 11 + T cells from healthy donors were assessed for CD1d recognition and cytokine production. Consistent with many of the cells being CD1d reactive invariant NK T cells, the cells produced substantial quantities of both IL-4 and IFN- ⁇ in response to CD1d transfected C1R cells, but not mock transfected C1R cells (FIG. 20C). The CD1d specificity of this recognition was further demonstrated by blocking with an anti-CD1d monoclonal antibody, 51.1, but not an isotype matched control Ab.
  • cytokine responses to CD1d were equivalent to those obtained after polyclonal stimulation of these T cells with PHA, confirming that most of the cells were indeed CD1d-reactive T cells. These responses were all comparable to those obtained previously with invariant NK T cell clones from healthy donors (Exley et al., J.Exp.Med. 186:109, 1997; Exley et al., J.Exp.Med. 188:867, 1998), indicating that these latter in vitro established clones reflected the functional status of the cells in vivo.
  • Invariant NK T cells from patients with advanced androgen independent prostate cancer were examined similarly. Relative to the healthy donors, there was a decrease in the total number of cultured cells recovered from patients with advanced androgen ablation refractory prostate cancer and a marked decrease in the fraction of expanded cells that were V ⁇ 24 + V ⁇ 11 + invariant NK T cells (FIGS. 21A and 21B, 98.5% versus 13.6% invariant NK T cells in a healthy donor and androgen ablation refractory prostate cancer patient, respectively).
  • invariant NK T cells have reduced activity in prostate cancer patients.
  • these invariant NK T cells produced less IFN- ⁇ (a Th1 effector).
  • incubation with IL-12 increased the activity of these invariant NK T cells to produce IFN- ⁇ .
  • cytokine production 1 ⁇ 10 5 cells/well in 96 well plates were co-cultured with an equal number of CD1d or mock transfected C1R cells in RPMI 1640 medium with 10% FBS, 20 U/ml IL-2, and 1 ng/ml PMA, as described previously (Exley et al., J.Exp.Med. 186:109, 1997). Cellular responses to CD1d were blocked with an anti-CD1d antibody, 51.1 at 10 ⁇ g/ml (Exley et al., J.Exp.Med. 186:109, 1997; Exley et al., Immunology 100:37, 2000). Supernatants were collected at 48 hours and 72 hours for IL-4 and IFN- ⁇ measurements, respectively.
  • cytokine levels were determined in triplicates by capture ELISA with matched antibody pairs in relation to cytokine standards (Endogen, Inc. Cambridge, Mass.). The limit of detection range of these assays for both IFN- ⁇ and IL-4 was 10-50 pg/ml.
  • Prostate cancer patient derived invariant NK T cells proliferated and produced similar levels of IL-4 in response to CD1d transfected cells as invariant NK T cells from a health donor (FIG. 22A). However, their production of IFN-was markedly reduced relative to invariant NK T cells from the healthy donor (FIG. 22B).
  • Analysis of IL-4 versus IFN- ⁇ production by ⁇ -GalCer expanded invariant NK T cells from a series of advanced prostate cancer patients and healthy donors confirmed a striking loss of IFN- ⁇ production by the cells derived from prostate cancer patients (FIG. 22C log scale for IFN- ⁇ ). This loss of IFN- ⁇ relative to IL-4 was most evident when IFN- ⁇ /IL-4 production ratios were compared, with a difference of approximately 50-fold between the prostate cancer and healthy donor derived NK T cell lines (FIG. 22E).
  • NK T cells may contribute to the anti-tumor effects of IL-12
  • Prostate cancer derived invariant NK T cells were treated with IL-12 (1 ng/ml) during the last week of culture to determine whether they could respond to this cytokine.
  • the IL-12 treated cells showed a marked increase in IFN- ⁇ production, and had ratios of IFN- ⁇ /IL-4 production that were comparable to those in the healthy donors (FIG. 22E, Cancer+IL-12).
  • MS Multiple sclerosis
  • CNS central nervous system
  • NK T cells This smaller number of NK T cells in MS patients suggests that increasing the number and/or activity of NK T cells may be useful for the treatment or prevention of MS, such as Relapsing Remitting, Primary Progressive, or Chronic Progressive MS. Additionally, samples from MS patients may be taken at various time points to determine whether the number of NK T cells in the periphery of these MS patients fluctuates over time and correlates with the status of their disease symptoms.
  • CD1d-restricted T cells may play an immunoregulatory role in multiple immunologic disorders.
  • the 6B11-FITC antibody in combination with the V ⁇ 24-PE antibody were used to determine the frequency of circulating CD1d-restricted T cells in diabetic patients on the day of diagnosis and in the HIV MACS cohort followed at UCLA medical center. PBL were stained and analyzed by FACS as described above. In addition, dendritic cell (DC) subtypes were determined for the HIV MACS cohort. The results are summarized in FIGS. 23 and 24.
  • An identification of the pattern of genes activated in a particular T cell type may provide information predictive of the function of that T cell subpopulation. For example, changes in gene expression patterns in the cytokine/chemokine family are particularly relevant given the association of cytokine secretion and the in vivo function for T cells. As described below, the gene expression pattern for V ⁇ 24J ⁇ Q T cells was determined to better understand the role of these cells in autoimmunity and type 1 diabetes. These method may also be used to determine the role of any other T cells subpopulation of interest in the development and/or progression of any disease or condition.
  • genes were grouped into six distinct expression patterns, using the Self-Organizing Map algorithm (Tamayo et al., Proc. Nat. Acad. Sci. USA 96, 2907-2912, 1999).
  • JunB and GATA3 were recently reported to be preferentially expressed in Th2 T cells.
  • the transcription factor NFAT4 thought to act in part as a suppressor of IL-4 transcription was overexpressed in the IL-4-null clone relative to the IL-4 + clone. Based on this data, the discordant regulation of other genes such as transcription factors might be predicted to be important for controlling Th-phenotype.
  • a model for regulated genes whose expression concurs with multiple independent biological observations is presented in FIG. 25B.
  • CD1d on Myeloid Dendritic Cells Stimulates Cytokine Secretion from and Cytolytic Activity of V ⁇ 24J ⁇ Q T Cells
  • DC are a distinct population of bone marrow derived antigen presenting cells that play a key role in initiating T cell responses. In humans, three populations of DC are found at very low frequencies (0.03-0.3%) in peripheral blood. Human DC include two myeloid populations—Langerhans cell precursors (Lin ⁇ /CD11c + /CD1a + /I1-3R ⁇ ) and tissue DC (Lin ⁇ /CD11c + /CD1a ⁇ /IL-3R ⁇ )— and a “plasmacytoid” DC (PDC) population (Lin ⁇ /CD11c ⁇ /CD1a ⁇ /IL-3R + ).
  • PDC plasmacytoid
  • the PDC population also express CXCR3 and CD62L that facilitate homing to the high endothelial venule and movement into lymphoid tissues.
  • PDC undergo activation in lymphoid through ligation of CD40 or by exposure to LPS and produce a variety of cytokines and upregulate T cell stimulatory capacity.
  • the MDC occupy either the epidermis or dermal and other tissue sites, respectively.
  • MDC reside in peripheral tissues in an immature state and readily take up antigens until such time that they receive a signal provided by infection or tissue damage. At this point they become activated and migrate to the draining lymph nodes where they readily produce cytokine such as IL-12p70 and activate naive T cells.
  • Recent studies support a differential role for the MDC and PDC populations in directing the development of Th1 and Th2 responses. Studies in mice and humans also suggest that defective MDC maturation and function may play a role in type 1 diabetes pathogenesis.
  • PBMC peripheral blood mononuclear cells
  • irradiated feeder PBMC 50,000 cells/well
  • irradiated 721.221 lymphoblastoid cells 500 cells/well
  • PHA-P 1 ⁇ g/mL
  • IL-2 10 U/ml
  • IL-7 10 U/mL
  • RPMI 1640 Sigma
  • Clones were then propagated with periodic restimulation by anti-CD3 antibody in the presence of irradiated allogenic feeder PBMC and anti-CD3 antibody. Clones were confirmed to be positive for V ⁇ 24 and NKR-P1A by flow cytometry and to have the V ⁇ 24J ⁇ Q CDR3 TCR by sequencing.
  • V ⁇ 24J ⁇ Q T cell clones were stimulated (25,000/well) with plate-bound anti-CD3 or control isotype antibody for 4, 8, or 24 hours. Supernatants were collected and assayed for IL-4, IFN- ⁇ , macrophage inflammatory protein-1- ⁇ (MIP-1- ⁇ ), MIP-1- ⁇ , TNF- ⁇ , and GM-CSF by quantitative ELISA (Quantikine kits, R &D Systems, Minneapolis, Minn., USA). After 24 hours, 1 ⁇ Ci/well of [ 3 H-]thymidine (Dupont NEN, Boston, Mass.) was added and incorporation measured as described (Wilson et al., Nature 391:177, 1998).
  • CD1 isoforms CD1a, CD1b, CD1c, CD1d and pSR- ⁇ -neo vector alone
  • Monocyte-derived dendritic cells were generated from fresh PBMC using an adaptation of previously published methods (O'Doherty et al., Journal of Experimental Medicine 178:1067, 1993), or positively selected by anti-CD14 microsphere enrichment as described in the manufacturers protocols (Miltenyi Biotec, Auburn, Calif., USA). Briefly, freshly isolated PBMC prepared from allogenic of sygneic donors were enriched for monocytes by adherence and washing.
  • the remaining monocytes were cultured in R10 supplemented with recombinant human IL-4 (rhIL-4; Genzyme) and rhGM-CSF at 1000 U/ml each for an additional 7 days, yielding a non-adherent population of cells that were at least 90% CD1a + /DR + /CD3 ⁇ /CD14 ⁇ by flow cytometric analysis.
  • Cytolytic activity by V ⁇ 24J ⁇ Q T cell clones was determined by measuring the specific release of 51 Cr at fours hours.
  • Target cells were labeled with 50 uCi Na 2 51 Cr (New England Nuclear, North Billerica Mass.) for one hour and washed twice. Cytolytic activity was determined in standard chromium release assays with U-bottom 96-well microtiter plates containing 10 4 labeled target cells per well, with the indicated ratios of effector cells. After a 4 hour incubation at 37° C., the supernatants were harvested and counted on a gamma counter (Cobra, Packard, Downer's Grove Ill.).
  • Percent specific lysis was calculated as [(experimental release ⁇ spontaneous release)/(maximal release ⁇ spontaneousrelease)] ⁇ 100.
  • cytolysis was tested under conditions of calcium chelation, in the presence of EGTA and MgCl 2 each at 4 mM.
  • the 42.1 anti-CD1d monoclonal antibody was a kind gift from Dr. Steven Porcelli (Brigham & Women's Hospital).
  • F(ab′) 2 fragments of 42.1 and IgG1 control antibodies were prepared with an Immunopure F(ab′) 2 kit, Pierce (Rockford, Ill.).
  • Goat F(ab′) 2 Anti-mouse IgG-FITC, human adsorbed, was obtained from Caltag (Burlingham, Calif., USA)
  • FcR-Blocking reagentTM human IgG was obtained from Miltenyi Biotec (Auburn, Calif., USA).
  • NOR3.2 was obtained from Biosource International (Camarillo, Calif., USA).
  • Anti-V ⁇ 24, anti-V ⁇ 11, anti- ⁇ TCR, and anti-CD83 were obtained from Immunotech (Coulter/Beckman, Fullerton, Calif., USA).
  • Anti-CD1a, anti-CD4, anti-CD8, anti-CD40L, anti-CD80, anti-CD86, and HLA-DR were obtained from Pharmingen (San Diego, Calif., USA).
  • Anti-CD3, clone UCHT1 was obtained from Ancell (Bayport, Minn., USA) and IgG1 control from Sigma (St. Louis, Mo.).
  • Immunoprecipitates of CD1d from lysates of 5 ⁇ 10 5 C1R/CD1d cells, 4 ⁇ 10 7 dendritic cells, and 4 ⁇ 10 7 control C1R/neo cells were prepared using monoclonal antibody 42.1 coupled to protein A beads.
  • the immunoprecipitates were resolved by SDS-PAGE (5-15%), and probed with an affinity purified rabbit anti-CD1d polyclonal antibody (Exley et al., Journal of Experimental Medicine 188:867, 1998). Bands were visualized by chemiluminescence.
  • NOR3.2 monoclonal antibody was used to determine CD1d expression in fixed, paraffin-embedded tissue by immunoperoxidase staining (Vectastain ABC elite kit with visualization using NovaRed, Vector Laboratories, Burlingame, Calif.). Staining was done as per manufacturer's specifications with NOR3.2 used at 1:100 dilution. The specificity of the signal was confirmed by blocking experiments using a GST-CD1d fusion protein as compared to the GST protein alone.
  • a detailed analysis of the transcriptional profile of V ⁇ 24J ⁇ Q T cells was performed using high-density oligonucleotide arrays. Activation of clones derived from normal donors resulted in the expression of numerous effector molecules believed to be important for the recruitment and differentiation of myeloid dendritic cells (FIGS. 26A and 26B). Among these were four of sixteen chemokines examined and included MIP-1 ⁇ and MIP-1 ⁇ , which are thought to recruit macrophages and immature dendritic cells in vivo. Also produced were GM-CSF, IL-4, and TNF- ⁇ , cytokines involved in the differentiation and maturation of myeloid dendritic cells and their subsequent maturation.
  • Activation also induced the expression of eight of twenty-six cytokines tested. These cytokines, as well as CD40 ligand and 4-1BB, were produced by each of the V ⁇ 24J ⁇ Q T cell clones examined (FIGS. 26 A- 26 D, 27 A, and 27 B).
  • activated V ⁇ 24J ⁇ Q T cells expressed enhanced levels of perforin and granzyme B, proteins usually associated with classic cytotoxic T cells.
  • their immunomodulatory functions might not be limited to cytokine release, but could involve cytolytic activities as well.
  • CD1d-restricted triggering of the invariant TCR activates the secretion of cytokines and a concurrent cytolytic response, a situation similar to that observed for cytotoxic T cells triggered by MHC class I and peptide epitopes.
  • CD1d is Expressed on Myeloid-lineage Dendritic Cells
  • V ⁇ 24J ⁇ Q T cell clones The combination of cytokines and cytolytic proteins produced by V ⁇ 24J ⁇ Q T cell clones suggested an effector role beyond that of simple Th2 priming by IL-4 secretion, as previously proposed. Given these observations, immune regulation by V ⁇ 24J ⁇ Q T cells might involve interaction with myeloid dendritic cells which are important for the generation of Th1-like responses. Immature dendritic cells were derived from peripheral blood monocytes differentiated in vitro with IL-4 and GM-CSF and subsequently matured with monocyte conditioned media (O'Doherty et al., Journal of Experimental Medicine 178:1067, 1993). Peripheral blood monocytes are known to express low levels of CD1d, which is promptly lost on culture in vitro.
  • CD1d was not found to be expressed by follicular dendritic cells or follicle tingle body macrophages, and was largely absent from sinus histiocytes, i.e. CD1d expression was targeted to T cell-dependent lymphoid regions. While surveying other histiocytic/monocytic populations in other forms of reactive lymph nodes processes, striking CD1d staining was found on epithelioid histiocytes in both caseating granulomas of M. tuberculosis infections and other non-mycobacterial granulomas. Murine V ⁇ 14J ⁇ 281 T cells have been shown to be required for granuloma formation after challenge with lipid extracts from M. tuberculosis .
  • V ⁇ 24J ⁇ Q T cells The ability of V ⁇ 24J ⁇ Q T cells to interact with dendritic cells was confirmed by testing several V ⁇ 24J ⁇ Q T cell clones for cytolysis of DC from multiple healthy donors of differing MHC haplotypes (FIGS. 29A and 29B). Both allogenic and autologous dendritic cells were lysed by the clones, indicating that killing was neither MHC-restricted nor alloreactive. Furthermore, cytolysis was completely abrogated by calcium chelation and markedly inhibited by the anti-CD1d monoclonal antibody 42.1 (FIG. 29C). These data suggest that killing was mediated via the perforin/granzyme pathway and required CD1d.
  • V ⁇ 24J ⁇ Q T cells Comparison of immature (CD83 ⁇ ) versus mature (CD83 + ) phenotype dendritic cells demonstrated no consistent difference in recognition by V ⁇ 24J ⁇ Q T cells (FIG. 29A). Activation of V ⁇ 24J ⁇ Q T cells by dendritic cells also resulted in secretion of both IL-4 and IFN- ⁇ (FIG. 29D). Thus, exposure of V ⁇ 24J ⁇ Q T cells to dendritic cells expressing CD1d triggered both cytolytic functions and cytokine release.
  • V ⁇ 24J ⁇ Q T cells After activation by anti-CD3, V ⁇ 24J ⁇ Q T cells were found to be capable of secreting a broad panel of cytokines, chemokines, and co-stimulatory proteins important for the recruitment and differentiation of myeloid dendritic cells including IL-4 and GM-CSF.
  • Myeloid dendritic cells cultured in the presence of these gene products expressed CD1d and became specific targets for CD1d-restricted killing by V ⁇ 24J ⁇ Q T cells.
  • CD1d was preferentially expressed on myeloid dendritic cells in the paracortical T cell zones of lymph nodes corroborating the in vitro expression data.
  • DC1 human myeloid-derived dendritic cells
  • DC2 lymphoid-derived dendritic cells
  • their immunomodulatory function is not limited to Th2 bias induced by IL-4 secretion (Maldonado-Lopez et al., J Exp Med 189:587, 1999; Rissoan et al., Science 283:1183, 1999; Reid et al., Curr Opin Immunol 12:114, 2000).
  • DC1 cells secreted high levels of IL-12 and induced T cells with a Th1 phenotype.
  • V ⁇ 24J ⁇ Q T cells The site of interaction between V ⁇ 24J ⁇ Q T cells and dendritic cells is presently unknown.
  • the paucity of V ⁇ 24J ⁇ Q T cells in a typical lymph node suggests it may occur extranodally and that the CD1d positive dendritic cells present within the lymph node may be those which escape peripheral destruction (Bendelac et al., Annual Review of Immunology 15:535, 1997; Porcelli et al., Annu Rev Immunol 17:297, 1999).
  • the parallel tissue distributions of V ⁇ 24J ⁇ Q T cells within the reticuloendothelial system and the in vivo expression pattern of CD1d are strong evidence for a key role for their interaction in regulating the generation of cellular immune responses.
  • the CD1d-restricted T cells are distributed in the liver, gut, spleen, lymph nodes, and thymus (Bendelac et al., Annual Review of Immunology 15:535, 1997; Porcelli et al., Annu Rev Immunol 17:297, 1999), sites of active antigen sampling and presentation by professional antigen presenting cells. Unregulated dendritic cells have also been previously shown to be capable of initiating and maintaining autoimmunity by the presentation of tissue-specific self-antigens.
  • V ⁇ 24J ⁇ Q T cells are activated by CD1d on myleloid dendritic cells (DC1) to secrete chemokines and cytokines important for the recruitment and differentiation of dendritic cells and thus play an important role in modulating dendritic cell function (FIG. 30).
  • DC1d myleloid dendritic cells
  • This interaction also activates the cytolytic functions of V ⁇ 24J ⁇ Q T cells resulting in negative regulation of Th1 cellular immune responses through cytolysis of dendritic cells.
  • This system may be reciprocal to the negative regulation of lymphoid dedritic cells (DC2) by mature T cells which serves to limit Th2 cellular responses (Rissoan et al., Science 283:1183, 1999).
  • deficient DC maturation and function may contribute to autoimmune disease such as type 1 diabetes
  • insufficient antigen presenting cell activity for the generation of regulatory cells (Th2 cells, CD4 + /CD25 + T cells) or for the induction of death in effector T cells.
  • regulatory cells Th2 cells, CD4 + /CD25 + T cells
  • intrinsic defects in the maturation of MDC may contribute to defects in NK T cell numbers or function.
  • defects in NK T cells my reciprocally limit DC maturation or lead to a failure to kill these antigen presenting cells, thus allowing a persistent and untoward immune response.
  • any of the antibodies of the invention may be tested in an in vivo animal model to determine the pharmacological and pharmacokinetic properties of the antibodies. For example, the half-life, bio-distribution, and efficacy of the antibody may be determined.
  • One possible method involves the administration of human invariant T cells or any other T cell population of interest to a SCID or otherwise immune-deficient animal such as a mouse and administration of an anti-invariant T cell antibody or any other antibody of the invention to the animal to determine whether the antibody modulates the activity or number of the administered T cells in vivo.
  • a population of T cells that contains 1-10 million T cells of interest is administered i.v. or to any site of interest.
  • One or more antibodies are administered, prior to, concurrent with, or following administration of the T cells.
  • the antibody may be administered at any point during the lifetime of the administered T cells in the host animal.
  • Approximately 1-100 ug of the antibody is administered in the same site or in a different site as the site of administration of the T cells. If detectably labeled antibodies are used, the location and amount of administered antibody and/or T cells may be monitored in vivo based on fluorescence or radioactivity.
  • histology, immunological, and/or biochemical measurements may be performed ex vivo on tissues from the animal.
  • the biological activity of the antibody or T cell subpopulation may be measured by analyzing the amount or activity of cytokines in a serum or tissue sample.
  • the activation of other cells, such as other T cells, by the administered T cell subpopulation may be measured.
  • the number of CD69 + T cells may be measured by FACS sorting with an anti-CD69 antibody.
  • the antibodies of the present invention may be administered to a mammal, possibly in addition to the administration of a cytokine, for the in vivo expansion of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells for the treatment or prevention of an autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, or cancer. As described in Example 7, all modes of administration, dosing, and frequency are contemplated.
  • compositions containing one or more antibodies of the invention may be prepared as described previously in Remingtion's Pharmaceutical Sciences by E. W. Martin.
  • Pharmaceutical stabilizing compounds, delivery vehicles, or carrier vehicles may be used.
  • human serum albumin or other human or animal proteins may be used.
  • Phospholipid vesicles or liposomal suspensions are possible pharmaceutically acceptable carriers or delivery vehicles.
  • An antibody of the invention that is covalently linked to a fluorescent label or radiolabel may be used to visualize the in vivo distribution, quantity, or migration of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.
  • This imaging of NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells may be used to identify subjects who are at risk for or have an autoimmune disease, viral infection, bacterial infection, parasitic infection, infection by a eukaryotic pathogen, allergy, asthma, inflammatory condition, graft versus host disease, graft rejection, immunodeficiency disease, spontaneous abortion, pregnancy, or cancer.
  • this method may be used to determine the effect of a therapy for one of the above diseases on NK T cells, CD1d-reactive T cells, or J ⁇ Q + T cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Reproductive Health (AREA)
  • Pulmonology (AREA)
  • Communicable Diseases (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Virology (AREA)
US09/885,768 2000-06-19 2001-06-19 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations Abandoned US20020164331A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US09/885,768 US20020164331A1 (en) 2000-06-19 2001-06-19 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
US11/541,958 US8138314B2 (en) 2000-06-19 2006-10-02 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
US13/423,683 US20120258040A1 (en) 2000-06-19 2012-03-19 Compositions and methods of monoclonal and polyclonal antibodies specific for t cell subpopulations
US14/495,502 US20150299320A1 (en) 2000-06-19 2014-09-24 Compositions and methods of monoclonal and polyclonal antibodies specific for t cell subpopulations

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US21246600P 2000-06-19 2000-06-19
US09/885,768 US20020164331A1 (en) 2000-06-19 2001-06-19 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/541,958 Continuation US8138314B2 (en) 2000-06-19 2006-10-02 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations

Publications (1)

Publication Number Publication Date
US20020164331A1 true US20020164331A1 (en) 2002-11-07

Family

ID=22791141

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/885,768 Abandoned US20020164331A1 (en) 2000-06-19 2001-06-19 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
US11/541,958 Expired - Fee Related US8138314B2 (en) 2000-06-19 2006-10-02 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
US13/423,683 Abandoned US20120258040A1 (en) 2000-06-19 2012-03-19 Compositions and methods of monoclonal and polyclonal antibodies specific for t cell subpopulations
US14/495,502 Abandoned US20150299320A1 (en) 2000-06-19 2014-09-24 Compositions and methods of monoclonal and polyclonal antibodies specific for t cell subpopulations

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/541,958 Expired - Fee Related US8138314B2 (en) 2000-06-19 2006-10-02 Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
US13/423,683 Abandoned US20120258040A1 (en) 2000-06-19 2012-03-19 Compositions and methods of monoclonal and polyclonal antibodies specific for t cell subpopulations
US14/495,502 Abandoned US20150299320A1 (en) 2000-06-19 2014-09-24 Compositions and methods of monoclonal and polyclonal antibodies specific for t cell subpopulations

Country Status (10)

Country Link
US (4) US20020164331A1 (de)
EP (2) EP1297017B1 (de)
JP (2) JP5184732B2 (de)
AU (1) AU2001268599A1 (de)
CA (1) CA2413866C (de)
DK (2) DK1297017T3 (de)
ES (2) ES2586206T3 (de)
HK (2) HK1158219A1 (de)
PT (1) PT1297017E (de)
WO (1) WO2001098357A2 (de)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020192681A1 (en) * 2000-10-24 2002-12-19 Whitehead Institute For Biomedical Research Response of dendritic cells to a diverse set of pathogens
US20040087485A1 (en) * 2000-12-25 2004-05-06 Yaron Iian Educated nkt cells and their uses in the treatment of immune-related disorders
US20060078994A1 (en) * 2004-10-07 2006-04-13 Don Healey Mature dendritic cell compositions and methods for culturing same
US20060093612A1 (en) * 2002-05-02 2006-05-04 Srivastava Pramod K Use of heat shock proteins to enhance efficacy of antibody therapeutics
US20060116332A1 (en) * 2004-11-02 2006-06-01 The Board Of Trustees Of The Leland Stanford Junior University Methods for inhibition of NKT cells
US20060134126A1 (en) * 2002-01-23 2006-06-22 Daniel Zimmerman Peptide constructs for treating disease
US20060216316A1 (en) * 2005-03-28 2006-09-28 Dhodapkar Madhav V In vivo expanded NKT cells and methods of use thereof
US20070082400A1 (en) * 2004-10-07 2007-04-12 Donald Healey Mature dendritic cell compositions and methods for culturing same
WO2005032463A3 (en) * 2003-09-30 2008-10-16 Enzo Therapeutics Inc Educated nkt cells and their uses in the treatment of immune-related disorders
US8617884B2 (en) 2002-06-28 2013-12-31 Life Technologies Corporation Methods for eliminating at least a substantial portion of a clonal antigen-specific memory T cell subpopulation
WO2018031507A1 (en) * 2016-08-09 2018-02-15 Angimmune, Llc Treatment of cancer using a combination of immunomodulation and check point inhibitors
US9932402B2 (en) 2011-10-27 2018-04-03 Nkt Therapeutics Inc. Humanized antibodies to iNKT
US10226476B2 (en) 2001-03-26 2019-03-12 Dana-Farber Cancer Institute, Inc. Method of attenuating reactions to skin irritants
US10329315B2 (en) 2012-10-12 2019-06-25 The Brigham And Women's Hospital, Inc. Glycosphingolipids and methods of use thereof

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003065985A2 (en) * 2002-02-01 2003-08-14 Schering Corporation Uses of mammalian cytokine; related reagents
WO2003092615A2 (en) * 2002-05-01 2003-11-13 Beth Israel Deaconess Medical Center Use of anti-cd1 antibodies for the modulation of immune responses
JP5053101B2 (ja) * 2005-01-28 2012-10-17 ブリガム ヤング ユニヴァーシティー CD1d−拘束NKT細胞の細菌糖脂質による活性化
WO2012018950A1 (en) * 2010-08-03 2012-02-09 Beth Israel Deaconess Medical Center Methods and compositions for treatment of metabolic disorders
FI3556774T3 (fi) 2011-03-11 2024-03-15 Beth Israel Deaconess Medical Ct Inc Anti-cd40-vasta-aineita ja niiden käyttötapoja
US20130273089A1 (en) 2011-11-03 2013-10-17 Tolera Therapeutics, Inc. Antibody and methods for selective inhibition of t-cell responses
CN104870011A (zh) 2011-11-03 2015-08-26 托莱拉医疗股份有限公司 选择性抑制t细胞应答的抗体和方法
CN104769104A (zh) 2011-12-12 2015-07-08 细胞药物有限公司 扩大t细胞的方法
GB201121308D0 (en) 2011-12-12 2012-01-25 Cell Medica Ltd Process
WO2013119947A1 (en) 2012-02-09 2013-08-15 Baylor College Of Medicine Pepmixes to generate multiviral ctls with broad specificity
SI3307322T1 (sl) 2015-09-04 2021-08-31 Primatope Therapeutics Inc. Humanizirana protitelesa proti CD40 in njihove uporabe
WO2017049291A1 (en) 2015-09-18 2017-03-23 Baylor College Of Medicine Immunogenic antigen identification from a pathogen and correlation to clinical efficacy
CN107557333B (zh) * 2017-10-16 2018-07-31 北京鼎成肽源生物技术有限公司 一种用于细胞治疗的新抗原呈递细胞的制备方法
WO2020232247A1 (en) 2019-05-14 2020-11-19 Provention Bio, Inc. Methods and compositions for preventing type 1 diabetes
JP2023501722A (ja) * 2019-11-14 2023-01-18 マレンゴ・セラピューティクス,インコーポレーテッド 抗tcr抗体分子およびその使用
MX2023004964A (es) * 2020-11-06 2023-06-29 Nat Center Neurology & Psychiatry Agente terapéutico para enfermedades progresivas ocasionadas por el incremento en células t positivas a cd4 positivas a eomes.

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5853737A (en) * 1992-12-10 1998-12-29 Brigham And Women's Hospital Method for inducing a CD1-restricted immune response

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7119248B1 (en) * 1994-04-12 2006-10-10 Miltenyi Biotec Gmbh Antibodies against epitopes with homology to self antigens, methods of preparation and applications thereof
US20020068301A1 (en) * 1997-05-28 2002-06-06 Hung-Sen Lai Cyclic peptide libraries and methods of use thereof to identify binding motifs
US6162609A (en) * 1997-12-31 2000-12-19 The Brigham And Women's Hospital, Inc. Diagnostic and therapeutic methods based upon Vα24JαQT cells
DE19905048A1 (de) * 1999-02-08 2000-08-10 Gsf Forschungszentrum Umwelt Verwendung depletorisch oder mitogen wirkender Antikörper in der Immuntherapie

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5853737A (en) * 1992-12-10 1998-12-29 Brigham And Women's Hospital Method for inducing a CD1-restricted immune response

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020192681A1 (en) * 2000-10-24 2002-12-19 Whitehead Institute For Biomedical Research Response of dendritic cells to a diverse set of pathogens
US20040087485A1 (en) * 2000-12-25 2004-05-06 Yaron Iian Educated nkt cells and their uses in the treatment of immune-related disorders
US10226476B2 (en) 2001-03-26 2019-03-12 Dana-Farber Cancer Institute, Inc. Method of attenuating reactions to skin irritants
US20060134126A1 (en) * 2002-01-23 2006-06-22 Daniel Zimmerman Peptide constructs for treating disease
US20060093612A1 (en) * 2002-05-02 2006-05-04 Srivastava Pramod K Use of heat shock proteins to enhance efficacy of antibody therapeutics
US8617884B2 (en) 2002-06-28 2013-12-31 Life Technologies Corporation Methods for eliminating at least a substantial portion of a clonal antigen-specific memory T cell subpopulation
US9528088B2 (en) 2002-06-28 2016-12-27 Life Technologies Corporation Methods for eliminating at least a substantial portion of a clonal antigen-specific memory T cell subpopulation
EP2465526A3 (de) * 2003-09-30 2012-08-29 Enzo Therapeutics, Inc. a fully owned subsidiary of Enzo Biochem, Inc. Gebildete NKT-Zellen und deren Verwendung bei der Behandlung von immunbedingten Störungen
CN101389349B (zh) * 2003-09-30 2012-09-05 恩佐治疗学股份有限公司 驯化的nkt细胞及其在治疗免疫相关病症中的应用
WO2005032463A3 (en) * 2003-09-30 2008-10-16 Enzo Therapeutics Inc Educated nkt cells and their uses in the treatment of immune-related disorders
EP2465528A3 (de) * 2003-09-30 2012-09-05 Enzo Therapeutics, Inc. a fully owned subsidiary of Enzo Biochem, Inc. Gebildete NKT-Zellen und deren Verwendung bei der Behandlung von immunbedingten Störungen
EP2465529A3 (de) * 2003-09-30 2012-09-05 Enzo Therapeutics, Inc. a fully owned subsidiary of Enzo Biochem, Inc. Gebildete NKT-Zellen und deren Verwendung bei der Behandlung von immunbedingten Störungen
EP2465525A3 (de) * 2003-09-30 2012-08-29 Enzo Therapeutics, Inc. a fully owned subsidiary of Enzo Biochem, Inc. Gebildete NKT-Zellen und deren Verwendung bei der Behandlung von immunbedingten Störungen
US20070082400A1 (en) * 2004-10-07 2007-04-12 Donald Healey Mature dendritic cell compositions and methods for culturing same
US9556455B2 (en) 2004-10-07 2017-01-31 Argos Therapeutics, Inc. Mature dendritic cell compositions and methods for culturing same
US11248209B2 (en) 2004-10-07 2022-02-15 Coimmune, Inc. Mature dendritic cell compositions and methods for culturing same
US20080145931A1 (en) * 2004-10-07 2008-06-19 Argos Therapeutics, Inc. Mature Dendritic Cell Compositions and Methods of Culturing Same
US8513008B2 (en) 2004-10-07 2013-08-20 Argos Therapeutics, Inc. Mature dendritic cell compositions and methods for culturing same
US20060078994A1 (en) * 2004-10-07 2006-04-13 Don Healey Mature dendritic cell compositions and methods for culturing same
US10184108B2 (en) 2004-10-07 2019-01-22 Argos Therapeutics, Inc. Mature dendritic cell compositions and methods for culturing same
US8822223B2 (en) 2004-10-07 2014-09-02 Argos Therapeutics, Inc. Mature dendritic cell compositions and methods for culturing same
US9523077B2 (en) 2004-10-07 2016-12-20 Argos Therapeutics, Inc. Mature dendritic cell compositions and methods for culturing same
US20060116332A1 (en) * 2004-11-02 2006-06-01 The Board Of Trustees Of The Leland Stanford Junior University Methods for inhibition of NKT cells
US8679499B2 (en) 2004-11-02 2014-03-25 The Board Of Trustees Of The Leland Stanford Junior University Methods for relieving asthma-associated airway hyperresponsiveness
US7682614B2 (en) * 2004-11-02 2010-03-23 The Board Of Trustees Of The Leland Stanford Junior University Methods for inhibition of NKT cells
US7837990B2 (en) * 2005-03-28 2010-11-23 The Rockefeller University In vivo expanded NKT cells and methods of use thereof
US20060216316A1 (en) * 2005-03-28 2006-09-28 Dhodapkar Madhav V In vivo expanded NKT cells and methods of use thereof
US9932402B2 (en) 2011-10-27 2018-04-03 Nkt Therapeutics Inc. Humanized antibodies to iNKT
US10329315B2 (en) 2012-10-12 2019-06-25 The Brigham And Women's Hospital, Inc. Glycosphingolipids and methods of use thereof
WO2018031507A1 (en) * 2016-08-09 2018-02-15 Angimmune, Llc Treatment of cancer using a combination of immunomodulation and check point inhibitors

Also Published As

Publication number Publication date
CA2413866A1 (en) 2001-12-27
JP5184732B2 (ja) 2013-04-17
ES2586206T3 (es) 2016-10-13
HK1054949A1 (en) 2003-12-19
EP2336187B8 (de) 2016-07-06
DK1297017T3 (da) 2012-09-17
US8138314B2 (en) 2012-03-20
DK2336187T3 (en) 2016-08-22
PT1297017E (pt) 2012-09-04
EP2336187A2 (de) 2011-06-22
WO2001098357A3 (en) 2003-01-03
JP2004501165A (ja) 2004-01-15
US20070160600A1 (en) 2007-07-12
EP2336187A3 (de) 2012-01-11
ES2389645T3 (es) 2012-10-30
HK1158219A1 (zh) 2012-07-13
WO2001098357A2 (en) 2001-12-27
EP1297017B1 (de) 2012-06-13
CA2413866C (en) 2014-09-02
JP2013014587A (ja) 2013-01-24
US20120258040A1 (en) 2012-10-11
EP2336187B1 (de) 2016-05-04
US20150299320A1 (en) 2015-10-22
EP1297017A2 (de) 2003-04-02
AU2001268599A1 (en) 2002-01-02

Similar Documents

Publication Publication Date Title
US8138314B2 (en) Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
KR102618231B1 (ko) 변형된 만능성 줄기 세포, 및 제조 및 사용 방법
EP2893003B1 (de) Selektive und kontrollierte expansion von nk zellen
KR20200024770A (ko) γδ T 세포의 증식, 조성물 및 이의 사용 방법
JP2022533621A (ja) 認識分子を含む操作された免疫細胞
US7527972B2 (en) Uses of bispecific antibody coated dendritic cells pulsed with antigens and GM-CSF in immune regulation
AU2023201286A1 (en) Chimeric antigen receptor therapy T cell expansion kinetics and uses thereof
US20210155941A1 (en) Compositions and methods for making engineered t cells
EP3191109B1 (de) Zellpopulation zur verwendung bei der behandlung von krebs
AU2007202851A1 (en) Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations
WO2024054863A1 (en) T-cell receptor that targets egfr mutation and methods of using the same
JP2003033175A (ja) 選択的免疫応答抑制を誘導する末梢血樹状細胞サブセット

Legal Events

Date Code Title Description
AS Assignment

Owner name: BETH ISRAEL DEACONESS MEDICAL CENTER, MASSACHUSETT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EXLEY, MARK A.;BALK, STEVEN P.;REEL/FRAME:012442/0268;SIGNING DATES FROM 20010907 TO 20010913

Owner name: DANA-FARBER CANCER INSTITUTE, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WILSON, SAMUEL B.;REEL/FRAME:012442/0307

Effective date: 20010913

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION D/B/A MASSACHUSET

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNMENT PREVIOUSLY RECORDED ON REEL 012442 FRAME 0307;ASSIGNOR:WILSON, SAMUEL B.;REEL/FRAME:022192/0708

Effective date: 20081031

Owner name: DANA-FARBER CANCER INSTITUTE, MASSACHUSETTS

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNMENT PREVIOUSLY RECORDED ON REEL 012442 FRAME 0307;ASSIGNOR:WILSON, SAMUEL B.;REEL/FRAME:022192/0708

Effective date: 20081031

Owner name: THE GENERAL HOSPITAL CORPORATION D/B/A MASSACHUSET

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNMENT PREVIOUSLY RECORDED ON REEL 012442 FRAME 0307. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNEES ARE "THE GENERAL HOSPITAL CORPORATION AND DANA-FARBER CANCER INSTITUTE," NOT "DANA-FARBER CANCER INSTITUTE";ASSIGNOR:WILSON, SAMUEL B.;REEL/FRAME:022192/0708

Effective date: 20081031

Owner name: DANA-FARBER CANCER INSTITUTE, MASSACHUSETTS

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNMENT PREVIOUSLY RECORDED ON REEL 012442 FRAME 0307. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNEES ARE "THE GENERAL HOSPITAL CORPORATION AND DANA-FARBER CANCER INSTITUTE," NOT "DANA-FARBER CANCER INSTITUTE";ASSIGNOR:WILSON, SAMUEL B.;REEL/FRAME:022192/0708

Effective date: 20081031