US20060147428A1 - Allogeneic and xenogeneic transplantation - Google Patents

Allogeneic and xenogeneic transplantation Download PDF

Info

Publication number
US20060147428A1
US20060147428A1 US11/151,744 US15174405A US2006147428A1 US 20060147428 A1 US20060147428 A1 US 20060147428A1 US 15174405 A US15174405 A US 15174405A US 2006147428 A1 US2006147428 A1 US 2006147428A1
Authority
US
United States
Prior art keywords
recipient
cells
donor
graft
cell
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
US11/151,744
Inventor
David Sachs
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.)
General Hospital Corp
Original Assignee
General Hospital Corp
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
Priority claimed from US08/114,072 external-priority patent/US5624823A/en
Priority claimed from PCT/US1994/001616 external-priority patent/WO1995021527A1/en
Priority claimed from US08/243,653 external-priority patent/US5658564A/en
Priority claimed from PCT/US1994/005527 external-priority patent/WO1994026289A1/en
Priority claimed from US08/266,427 external-priority patent/US5614187A/en
Priority claimed from US08/451,210 external-priority patent/US6296846B1/en
Priority claimed from US08/458,720 external-priority patent/US5876708A/en
Priority to US11/151,744 priority Critical patent/US20060147428A1/en
Application filed by General Hospital Corp filed Critical General Hospital Corp
Publication of US20060147428A1 publication Critical patent/US20060147428A1/en
Assigned to NIH - DEITR reassignment NIH - DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE GENERAL HOSPITAL CORPORATION
Assigned to NIH - DEITR reassignment NIH - DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE GENERAL HOSPITAL CORPORATION
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
    • 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
    • A61K35/14Blood; Artificial blood
    • A61K35/16Blood plasma; Blood serum
    • 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
    • A61K35/26Lymph; Lymph nodes; Thymus; Spleen; Splenocytes; Thymocytes
    • 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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem 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
    • A61K35/37Digestive system
    • A61K35/407Liver; Hepatocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/001Preparations to induce tolerance to non-self, e.g. prior to transplantation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1282Devices used in vivo and carrying the radioactive therapeutic or diagnostic agent, therapeutic or in vivo diagnostic kits, stents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-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
    • C07K16/2803Immunoglobulins [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
    • C07K16/2812Immunoglobulins [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 CD4
    • 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
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • 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
    • C12N2510/00Genetically modified cells

Definitions

  • This invention relates to the replacement of thymus function and to the induction or restoration of immunological tolerance.
  • the invention further relates to tissue and organ transplantation.
  • the thymus is the central organ for the development of mature, self-tolerant T cells that recognize peptide antigens in the context of self major histocompatibility (MHC) antigens.
  • MHC self major histocompatibility
  • the requirement for self MHC molecules to present antigen is termed MHC restriction.
  • Athymic individuals do not have an organ in which to generate normal numbers of MHC restricted T cells and are therefore immunoincompetent.
  • host T cells of an athymic T cell depleted host which has received a thymic graft can mature in the donor thymic tissue, e.g., in xenogeneic thymic tissue.
  • Host T cells which mature in the implanted xenogeneic thymic tissue are immunocompetent.
  • the invention features, in one aspect, a method of restoring or inducing immunocompetence (or restoring or promoting the thymus-dependent ability for T cell progenitors to mature or develop into functional mature T cells) in a host or recipient, e.g., a primate host or recipient, e.g., a human, which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells for a normal immune response.
  • a host or recipient e.g., a primate host or recipient, e.g., a human
  • the invention includes the step of introducing into the primate host, donor thymic tissue, e.g., xenogeneic thymic tissue, preferably fetal or neonatal thymic tissue, so that host T cells can mature in the implanted thymic tissue.
  • donor thymic tissue e.g., xenogeneic thymic tissue, preferably fetal or neonatal thymic tissue
  • the donor of the thymic tissue is a xenogeneic species and: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the host is a primate, e.g., a human, and the thymic tissue is swine, e.g., miniature swine, thymic tissue, or primate thymic tissue.
  • the thymic tissue is xenogeneic and: liver or spleen tissue, preferably fetal or neonatal liver or spleen tissue, is implanted with the thymic tissue; donor hematopoietic cells, e.g., cord blood stem cells or fetal or neonatal liver or spleen cells, are administered to the recipient, e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously; the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • liver or spleen tissue preferably fetal or neonatal liver or spleen tissue
  • donor hematopoietic cells e.g., cord blood stem cells or fetal or neonatal liver or spleen cells
  • the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • the method includes: (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient natural killer (NK) cells, e.g., by introducing into the recipient an antibody capable of binding to NK cells of the recipient, to prevent NK mediated rejection of the thymic tissue; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host T cell function, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host CD4 + cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4 + cells of the recipient.
  • NK natural killer
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, or the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient (preferably prior to thymic tissue transplantation).
  • low dose e.g., between about 100 and 400 rads, whole body irradiation
  • the administration of a myelosuppressive drug e.g., or the administration of a hematopoietic stem cell inactivating or depleting antibody
  • thymic tissue or hematopoietic stem cell transplantation inactivating thymic T cells by one or more of: irradiating the host with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the host's blood, e.g., by contacting the host's blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • the method further includes the step of identifying a host or recipient which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells for a normal immune response.
  • a graft which is obtained from a different organ than is the thymic tissue is implanted in the recipient; and the recipient does not receive hematopoietic stem cells, e.g., bone marrow cells, from the donor or the donor species.
  • hematopoietic stem cells e.g., bone marrow cells
  • tolerance to the donor tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient.
  • a donor antigen e.g., a donor MHC gene
  • human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and a class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue.
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • xenogeneic thymic tissue can be used to induce tolerance to a xenogeneic graft in a recipient.
  • the invention features, a method of inducing tolerance in a recipient mammal, e.g., a primate, e.g., a human, of a first species to a graft obtained from a mammal of a second species, e.g., a discordant species.
  • the method includes: prior to or simultaneous with transplantation of the graft, introducing into the recipient mammal thymic tissue, e.g., thymic epithelium, preferably fetal or neonatal thymic tissue, of the second species; and (optionally) implanting the graft in the recipient.
  • the thymic tissue prepares the recipient for the graft that follows, by inducing immunological tolerance at the T-cell level.
  • the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the recipient is a human and the thymic tissue is swine, e.g., miniature swine, thymic tissue, or primate thymic tissue.
  • liver or spleen tissue preferably fetal or neonatal liver or spleen tissue
  • donor hematopoietic cells e.g., cord blood stem cells or fetal or neonatal liver or spleen cells
  • the recipient e.g., a suspension of fetal liver cells administered intraperitoneally or intravenously
  • the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • the method includes (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting host CD4 + cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4 + cells of the recipient.
  • NK natural killer
  • An anti-mature T cell antibody which lyses T cells as well as NK cells can be administered. Lysing T cells is advantageous for both thymic tissue and xenograft survival.
  • Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable.
  • Monoclonal preparations can be used in the methods of the invention.
  • the recipient does not receive hematopoietic cells from the donor or the donor species: the same mammal of the second species is the donor of both the graft and the thymic tissue; the donor mammal is a swine, e.g., a miniature swine; an anti-human thymocyte polyclonal anti-serum, obtained, e.g., from a horse or pig is administered to the recipient.
  • the same mammal of the second species is the donor of both the graft and the thymic tissue
  • the donor mammal is a swine, e.g., a miniature swine
  • an anti-human thymocyte polyclonal anti-serum obtained, e.g., from a horse or pig is administered to the recipient.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • thymic T cells include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • the method further includes the step of identifying a host or recipient which is in need of a graft.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • tolerance to the xenogeneic thymic tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient.
  • a donor antigen e.g., a donor MHC gene
  • human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue.
  • a recipient primate e.g., a human
  • expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor.
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • the invention features, a method of restoring or inducing immunocompetence in a recipient, e.g., a primate recipient, e.g., a human, at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells to provide a normal immune response.
  • the invention includes the steps of introducing into the primate recipient, donor thymic tissue, e.g., xenogeneic thymic tissue, so that recipient T cells can mature in the implanted donor thymic tissue.
  • the thymic tissue is preferably fetal or neonatal thymic tissue.
  • the thymic tissue is xenogeneic and: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the recipient is a human and the thymic tissue is vertebrate, e.g., swine, e.g., miniature swine, thymic tissue, or primate thymic tissue.
  • Acceptance of a graft, especially a xenogeneic graft will depend on the stage of the immune disorder. Generally, the more advanced the disorder the more compromised the recipient immune system and the easier it is to induce acceptance of donor thymic tissue. In some cases, the tolerizing effect of the graft itself will be sufficient to provide for acceptance of xenogeneic thymus. In other cases, additional measures will be needed. Thus, the method can include other steps which facilitate acceptance of the donor tissue or otherwise optimize the method.
  • liver or spleen tissue preferably fetal or neonatal liver or spleen tissue
  • thymic tissue is implanted with the thymic tissue
  • donor hematopoietic cells e.g., cord blood stem cells or fetal or neonatal liver or spleen cells
  • the recipient e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously
  • the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • the method includes (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; the method includes (preferably prior to or at the time of introducing the thymic tissue into the recipient), depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host CD4 + cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4 + cells of the recipient.
  • NK natural killer
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • thymic tissue or hematopoietic stem cell transplantation inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • the method further includes the step of identifying a host or recipient which is at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells to provide a normal immune response.
  • an acquired immune disorder e.g., a human at risk for AIDS
  • tolerance to donor tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient.
  • a donor antigen e.g., a donor MHC gene
  • human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and a class II MHC gene, and the cells implanted in a human recipient who will receive swine thymic tissue.
  • a recipient primate e.g., a human
  • expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor.
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • the invention features, a method of restoring or inducing immunocompetence in a recipient, e.g., a primate recipient, e.g., a human, at risk for an acquired immune disorder, (e.g., a human at risk for AIDS) which is unable to produce a normal or sufficient number of mature functional T cells to provide normal immune function.
  • a recipient e.g., a primate recipient, e.g., a human
  • an acquired immune disorder e.g., a human at risk for AIDS
  • the invention includes the steps of introducing into the primate recipient, donor hematopoietic stem cells, so that donor T cells can mature in the recipient thymus.
  • the donor stem cells are from a xenogeneic donor and: the xenograft hematopoietic stem cells are from a discordant species; the hematopoietic stem cells are from a concordant species; the recipient is a human and the hematopoietic stem cells are vertebrate, e.g., swine, e.g., miniature swine, hematopoietic stem cells, or primate hematopoietic stem cells.
  • Acceptance of the donor cells will depend on the stage of the immune disorder. Generally, the more advanced the disorder the more compromised the recipient immune system and the easier it is to induce acceptance of donor, especially xenogeneic donor, tissue. In some cases, the tolerizing effect of the stem cells themselves will be sufficient to provide for acceptance. In other cases, additional measures will be needed. Thus, the method can include other steps which facilitate acceptance of the donor cells or otherwise optimize the method.
  • liver or spleen tissue preferably fetal or neonatal liver or spleen tissue
  • the donor hematopoietic cells e.g., cord blood stem cells or fetal or neonatal liver or spleen cells
  • the recipient e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously.
  • the method includes, (preferably prior to or at the time of introducing the donor cells into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; the method includes (preferably prior to or at the time of introducing the thymic tissue into the recipient), depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host CD4 + cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4 + cells of the recipient.
  • NK natural killer
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • thymic tissue or hematopoietic stem cell transplantation inactivating thymic T cells by one or more of: irradiating the recipient mamma i with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • the method further includes the step of identifying a host or recipient which is at risk for an acquired immune disorder, (e.g., a human at risk for AIDS) and which is unable to produce a normal or sufficient number of mature functional T cells to provide normal immune function.
  • an acquired immune disorder e.g., a human at risk for AIDS
  • tolerance to donor tissue can be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient.
  • a donor antigen e.g., a donor MHC gene
  • human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and a class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue.
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • the invention features, a method of restoring or inducing immunocompetence in a recipient, e.g., a primate recipient, e.g., a human, at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), which is thymus-function deficient and thus unable to produce a normal number of mature functional T cells or a sufficient number of mature functional T cells for a normal immune response.
  • a primate recipient e.g., a human
  • an acquired immune disorder e.g., a human at risk for AIDS
  • the invention includes the steps of introducing into the primate recipient, donor thymic tissue, preferably, xenogeneic thymic tissue, and donor hematopoietic stem cells, preferably xenogeneic hematopoietic stem cells, so that donor T cells can mature in the implanted donor thymic tissue.
  • donor thymic tissue preferably, xenogeneic thymic tissue
  • donor hematopoietic stem cells preferably xenogeneic hematopoietic stem cells
  • the thymic graft is a xenograft and: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the recipient is a human and the thymic tissue is vertebrate, e.g., swine, e.g., immature swine, thymic tissue, or primate thymic tissue.
  • the xenograft hematopoietic stem cells are from a discordant species; the hematopoietic stem cells are a concordant species; the recipient is a human and the hematopoietic stem cells are vertebrate, e.g., swine, e.g., miniature swine, hematopoietic stem cells, or primate hematopoietic stem cells.
  • the donor of the thymic graft and the donor of the stem cells are: the same organism; from the same species; syngeneic; matched at least one MHC locus; matched at least one class I MHC locus; matched at least one class II MHC locus; sufficiently MHC matched that one will not reject a graft from the other; miniature swine from a herd which is completely or partially inbred.
  • Acceptance of donor tissue will depend on the stage of the immune disorder. Generally, the more advanced the disorder the more compromised the recipient immune system and the easier it is to induce acceptance of donor tissue. In some cases, the tolerizing effect of the donor tissue itself will be sufficient to provide for acceptance. In other cases, additional measures will be needed.
  • the method can include other steps which facilitate acceptance of donor tissue or otherwise optimize the method.
  • liver or spleen tissue preferably fetal or neonatal liver or spleen tissue
  • thymic tissue is implanted with the thymic tissue
  • donor hematopoietic cells e.g., cord blood cells or fetal or neonatal liver or spleen cells
  • the recipient e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously
  • the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • the method includes, (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; the method includes, (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient mammal; (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting host CD4 + cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4 + cells of the recipient.
  • NK natural killer
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • thymic tissue or hematopoietic stem cell transplantation inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • a drug which depletes or inactivates natural antibodies e.g., deoxyspergualin
  • the administration of an anti-IgM antibody e.g., anti-IgM antibody
  • the adsorption of natural antibodies from the recipient's blood e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species
  • the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • the method further includes the step of identifying a host or recipient which is at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), and which is thymus-function deficient and thus unable to produce a normal number of mature functional T cells or a sufficient number of mature functional T cells for a normal immune response.
  • an acquired immune disorder e.g., a human at risk for AIDS
  • tolerance to donor tissue can be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient.
  • a donor antigen e.g., a donor MHC gene
  • human recipient stem cells can be engineered to express a swine MHC gene, e.g. a swine class I or class II MHC gene, or both a class I and a class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue.
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • the invention features, a method of inducing immunological tolerance in a recipient mammal, e.g., a primate, e.g., a human, of a first species to a graft obtained from a donor mammal of a second species, e.g., a discordant species e.g., a discordant primate species.
  • a recipient mammal e.g., a primate, e.g., a human
  • a graft obtained from a donor mammal of a second species
  • a discordant species e.g., a discordant primate species.
  • the method includes: prior to or simultaneous with transplantation of the graft, introducing into the recipient mammal hematopoietic stem cells, e.g., bone marrow cells, or fetal liver or spleen cells, of the second species; (preferably, the hematopoietic stem cells home to a site in the recipient mammal); optionally, (preferably prior to introducing the hematopoietic stem cells into the recipient mammal), depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient mammal an antibody capable of binding to natural killer (NK) cells of the recipient mammal, to prevent NK mediated rejection of the hematopoietic cells; and (optionally) implanting the graft in the recipient.
  • hematopoietic stem cells e.g., bone marrow cells, or fetal liver or spleen cells
  • the recipient mammal hematopoietic stem cells home to a site in the
  • the hematopoietic cells prepare the recipient for the graft that follows, by inducing tolerance at both the B-cell and T-cell levels.
  • hematopoietic cells are fetal liver or spleen, or bone marrow cells, including immature cells (i.e., undifferentiated hematopoietic stem cells; these desired cells can be separated out of the bone marrow prior to administration), or a complex bone marrow sample including such cells can be used.
  • anti-NK antibody is anti-human thymocyte polyclonal anti-serum.
  • a second, anti-mature T cell antibody can be administered as well, which lyses T cells as well as NK cells. Lysing T cells is advantageous for both bone marrow and xenograft survival.
  • Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable.
  • Monoclonal preparations can be used in the methods of the invention.
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species-specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus; and the step of prior to hematopoietic stem cell transplantation, introducing into the recipient mammal an antibody capable of binding to mature T cells of the recipient mammal.
  • donor species-specific stromal tissue preferably hematopoietic stromal tissue, e.g., fetal liver or thymus
  • hematopoietic stem cell transplantation introducing into the recipient mammal an antibody capable of binding to mature T cells of the recipient mammal.
  • Preferred embodiments include those in which: the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells; the hematopoietic stem cells are introduced simultaneously with, or prior to, the antibody; the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells, and the hematopoietic stem cells are introduced simultaneously with, or prior to, the antibody.
  • Preferred embodiments include those in which: the same mammal of the second species is the donor of both the graft and the hematopoietic cells; the donor mammal is a swine, e.g., a miniature swine; the introduction is by intravenous injection; and an anti-human thymocyte polyclonal anti-serum; obtained, e.g. from a horse or pig is administered.
  • the same mammal of the second species is the donor of both the graft and the hematopoietic cells
  • the donor mammal is a swine, e.g., a miniature swine
  • the introduction is by intravenous injection
  • an anti-human thymocyte polyclonal anti-serum obtained, e.g. from a horse or pig is administered.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • thymic tissue or hematopoietic stem cell transplantation inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • Preferred embodiments include: (preferably prior to hematopoietic stem cell transplantation) depleting, inactivating, or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to mature T cells of the recipient.
  • the graft is obtained from a different organ than the hematopoietic stem cells.
  • Preferred embodiments include those in which: the primate is a cynomolgus monkey; the primate is a human; the stromal tissue is fetal or neonatal liver; the stromal tissue is fetal or neonatal thymus; the mammal is a swine; e.g., a miniature swine; the graft is a liver; the graft is a kidney.
  • tolerance to the xenogeneic thymic tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient.
  • a donor antigen e.g., a donor MHC gene
  • human recipient stem cells can be engineered to express a swine class I or class II MHC gene, or both a class I and II gene, and the cells implanted in a human recipient who will receive swine thymic tissue.
  • a recipient primate e.g., a human
  • expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor.
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • the invention features a method of inducing immunological tolerance in a recipient mammal, e.g., a primate, e.g., a human to a graft obtained from a donor mammal of the same species.
  • the method includes the following: (preferably prior to or simultaneous with transplantation of the graft) introducing into the recipient mammal hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, obtained from a mammal (preferably, the hematopoietic stem cells home to a site in the recipient mammal); and, preferably, introducing the graft into the recipient.
  • hematopoietic stem cells e.g., bone marrow cells or fetal liver or spleen cells
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species-specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus; and prior to hematopoietic stem cell transplantation, depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient mammal an antibody capable of binding to mature T cells of the recipient mammal.
  • donor species-specific stromal tissue preferably hematopoietic stromal tissue, e.g., fetal liver or thymus
  • depleting, inactivating or inhibiting recipient T cells e.g., by introducing into the recipient mammal an antibody capable of binding to mature T cells of the recipient mammal.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • thymic tissue or hematopoietic stem cell transplantation inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • an anti T cell antibody e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • the method includes: (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient natural killer (NK) cells, e.g., by introducing into the recipient an antibody capable of binding to NK cells of the recipient, to prevent NK mediated rejection of the thymic tissue; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host T cell function, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient.
  • NK natural killer
  • T cell help which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance.
  • immunosuppressant e.g., cyclosporine
  • T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression.
  • the invention further provides several methods of inducing tolerance to foreign antigens, e.g., to antigens on allogeneic or xenogeneic tissue or organ grafts. These methods can be used individually or in combination with one another. For example, it has been found that the short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA), can significantly prolong graft acceptance.
  • a help reducing agent e.g., a short high dose course of cyclosporine A (CsA)
  • CsA cyclosporine A
  • the short term help reduction-methods of the invention can be combined with one or more other methods for prolonging graft acceptance.
  • a short course of high dose cyclosporine treatment to induce tolerance to unmatched donor class I and other minor unmatched donor antigens can be combined with implantation of retrovirally transformed bone marrow cells to induce tolerance to unmatched donor class II.
  • a short course of high dose cyclosporine administered to induce tolerance to unmatched donor class I and other minor antigens can also be combined with implantation of donor bone marrow cells to induce tolerance to unmatched donor class II.
  • the invention features, in one aspect, a method of inducing tolerance in a recipient mammal, e.g., a primate, e.g., a human, to an allograft from a donor primate including: implanting the graft in the recipient; and administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine.
  • the short course of help reducing treatment is generally administered at about the time the graft is introduced into the recipient.
  • the recipient is mismatched at a first locus which affects graft rejection, e.g., an MHC class I or II locus, or a minor antigen locus, and matched, or tolerant of a mismatch, at a second locus which affects graft rejection, e.g., an MHC class I or II locus, or a minor antigen locus.
  • a first locus which affects graft rejection e.g., an MHC class I or II locus, or a minor antigen locus
  • Matching at the second locus can be achieved by selection of a recipient or donor of the appropriate genotype.
  • the recipient can be rendered tolerant of a mismatch at the second locus by any method of tolerance induction, e.g., by administering donor bone marrow tissue to the recipient to induce tolerance to donor antigens expressed on the donor bone marrow, by expressing an MHC antigen of the donor from a stem cell of the recipient to induce tolerance to the donor antigen, or by altering the immunological properties of the graft, e.g., by masking, cleaving, or otherwise modifying cell surface molecules on the graft.
  • tolerance induction e.g., by administering donor bone marrow tissue to the recipient to induce tolerance to donor antigens expressed on the donor bone marrow, by expressing an MHC antigen of the donor from a stem cell of the recipient to induce tolerance to the donor antigen, or by altering the immunological properties of the graft, e.g., by masking, cleaving, or otherwise modifying cell surface molecules on the graft.
  • any of the methods which can be used to match or induce tolerance to the second locus can be used to match or induce tolerance to a third locus which affects graft rejection, e.g., an MHC class I or II locus, or a minor antigen locus.
  • the recipient and donor are matched at a class II locus and the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on the graft.
  • tolerance to a class II antigen is induced by a method other than a short course of a help reducing treatment, and the short course of help reducing treatment induces tolerance to unmatched class I and minor antigens on the graft.
  • the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen (in humans this is usually 8-12 days, preferably about 10 days); in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for mature T cells of the recipient to initiate rejection of an antigen after first being stimulated by the antigen.
  • the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g. in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient.
  • the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone.
  • the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative, or the short course is postoperative; or the donor and recipient are class I matched.
  • Methods of inducing tolerance by a short-term administration of a help reducing agent e.g., a short high dose course of cyclosporine A (CsA)
  • CsA cyclosporine A
  • methods for inducing tolerance e.g., methods for the implantation of transduced bone marrow cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 008/126,122, filed on Sep. 23, 1993.
  • the invention features a method of inducing tolerance in a recipient mammal, e.g., a primate, e.g., a human, of a first species to a graft from a mammal, e.g., a swine, e.g., a miniature swine, of a second species, which graft preferably expresses a major histocompatibility complex (MHC) antigen.
  • MHC major histocompatibility complex
  • the method includes inserting DNA encoding an MHC antigen of the second species into a hematopoietic stem cell, e.g., a bone marrow hematopoietic stem cell, of the recipient mammal; allowing the MHC antigen encoding DNA to be expressed in the recipient; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine treatment.
  • the short course of help reducing treatment is generally administered at about the time the graft is introduced into the recipient.
  • the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on a graft which is introduced into the recipient subsequent to expression of the MHC antigen.
  • the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient.
  • the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone.
  • the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative; or the short course is postoperative.
  • Preferred embodiments include those in which: the cell is removed from the recipient mammal prior to the DNA insertion and returned to the recipient mammal after the DNA insertion; the DNA is obtained from the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is syngeneic with the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is MHC matched, and preferably identical, with the individual mammal from which the graft is obtained; the DNA includes an MHC class I gene; the DNA includes an MHC class II gene; the DNA is inserted into the cell by transduction, e.g., by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient for at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of: prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the step of, prior to implantation of a graft, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species.
  • an organ e.g., a liver or a kidney
  • In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.
  • the method further includes, prior to hematopoietic stem cell transplantation, introducing into the recipient an antibody capable of binding to mature T cells of said recipient mammal.
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a liver or a kidney.
  • the invention features a method of inducing tolerance in a recipient mammal, preferably a primate, e.g., a human, to a graft obtained from a donor of the same species, which graft preferably expresses an MHC antigen.
  • a primate e.g., a human
  • the method includes: inserting DNA encoding an MHC antigen of the donor into a hematopoietic stem cell, e.g., bone marrow hematopoietic stem cell, of the recipient; allowing the MHC antigen encoding DNA to be expressed in the recipient; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine.
  • the short course of help reducing treatment is generally administered at about the time the graft is introduced into the recipient.
  • the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on a graft which is introduced into the recipient subsequent to expression of the MHC antigen.
  • the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient.
  • the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone
  • the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative, or the short course is postoperative; or the donor and recipient are class I matched.
  • Preferred embodiments include those in which: the cell is removed from the recipient prior to the DNA insertion and returned to the recipient after the DNA insertion; the DNA includes a MHC class I gene; the DNA includes a MHC class II gene; the DNA is inserted into the cell by transduction, e.g. by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • the method further includes, prior to hematopoietic stem cell transplantation, introducing into the recipient an antibody capable of binding to mature T cells of said recipient mammal.
  • the graft is a liver or a kidney.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of: prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the step of, prior to implantation of a graft, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species.
  • an organ e.g., a liver or a kidney
  • In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.
  • Methods of inducing tolerance with a short-term administration of a help reducing agent e.g., a short high dose course of cyclosporine A (CsA)
  • CsA cyclosporine A
  • methods of inducing tolerance which use the implantation of donor stem cells to induce tolerance to an antigen e.g., the methods described in U.S. Ser. No. 07/838,595, filed Feb. 19, 1992.
  • the invention features a method of inducing tolerance in a recipient mammal of a first species, e.g., a primate, e.g., a human, to a graft obtained from a mammal of a second, preferably discordant species, e.g., a swine, e.g., a miniature swine, or a discordant primate species.
  • a first species e.g., a primate, e.g., a human
  • a graft obtained from a mammal of a second, preferably discordant species, e.g., a swine, e.g., a miniature swine, or a discordant primate species.
  • the method includes: preferably prior to or simultaneous with transplantation of the graft, introducing, e.g., by intravenous injection, into the recipient mammal, hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, of the second species (preferably the hematopoietic stem cells home to a site in the recipient mammal); (optionally) inactivating the natural killer cells of the recipient mammal, e.g., by prior to introducing the hematopoietic stem cells into the recipient mammal, introducing into the recipient mammal an antibody capable of binding to natural killer cells of said recipient mammal; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine.
  • the short course of help reducing treatment is generally administered at the time at the graft is introduced into the recipient.
  • the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on the graft which is introduced into the recipient.
  • the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times, the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient.
  • the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone.
  • the help reducing treatment is begun before or at about the time the graft is introduced; or the short course is perioperative, the short course is postoperative.
  • the hematopoietic cells prepare the recipient for the graft that follows, by inducing tolerance at both the B-cell and T-cell levels.
  • hematopoietic cells are fetal liver or spleen, or bone marrow cells, including immature cells (i.e., undifferentiated hematopoietic stem cells; these desired cells can be separated out of the bone marrow prior to administration), or a complex bone marrow sample including such cells can be used.
  • anti-NK antibody is anti-human thymocyte polyclonal anti-serum.
  • a second anti-mature T cell antibody can be administered as well, which lyses T cells as well as NK cells. Lysing T cells is advantageous for both bone marrow and xenograft survival.
  • Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable.
  • Monoclonal preparations can be used in the methods of the invention.
  • stromal tissue preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells; the hematopoietic stem cells are introduced simultaneously with, or prior to, the antibody.
  • the same mammal of the second species is the donor of one or both the graft and the hematopoietic cells; and the antibody is an anti-human thymocyte polyclonal anti-serum, obtained, e.g., from a horse or pig.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of: prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • the method further includes, prior to hematopoietic stem cell transplantation, introducing into the recipient an antibody capable of binding to mature T cells of said recipient mammal.
  • the method further includes inactivating T cells of the recipient, e.g., by, prior to introducing the hematopoietic stem cells into the recipient, introducing into the recipient an antibody capable of binding to T cells of the recipient.
  • the method includes the step of introducing into the recipient a graft obtained from the donor which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • the invention features a method of inducing tolerance in a recipient mammal, preferably a primate, e.g., a human, to a graft obtained from a donor, e.g., of the same species.
  • the method includes: preferably prior to or simultaneous with transplantation of the graft, introducing, e.g., by intravenous injection, into the recipient, hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, of a mammal, preferably the donor (preferably the hematopoietic stem cells home to a site in the recipient); (optionally), inactivating T cells of the recipient, e.g., by, prior to introducing the hematopoietic stem cells into the recipient, introducing into the recipient an antibody capable of binding to T cells of the recipient; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment,
  • the short course of help reducing treatment induces tolerance to unmatched class I and minor antigens on the graft which is introduced into the recipient.
  • the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient.
  • the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone
  • the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative, the short course is postoperative; the donor and recipient are class I matched.
  • the hematopoietic stem cells are introduced simultaneously with, or prior to administration of the antibody; the antibody is an antihuman thymocyte polyclonal anti-serum; and the anti-serum is obtained from a horse or pig.
  • NK cells of the recipient include: the further step of, prior to hematopoietic stem cell transplantation, inactivating or depleting NK cells of the recipient, e.g., by introducing into the recipient mammal an antibody capable of binding to NK cells of the recipient mammal; and those in which the same individual is the donor of both the graft and the bone marrow.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of, prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the further step of, prior to bone marrow transplantation, adsorbing natural antibodies from the blood of the recipient by hemoperfusing an organ, e.g., the liver, or a kidney, obtained from the donor.
  • an organ e.g., the liver, or a kidney
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • donor species specific stromal tissue preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • the method includes the step of introducing into the recipient, a graft which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • Methods of inducing tolerance with short-term administration of a help reducing agent can be combined with yet other methods for inducing tolerance, e.g., with: methods which use the implantation of a xenogeneic thymic graft to induce tolerance, e.g., the methods described in U.S. Ser. No. 08/163, 912 filed on Dec. 7, 1993; methods of increasing the level of the activity of a tolerance promoting or GVHD inhibiting cytokine or decreasing the level of activity of a tolerance inhibiting or GVHD promoting cytokine, e.g., the methods described in U.S. Ser. No.
  • an immunosuppressant e.g., cyclosporine
  • the invention features a method of diminishing or inhibiting T cell activity, preferably the activity of thymic or lymph node T cells, in a recipient mammal, e.g., a primate, e.g., a human, which receives a graft from a donor mammal.
  • the method includes, inducing tolerance to the graft; administering to the recipient a short course of an immunosuppressive agent, e.g., cyclosporine, sufficient to inactivate T cells, preferably thymic or lymph node T cells; and preferably transplanting the graft into the recipient.
  • an immunosuppressive agent e.g., cyclosporine
  • Tolerance to the graft can be induced by any method, e.g., by any of the methods discussed herein.
  • tolerance can be induced by the administration of donor allogeneic or xenogeneic hematopoietic stem cells, the administration of genetically engineered autologous stem cells, by the administration of a short course of a help reducing agent, or by altering the immunological properties of the graft, e.g., by masking, cleaving, or otherwise modifying cell surface molecules of the graft.
  • the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period.
  • the short course is begun before or at about the time the treatment to induce tolerance is begun, e.g., at about the time, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient; the short course begins on the day the treatment to induce tolerance is begun, e.g., on the day, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the treatment to induce tolerance is begun, e.g., within 1, 2, 4, 6, 8, or 10 days before or after xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient.
  • the short course of an immunosuppressive is administered in conjunction with an anti-T cell antibody; the short course of an immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG antibody, or similar, preparations.
  • T cells e.g., thymic or lymph node T cells
  • the recipient mammal is other than a mouse or rat.
  • Methods of inactivating T cells, preferably thymic or lymph node T cells, of the invention can be combined with methods of inducing tolerance in which the inactivation of T cells is desirable.
  • the anti-T cell methods of the invention can be used in place of, or in addition to, methods for the inactivation of T cells called for, or useful in such methods of inducing tolerance.
  • anti-thymic or lymph node T cell methods of the invention can be used with methods for the implantation of transduced bone marrow cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 008/126,122, filed on Sep. 23, 1993.
  • the invention features a method of promoting, in a recipient mammal of a first species, the acceptance of a graft from a donor mammal of a second species, which graft, preferably, expresses a major histocompatibility complex (MHC) antigen.
  • MHC major histocompatibility complex
  • the method includes inserting DNA encoding an MHC antigen of the second species into a hematopoietic stem cell, e.g., a bone marrow hematopoietic stem cell, of the recipient mammal; allowing the MHC antigen encoding DNA to be expressed in the recipient; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells. (Thymic or lymph node T cells might otherwise inhibit the survival of the graft or engineered cells.)
  • a hematopoietic stem cell e.g., a bone marrow hematopoietic stem cell
  • an immunosuppressive agent e.g., a short course of cyclosporine treatment
  • the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period.
  • the recipient mammal is a primate, e.g., a human
  • the donor mammal is a swine, e.g., a miniature swine.
  • the short course is begun before or at about the time genetically engineered stem cells are introduced into the recipient; the short course begins on the day the genetically engineered stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the genetically engineered stem cells are introduced into the recipient.
  • the short course of an immunosuppressive agent is administered in conjunction with an anti-T cell antibody; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG, or similar, antibody preparations.
  • T cells e.g., thymic or lymph node T cells
  • Preferred embodiments include those in which: the cell is removed from the recipient mammal prior to the DNA insertion and returned to the recipient mammal after the DNA insertion; the DNA is obtained from the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is syngeneic with the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is MHC matched, and preferably identical, with the individual mammal from which the graft is obtained; the DNA includes an MHC class I gene; the DNA includes an MHC class II gene; the DNA is inserted into the cell by transduction, e.g., by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient for at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell implantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • low dose e.g., between about 100 and 400 rads
  • the method further includes the administration of thymic irradiation to the recipient, e.g., 700 rads of thymic irradiation.
  • Other preferred embodiments include: the step of depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species.
  • an organ e.g., a liver or a kidney
  • In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a liver or a kidney.
  • the invention features a method of promoting, in a recipient mammal, preferably a primate, e.g., a human, acceptance of a graft obtained from a donor of the same species, which graft expresses an MHC antigen.
  • a primate e.g., a human
  • the method includes: inserting DNA encoding an MHC antigen of the donor into a hematopoietic stem cell, e.g., a bone marrow hematopoietic stem cell, of the recipient; allowing the MHC antigen encoding DNA to be expressed in the recipient; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells. (Thymic or lymph node T cells might otherwise inhibit the survival of the graft or engineered cells.)
  • an immunosuppressive agent e.g., a short course of cyclosporine treatment
  • the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period.
  • the short course is begun before or at about the time genetically engineered stem cells are introduced into the recipient; the short course begins on the day the genetically engineered stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the genetically engineered stem cells are introduced into the recipient.
  • the short course of an immunosuppressive agent is administered in conjunction with an anti-T cell antibody; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG, or similar, antibody preparations.
  • T cells e.g., thymic or lymph node T cells
  • Preferred embodiments include those in which: the cell is removed from the recipient prior to the DNA insertion and returned to the recipient after the DNA insertion; the DNA includes a MHC class I gene; the DNA includes a MHC class II gene; the DNA is inserted into the cell by transduction, e.g. by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell implantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • low dose e.g., between about 100 and 400 rads
  • the method further includes the administration of thymic irradiation to the recipient, e.g., 700 rads of thymic irradiation.
  • Other preferred embodiments include: the step of depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species.
  • an organ e.g., a liver or a kidney
  • In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a liver or a kidney.
  • Methods of inactivating T cells, preferably thymic or lymph node T cells, of the invention can be combined with methods of inducing tolerance which use the implantation of donor stem cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, hereby incorporated by reference.
  • the invention features a method of promoting, in a recipient mammal of a first species, e.g., a primate, e.g., a human, acceptance of a graft obtained from a mammal of a second, preferably discordant species, e.g., a swine, e.g., a miniature swine, or a discordant primate species.
  • a first species e.g., a primate, e.g., a human
  • discordant species e.g., a swine, e.g., a miniature swine, or a discordant primate species.
  • the method includes: introducing, e.g., by intravenous injection, into the recipient mammal, hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, of the second species (preferably the hematopoietic stem cells home to a site in the recipient mammal); (optionally) inactivating natural killer cells of the recipient mammal, e.g., by, prior to introducing the hematopoietic stem cells into the recipient mammal, introducing into the recipient mammal an antibody capable of binding to natural killer cells of said recipient mammal; (optionally) inactivating T cells of the recipient mammal, e.g., by, prior to introducing the hematopoietic stem cells into the recipient mammal, introducing into the recipient mammal an antibody capable of binding to T cells of the recipient mammal; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g.,
  • the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period mentioned above.
  • the short course is begun before or at about the time stem cells are introduced into the recipient; the short course begins on the day the stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the stem cells are introduced into the recipient.
  • the short course of an immunosuppressive agent is administered in conjunction with one or both of an anti-T cell antibody, or thymic irradiation, e.g., 700 rads of thymic irradiation; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG antibody preparations.
  • the hematopoietic cells prepare the recipient for the graft that follows, by inducing tolerance at both the B-cell and T-cell levels.
  • hematopoietic cells are fetal liver or spleen, or bone marrow cells, including immature cells (i.e., undifferentiated hematopoietic stem cells; these desired cells can be separated out of the bone marrow prior to administration), or a complex bone marrow sample including such cells can be used.
  • anti-NK antibody is anti-human thymocyte polyclonal anti-serum.
  • a second anti-mature T cell antibody can be administered as well, which lyses T cells as well as NK cells. Lysing T cells is advantageous for both bone marrow and xenograft survival.
  • Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable.
  • Monoclonal preparations can be used in the methods of the invention.
  • stromal tissue preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells; the hematopoietic stem cells are introduced simultaneously with, or prior to, an anti-NK or T cell antibody.
  • the same mammal of the second species is the donor of one or both the graft and the hematopoietic cells; and the anti-T or anti-NK cell antibody is an anti-human thymocyte polyclonal anti-serum, obtained, e.g., from a horse or pig.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • low dose e.g., between about 100 and 400 rads
  • the method further includes the administration of thymic irradiation to the recipient, e.g., 300 to 700 rads of thymic irradiation.
  • Other preferred embodiments include: the step of prior to hematopoietic stem cell transplantation, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a graft which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • the stem cells are introduced into the recipient prior to or simultaneous with transplantation of the graft.
  • the invention features a method of promoting, in a recipient mammal, preferably a primate, e.g., a human, acceptance of a graft obtained from a donor of the same species.
  • the method includes: introducing, e.g., by intravenous injection into the recipient, hematopoietic stem cells, e.g.
  • bone marrow cells or fetal liver or spleen cells, of a mammal preferably the donor (preferably the hematopoietic stem cells home to a site in the recipient); (optionally) inactivating T cells of the recipient, e.g., by, prior to introducing the hematopoietic stem cells into the recipient, introducing into the recipient an antibody capable of binding to T cells of the recipient; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells.
  • an immunosuppressive agent e.g., a short course of cyclosporine treatment
  • the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period mentioned above.
  • the short course is begun before or at about the time stem cells are introduced into the recipient; the short course begins on the day the stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the stem cells are introduced into the recipient.
  • the short course of an immunosuppressive agent is administered in conjunction with one or both of an anti-T cell antibody, or thymic irradiation, e.g., 700 rads of thymic irradiation; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG antibody preparations.
  • the anti-T cell or NK cell antibody is an antihuman thymocyte polyclonal anti-serum; and the anti-serum is obtained from a horse or pig.
  • NK cells e.g., by introducing into the recipient mammal an antibody capable of binding to NK cells of the recipient mammal; and those in which the same individual is the donor of both the graft and the bone marrow.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • low dose e.g., between about 100 and 400 rads
  • the method further includes administering thymic irradiation to the recipient, e.g., 700 rads of thymic irradiation.
  • Other preferred embodiments include: the further step of, prior to bone marrow transplantation, adsorbing natural antibodies from the blood of the recipient by hemoperfusing an organ, e.g., the liver, or a kidney, obtained from the donor.
  • an organ e.g., the liver, or a kidney
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • donor species specific stromal tissue preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • Other preferred embodiments further include the step of introducing into the recipient, a graft obtained from the donor, e.g., a graft which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • the stem cells are introduced into the recipient prior to or simultaneous with transplantation of the graft.
  • Methods of inactivating T cells, preferably thymic or lymph node T cells, of the invention can be used with yet other methods of inducing tolerance in which the inactivation of thymic or lymph node T cells is desirable.
  • anti-thymic or lymph node T cell methods of the invention can be used with: methods which use the implantation of a xenogeneic thymic graft to induce tolerance, e.g., the methods described in U.S. Ser. No. 08/163, 912 filed on Dec.
  • Thymus-function deficient refers to a condition in which the ability of an individual's thymus to support the maturation of T cells is impaired as compared with a normal individual. Thymus deficient conditions include those in which the thymus or thymus function is essentially absent.
  • Tolerance refers to the inhibition of a graft recipient's ability to mount an immune response, e.g., to a donor antigen, which would otherwise occur, e.g., in response to the introduction of a non self MHC antigen into the recipient. Tolerance can involve humoral, cellular, or both humoral and cellular responses. The concept of tolerance includes both complete and partial tolerance. In other words, as used herein, tolerance include any degree of inhibition of a graft recipient's ability to mount an immune response, e.g., to a donor antigen.
  • a discordant species combination refers to two species in which hyperacute rejection occurs when vascular organs are grafted. Generally, discordant species are from different orders, while non-discordant species are from the same order. For example, rats and mice are non-discordant species, i.e. their MHC antigens are substantially similar, and they are members of the same order, rodentia.
  • Hematopoietic stem cell refers to a cell that is capable of developing into mature myeloid and/or lymphoid cells.
  • a hematopoietic stem cell is capable of the long-term repopulation of the myeloid and/or lymphoid lineages.
  • Stem cells derived from the cord blood of the recipient or the donor can be used in methods of the invention. See U.S. Pat. No. 5,192,553, hereby incorporated by reference, and U.S. Pat. No. 5,004,681, hereby incorporated by reference.
  • Miniature swine refers to completely or partially inbred miniature swine.
  • “Graft”, as used herein, refers to a body part, organ, tissue, cells, or portions thereof
  • “Stromal tissue”, as used herein, refers to the supporting tissue or matrix of an organ, as distinguished from its functional elements or parenchyma.
  • Restoring, inducing, or promoting immunocompetence means one or both of: (1) increasing the number of mature functional T cells in the recipient (over what would be seen in the absence of treatment with a method of the invention) by either or both, increasing the number of recipient-mature functional T cells or by providing mature functional donor-T cells, which have matured in the recipient; or (2) improving the immune-responsiveness of the recipient, e.g., as is measured by the ability to mount a skin response to a recall antigen, or improving the responsiveness of a of a T cell of the recipient, e.g., as measured by an in vitro test, e.g., by the improvement of a proliferative response to an antigen, e.g., the response to tetanus antigen or to an alloantigen.
  • a mature functional T cell is a T cell (of recipient or donor origin) which responds to microbial antigens and tolerant to recipient and donor tissue.
  • Lymph node or thymic T cell refers to T cells which are resistant to inactivation by traditional methods of T cell inactivation, e.g., inactivation by a single intravenous administration of anti-T cell antibodies, e.g., anti-bodies, e.g., ATG preparation.
  • Restoring or inducing the thymus-dependent ability for T cell progenitors to mature into mature T cells means either or both, increasing the number of functional mature T cells of recipient origin in a recipient, or providing mature functional donor T cells to a recipient, by providing donor thymic tissue in which T cells can mature.
  • the increase can be partial, e.g., an increase which does not bring the level of mature functional T cells up to a level which results in an essentially normal immune response or partial, e.g., an increase which falls short of bringing the recipient's level of mature functional T cells up to a level which results in an essentially normal immune response.
  • Methods of the invention will allow the induction of immunocompetence in patients suffering from an immunodeficiency, e.g., a T cell deficiency, e.g., a thymic based immunodeficiency, e.g., a congenital immunodeficiency due to thymic aplasia or dysfunction, an acquired immune disorder, e.g., AIDS, immunoincompetence resulting form a neoplastic disease, or immunoincompetence resulting from a medical procedure, e.g., chemotherapy or radiation treatment.
  • An acquired immune deficiency is one which is due primarily to other than genetic defects.
  • At risk for AIDS refers to being HIV positive or having AIDS.
  • An immunosuppressive agent capable of inactivating thymic or lymph node T cells is an agent, e.g., a chemical agent, e.g., a drug, which, when administered at an appropriate dosage, results in the inactivation of thymic or lymph node T cells.
  • agents e.g., cyclosporine, FK-506, and rapamycin.
  • Anti-T cell antibodies because they are comparatively less effective at inactivating thymic or lymph node T cells, are not preferred for use as agents.
  • An agent should be administered in sufficient dose to result in significant inactivation of thymic or lymph node T cells which are not inactivated by administration of an anti-T cell antibody, e.g., an anti-ATG preparation.
  • Putative agents, and useful concentrations thereof can be prescreened by in vitro or in vivo tests, e.g., by administering the putative agent to a test animal, removing a sample of thymus or lymph node tissue, and testing for the presence of active T cells in an in vitro or in vivo assay. Such prescreened putative agents can then be further tested in transplant assays.
  • “Short course of a immunosuppressive agent”, as used herein, means a transitory non-chronic course of treatment.
  • the treatment should begin before or at about the time the treatment to induce tolerance is begun, e.g., at about the time, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient.
  • the short course can begin on the day the treatment to induce tolerance is begun, e.g., on the day, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient or the short course can begin within 1, 2, 4, 6, 8, or 10 days before or after the treatment to induce tolerance is begun, e.g., within 1, 2, 4, 6, 8, or 10 days before or after xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient.
  • the short course can last for: a period equal to or less than about 8-12 days, preferably about 10 days, or a time which is approximately equal to or is less than two, three, four, five, or ten times the 8-12 or 10 day period. Optimally, the short course lasts about 30 days.
  • the dosage should be sufficient to maintain a blood level sufficient to inactivate thymic or lymph node T cells. A dosage of approximately 15 mg/kg/day has been found to be effective in primates.
  • Help reduction means the reduction of T cell help by the inhibition of the release of at least one cytokine, e.g., any of IL-2, IL-4, IL-6, gamma interferon, or TNF, from T cells of the recipient at the time of the first exposure to an antigen to which tolerance is desired.
  • the inhibition induced in a recipient's T cell secretion of a cytokine must be sufficient such that the recipient is tolerized to an antigen which is administered during the reduction of help.
  • the level of reduction is one which substantially eliminates the initial burst of IL-2 which accompanies the first recognition of a foreign antigen but which does not eliminate all mature T cells, which cells may be important in educating and producing tolerance.
  • a help reducing agent is an agent, e.g., an immunosuppressive drug, which results in the reduction of cytokine release.
  • help reducing agents are cyclosporine, FK-506, and rapamycin.
  • Anti-T cell antibodies because they can eliminate T cells, are not preferred for use as help reducing agents.
  • a help reducing agent must be administered in sufficient dose to give the level of inhibition of cytokine release which will result in tolerance.
  • the help reducing agent should be administered in the absence of treatments which promote cytokine, e.g., IL-2, release.
  • Putative agents help reducing agents can be prescreened by in vitro or in vivo tests, e.g., by contacting the putative agent with T cells and determining the ability of the treated T cells to release a cytokine, e.g., IL-2. The inhibition of cytokine release is indicative of the putative agent's efficacy as a help reducing agent.
  • Such prescreened putative agents can then be further tested in a kidney transplant assay.
  • a putative help reducing agent is tested for efficacy by administering the putative agent to a recipient monkey and then implanting a kidney from a class II matched class I and minor antigen mismatched donor monkey into the recipient. Tolerance to the donor kidney (as indicated by prolonged acceptance of the graft) is indicative that the putative agent is, at the dosage tested, a help reducing agent.
  • “Short course of a help reducing agent”, as used herein, means a transitory non-chronic course of treatment.
  • the treatment should begin before or at about the time of transplantation of the graft.
  • the treatment can begin before or at about the time of the recipient's first exposure to donor antigens.
  • the treatment lasts for a time which is approximately equal to or less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • the duration of the treatment can be extended to a time approximately equal to or less than two, three, four, five, or ten times, the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • the duration will usually be at least equal to the time required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • about 12 days of treatment is sufficient.
  • Experiments with cyclosporine A (10 mg/kg) in pigs show that 6 days is not sufficient.
  • Other experiments in monkeys show that IL-2 administered on day 8, 9, or 10 of cyclosporine A treatment will result in rejection of the transplanted tissue. Thus, 8, 9, or 10 days is probably not sufficient in pigs.
  • a dose of 10 mg/kg cyclosporine with a blood level of about 500-1,000 ng/ml is sufficient to induce tolerance to class II matched class I and minor antigen mismatched kidneys.
  • the same blood level, 500-1,000 ng/ml is sufficient to induce tolerance in pigs.
  • Long-term administration of 5 mg/kg prevents rejection (by long term immune suppression) but does not result in tolerance.
  • the help suppressing methods of the invention avoid the undesirable side effects of long-term or chronic administration of the broad spectrum immune suppressants often used in transplantation.
  • Long-term or chronic administration of drugs such as Prednisone, Imuran, CyA, and, most recently FK506, have all had an important impact on the field of transplantation.
  • all of these drugs cause nonspecific suppression of the immune system which must be titrated sufficiently to avoid rejection while not completely eliminating immune function.
  • Patients who must stay on chronic immunosuppressive therapy for the remainder of their lives face major complications arising from too much or too little immunosuppression, causing infection and rejection, respectively.
  • the help suppressing methods of the invention are based on the administration of a transitory short term high dose course of a help reducing treatment.
  • MHC antigen refers to a protein product of one or more MHC genes; the term includes fragments or analogs of products of MHC genes which can evoke an immune response in a recipient organism.
  • MHC antigens include the products (and fragments or analogs thereof) of the human MHC genes, i.e., the HLA genes.
  • MHC antigens in swine e.g., miniature swine, include the products (and fragments and analogs thereof) of the SLA genes, e.g., the DRB gene.
  • Recipient thymic or lymph node T cells are responsible for significant resistance to implanted grafts, e.g., to transplanted hematopoietic cells or transplanted organs. It has been found that the usual methods of T cell depletion or inactivation, e.g., the administration of anti-T cell antibodies, often fall short of an optimum level of T cell depletion or inactivation. In particular, such methods fail to provide optimum levels of depletion or inactivation of thymic or lymph node T cells.
  • Methods of the invention in which a short course of an immunosuppressant, e.g., cyclosporine, capable of inactivating recipient thymic or lymph node T cells is administered to the recipient result in more thorough inactivation of thymic or lymph node T cells and thus in improved acceptance of graft tissue.
  • an immunosuppressant e.g., cyclosporine
  • the retroviral methods of the invention allow the reconstitution of a graft recipient's bone marrow with transgenic autologous bone marrow cells expressing allogeneic or xenogeneic MHC genes. Expression of the transgenic MHC genes confers tolerance to grafts which exhibit the products of these or closely related MHC genes. Thus, these methods provide for the induction of specific transplantation tolerance by somatic transfer of MHC genes. Retroviral methods of the invention avoid the undesirable side effects of broad spectrum immune suppressants which are often used in transplantation.
  • BMT bone marrow transplantation
  • GVHD graft versus host disease
  • Retroviral methods of the invention which induce specific tolerance by reconstitution of the recipient's bone marrow with autologous (as opposed to allogeneic or heterologous) bone marrow cells, allow tolerance to be conferred with minimal risk of GVHD and with minimal need to remove T cells from the marrow inoculum.
  • Retroviral methods of the invention can be combined with the help suppression and T cell inactivation methods of the invention to prolong graft acceptance.
  • Hematopoietic cell transplant methods of the invention avoid the undesirable side effects of broad spectrum immune suppressants which are often used in transplantation. Hematopoietic cell transplant methods of the invention can be combined with the help suppression and T cell elimination methods of the invention to prolong graft acceptance.
  • FIG. 1A is a graph showing growth of fetal pig THY/LIV graft vs. time after transplantation in the presence of mature mouse T cells in the periphery.
  • FIG. 1B is a dot plot analysis of live peripheral white blood cells of a representative animal 16 weeks post-transplant. Upper left quadrant 0.5%; upper right quadrant 12.1%.
  • FIG. 2 is a graph of mouse anti-pig mixed lymphocyte reactions (MLR's) performed to determine whether or not mouse T cells which matured in pig thymus grafts were tolerant to pig antigens.
  • MLR's mouse anti-pig mixed lymphocyte reactions
  • FIG. 3 is a diagram of the GS4.5 retroviral construct.
  • FIG. 4 is a diagram of the GS4.5 proviral genome and the expected transcripts.
  • FIGS. 5 a and 5 b are representations of flow cytometry profile of transduced cells.
  • FIG. 6 is a diagram of the transduction assay.
  • FIG. 7 is a diagram of genetic maps of the C57BL/10, B10.AKM, and B10.MBR strains.
  • FIG. 8 is a diagram of the FACS profile of spleen cells from a recipient of transduced bone marrow.
  • FIGS. 9 a and 9 b are graphs of survival versus time in skin graft experiments.
  • FIGS. 10 a - d are diagrams of FACS analysis of thymocytes from graft rejecters, and controls.
  • FIG. 11 is a diagram of the N2-B19-H2b vector.
  • the following procedure was designed to promote the acceptance of a xenograft thymus by a host and thusly to either or both: 1, lengthen the time an implanted organ (a xenograft) survives in a xenogeneic host prior to rejection; and 2, provide xenogeneic thymic tissue in which host T cells can mature.
  • the organ can be any organ, e.g., a liver, e.g., a kidney, e.g., a heart.
  • the two main strategies are elimination of natural antibodies and transplantation of thymic tissue to induce tolerance.
  • Preparation of the recipient for either organ transplantation or thymus replacement includes any or all of the following steps. Preferably they are carried out in the following sequence.
  • a preparation of horse anti-human thymocyte globulin is intravenously injected into the recipient.
  • the antibody preparation eliminates mature T cells and natural killer cells. If not eliminated, mature T cells might promote rejection of both the thymic transplant and, after sensitization, the xenograft organ.
  • the ATG preparation also eliminates natural killer (NK) cells. NK cells probably have no effect on an implanted organ, but might act immediately to reject the newly introduced thymic tissue.
  • Anti-human ATG obtained from any mammalian host can also be used, e.g., ATG produced in pigs, although thus far preparations of pig ATG have been of lower titer than horse-derived ATG.
  • ATG is superior to anti-NK monoclonal antibodies, as the latter are generally not lytic to all host NK cells, while the polyclonal mixture in ATG is capable of lysing all host NK cells.
  • Anti-NK monoclonal antibodies can, however, be used. In a relatively severely immunocompromised individual this step may not be necessary.
  • host (or donor) T cells mature in the xenogeneic thymus they will be tolerant of the xenogeneic thymic tissue. Alternatively, as the host immune system is progressively restored, it may be desirable to treat the host to induce tolerance to the xenogeneic thymic tissue.
  • the recipient can be thymectomized.
  • recipient T cells do not have an opportunity to differentiate in the recipient thymus, but must differentiate in the donor thymus. In some cases it may be necessary to splenectomize the recipient in order to avoid anemia.
  • the recipient can be administered low dose radiation.
  • this step is thought to be beneficial in bone marrow transplantation (by creating hematopoietic space for newly injected bone marrow cells), it is of less importance in thymic grafts which are not accompanied by bone marrow transplantation.
  • a sublethal dose e.g., a dose about equal to 100, or more than 100 and less than about 400, rads, whole body radiation, plus 700 rads of local thymic radiation, can be used.
  • natural antibodies can be adsorbed from the recipient's blood. (This is of more importance in organ grafts but can be used in thymus replacement procedures as well.) Antibody removal can be accomplished by exposing the recipient's blood to donor or donor species antigens, e.g., by hemoperfusion of a liver of the donor species to adsorb recipient-natural antibodies. Pre-formed natural antibodies (nAb) are the primary agents of graft rejection. Natural antibodies bind to xenogeneic endothelial cells and are primarily of the IgM class. These antibodies are independent of any known previous exposure to antigens of the xenogeneic donor.
  • B cells that produce these natural antibodies tend to be T cell-independent, and are normally tolerized to self antigen by exposure to these antigens during development.
  • the mechanism by which newly developing B cells are tolerized is unknown.
  • the liver is a more effective adsorber of natural antibodies than the kidney. Again, this step may not be required, at least initially, in a relatively severely immunocompromised patient.
  • Donor thymic tissue preferably fetal or neonatal thymic tissue is implanted in the recipient. Fetal or neonatal liver or spleen tissue can be included.
  • Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • the donor of the implant and the individual that supplies the tolerance-inducing thymic graft should be the same individual or should be as closely related as possible. For example, it is preferable to derive implant tissue from a colony of donors that is highly or completely inbred.
  • the donor of the organ used for perfusion need not be closely related to the donor of the implant or thymic tissue.
  • Immunocompetent C57BL/10 (B10) mice were used to test the ability of pig thymus to induce specific tolerance to discordant pig antigens.
  • B10 mice were treated with a non-myeloablative conditioning regimen which has previously shown to permit induction of tolerance to rat xeno-antigens in mice, see e.g., Sharabi et al., 1990, J. Exp. Med. 172:195-202.
  • mice received depleting doses of anti-T cell and anti-NK cell mAbs, 7 Gy mediastinal irradiation and 3 Gy whole body irradiation (WBI), and then received fetal swine thymus/liver (THY/LIV) transplants under the kidney capsule followed by administration of 10 8 fetal liver cells (FLC) i.p.
  • Mice either received no further anti-T cell and anti-NK cell mAb treatments after 0 to 6 weeks post-tx, or were maintained on chronic mAb treatment for the duration of the experiment.
  • Swine THY/LIV grafts grew initially in treated euthymic mice, but stopped growing after T cell and natural killer (NK) cell-depleting monoclonal antibodies (mAbs) were discontinued, and these mice developed anti-pig IgG response.
  • NK natural killer
  • mAbs monoclonal antibodies
  • the grafts enlarged markedly and no anti-pig IgG response was observed.
  • Pig thymopoiesis was supported for at least 32 weeks during chronic mAb administration, although no pig T cells were detected in the periphery by flow cytometry (FCM).
  • Percentages of intra-graft CD4 + /CD8 ⁇ , CD4 ⁇ /CD8 + , CD4 + /CD8 + , and CD4 ⁇ /CD8 ⁇ pig thymocyte subsets were similar to those in normal pig thymus.
  • swine THY/LIV grafts grew markedly in adult thymectomized mice (ATX-THY/LIV) which received only a short (less than 6 weeks) course of mAb treatment post-transplant (post-tx).
  • FCM analysis of peripheral WBC in these mice 6 weeks after discontinuing mAb treatment revealed the presence of mature ( ⁇ -TCR hi ) mouse T cells.
  • ⁇ -TCR hi mature
  • the majority (more than 90%) of the ⁇ -TCR hi T cells were CD4 + /CD8 ⁇ .
  • FMC analyses 13 to 26 weeks post-tx demonstrated normal mouse thymocyte subsets in swine thymi. For example, 7.9% CD4 + /CD8 ⁇ , 2.9% CD4 ⁇ /CD8 + , 85.5% CD4 + /CD8 + , 3.7% CD4 ⁇ /CD8 ⁇ and 11.6% ⁇ -TCR hi thymocytes were found in a swine THY/LIV graft by FCM 17 weeks post-tx compared to 3.5% CD4 + /CD8 ⁇ , 3.2% CD4 ⁇ /CD8 + , 87.8% CD4 + /CD8 + , 5.5% CD4 ⁇ /CD8 ⁇ and 10.0% ⁇ -TCR hi thymocytes in a normal B10 thymus.
  • Fetal swine liver grafted without a thymus fragment did not grow in control nAb-treated ATX-B10 mice and ⁇ -TCR hi T cells did not appear in the periphery. Thus, the pig thymus was required for the development of mature mouse T cells.
  • mice were performed to determine whether or not mouse T cells which matured in pig thymus grafts were tolerant to pig antigens.
  • ATX-THY/LIV B10 mice H-2 b ) mounted no anti-B10 or anti-pig responses, but demonstrated normal allo-responses against a fully MHC-mismatched allogeneic stimulator, B10.BR (H-2 k ) ( FIG. 2 ).
  • fetal pig THY/LIV grafts were transplanted into both I-E + (BALB/c nude) and I-E ⁇ (ATX B10) recipients.
  • I-E + mice delete V ⁇ 11 T cells because of presentation in the thymus of an endogenous superantigen in association with I-E, whereas I-E ⁇ mice do not delete this T cell family.
  • the percentages of V ⁇ 11 T cells were therefore compared between I-E + and I-E ⁇ recipients of fetal pig thymus grafts in which murine T cells developed in pig thymi.
  • ATX B10 recipients were treated as described above.
  • BALB/c nude mice were depleted of NK cells and irradiated with 3 Gy WBI prior to transplant. These mice also developed large numbers of mature CD4 + T cells that migrated to the periphery. Complete deletion of V ⁇ 11 T cells was observed in the periphery of BALB/c nude recipients of fetal swine thymus grafts (Table I), indicating that mouse I-E also participated in negative selection of mouse T cells developing in pig thymi.
  • Negative selection is most likely carried out by murine Ia + dendritic cells which were detected predominantly in the cortico-medullary junction of swine thymus grafts by immunoperoxidase staining.
  • reduction in the percentage of V ⁇ 11 T cells was observed compared to normal B10 mice (mean 2.8% of T cells ⁇ 0.8 S.D., normal B10 5.2%, p ⁇ 0.005) suggesting that the pig SLA DR class II, which shares significant homology with mouse I-E class II, may participate in negative selection of mouse T cells developing in the pig thymus graft (Table I).
  • mice normal positive selection in grafted mice of a V ⁇ T cell family which is not deleted in I-E + or I-E ⁇ mice.
  • BALB/c nude mice were depleted of NK cells using rabbit anti-Asialo-Gm1 serum, and were given 3 Gy WBI, and fetal swine THY/LIV grafts implants under the kidney capsule, followed by injection of 10 8 FLC i.p. on Day 0.
  • ACK-lysed splenocytes or peripheral white blood cells red blood cells were removed by hypotonic shock
  • Murine F c R's were blocked using rat anti-mouse F c R mAb, 2.4G2, and then cells were stained with either fluoresceinated hamster anti-mouse V ⁇ 8.1/8.2 TCR (Pharmingen) or rat anti-mouse V ⁇ 11 TCR (Pharmingen) (green fluorescence) followed by phycoerythrin-conjugated rat anti-mouse CD4 and CD8 mAbs (Pharmingen) (orange fluorescence) and analyzed by two color FCM as described below.
  • Fluoresceinated murine mAb HOPCI with no known reactivity to mouse cells, or rat anti-mouse IgG1 (Zymed Laboratories, Inc.) was used as the negative control mAb in the green fluorescence.
  • Phycoerythrin-conjugated mAb Leu4 (Becton-Dickinson), was used as the negative control mAb in the orange fluorescence.
  • Approximately 5,000 gated CD4 + and CD8 + cells were usually collected for analysis of V ⁇ families. Non-viable cells were excluded using the vital nucleic acid stain, propidium iodide. Percentages of positive cells were determined as described below. Results are presented as the mean ⁇ SD of results obtained for individual mice.
  • CD4 + /CD8 ⁇ / ⁇ -TCR hi T cells which are released into the periphery are phenotypically normal, functional and tolerant to donor xeno-antigens, and to host antigens.
  • the lack of CD4 ⁇ /CD8 + / ⁇ -TCR hi repopulation in the periphery may be due to failure of mouse CD8 to interact with pig MHC class I molecules, as has been demonstrated for mouse anti-human responses, thereby preventing positive selection of CD8 + thymocytes by swine thymic epithelium. Since human CD8 + T cells are able to interact with pig MHC class I directly, human CD8 + T cells should mature effectively in swine fetal thymus grafts.
  • tolerance to pig antigens is not induced in euthymic mice which receive swine THY/LIV grafts because mouse T cell progenitors mature in the host thymus, which lacks the pig cells necessary to tolerize developing mouse thymocytes.
  • the non-myeloablative conditioning regimen used in this study permits engraftment of rat marrow and induction of donor-specific tolerance in murine recipients. Tolerance is thought to be induced in this model by rat dendritic cells detected at the cortico-medullary junction of the thymus of chimeric animals.
  • mice T cell progenitors home to the pig thymus graft and are tolerized pig antigens. No mouse TE is present in ATX hosts, but mouse dendritic cells are detectable in THY/LIV grafts and probably mediate the observed clonal deletion of cells reactive to host antigen.
  • V ⁇ 11 T cells in ATX B10 recipients of swine THY/LIV grafts suggests clonal deletion by swine cells, it is possible that there is a defect in the positive selection of this V ⁇ family on swine thymic stroma.
  • the normal percentages of V ⁇ 8.1/8.2 T cells in both ATX B10 and BALB/c nude recipients of swine THY/LIV grafts compared to those in normal B10 and BALB/c mice suggests that no defect in positive selection is present. The observed tolerance could be explained if the swine thymic stroma either clonally deletes or anergizes developing mouse thymocytes reactive to donor xeno-antigens.
  • mice were treated on a weekly basis post-tx with depleting doses of the same four mAbs for a period of 0-6 weeks. No difference in murine CD4 + T cell reconstitution or tolerance to pig antigens was observed in mice which were treated with no mAb post-tx compared to those which received 6 weeks mAb treatment post-tx. Some groups of control mice were maintained on chronic mAb treatment until the time of sacrifice.
  • ATX mice which received fetal swine liver grafts without a thymus fragment did not maintain their grafts, and developed less than 5% ⁇ -TRC + T cells in the periphery.
  • ATX mice were conditioned as described above. Exploratory laparotomies were performed at 5-6 and 15-19 weeks post-transplant to measure graft size. Mice were tail bled at regular intervals post-tx to obtain peripheral WBC which were prepared by hypotonic shock to remove red blood cells.
  • Percentages of positive cells were determined by subtracting the percentage of cells staining with the control mAb HOPC1 from the percentage of cells staining with the anti-mouse mAbs.
  • a dot plot analysis of live peripheral white blood cells of a representative animal 16 weeks post-tx (16 weeks after mAbs were discontinued) is shown in FIG. 2 .
  • 57% (27/47) of ATX mice treated with this protocol maintained swine grafts and reconstituted their CD4 + T cell compartment. In recent experiments this result was achieved in 90% (9/10) of mice treated with this regimen.
  • ATX-THY/LIV B10 H-2 b mice demonstrated specific unresponsiveness to pig antigens while maintaining normal allo-responsiveness to a fully MHC-mismatched B10.BR (H-2 k ) stimulator.
  • Specific unresponsiveness of B10 mice transplanted with fetal pig THY/LIV grafts to pig antigens in mixed lymphocyte reaction (MLR) was determined as follows. Control ATX-B10 mice which received a swine liver graft without a thymus fragment (ATX-LIV) mounted no responses to any stimulator, demonstrating the importance of the pig thymus graft in the development of functional mouse T cells.
  • Positive control anti-pig MLR was from a mouse immunized with a swine skin graft, since mice do not mount primary anti-pig responses.
  • Sterile splenocyte suspensions from normal B10 (right diagonal bar), normal B10 grafted with GG (SLA-I c /SLA-II d ) pig skin 12 weeks earlier (GG′-B10 solid bar), normal B10BR (crosshatched bar), and thymectomized B10 mice conditioned with the non-myeloablative regimen described above and transplanted with either a fetal pig (SLA-I d /SLA-II d ) THY/LIV graft (ATX-THY/LIV stippled bar), or a fetal pig liver graft only (ATX-LIV left diagonal bar) were ACK-lysed, washed and reconstituted in RPMI medium supplemented with 15% CPSR-2 (controlled processed serum replacement, Sigma), 4%
  • murine T cells are permitted to develop in a mouse thymus, they are not tolerized to pig antigens, and they reject pig thymus/liver grafts. (Thus, if the recipient has significant thymic function thymectomy is indicated.) If mouse T cells are continually depleted by mAb, swine thymopoiesis occurs in the swine thymus/liver graft. If a mouse is thymectomized but not chronically treated with anti-T cell mAb's, mouse thymopoiesis occurs in the pig thymus, and these cells are tolerized to pig antigens.
  • B10.BR full MHC mismatch to B10
  • C3H.SW minor antigen mismatch only skin grafts (1 mouse) were rejected by mouse T cells which had matured in a pig thymic graft, thus demonstrating their immunocompetence and ability to recognize minor antigens in a host MHC-restricted fashion.
  • the grafts were full thickness tail skin grafts on the upper thorax with a skin bridge separating them.
  • NK cells whole body irradiation, and thymic irradiation
  • inactivation or depletion of CD4 + cells is sufficient to allow growth of xenogeneic thymic tissue and maturation of host T cells in the xenogeneic thymic tissue.
  • the antibodies needed may differ depending on the species combination. E.g., in the case of a human recipient and a pig donor, because human CD8 will likely interact with pig class I molecules, it may also be necessary to administer anti-CD8 antibodies.
  • THY/LIV graft growth and host T cell development was seen in ATX mice treated with anti-CD4 antibodies and whole body irradiation.
  • B10 mice received 3 Gy whole body irradiation and fetal pig THY/LIV graft and 10 8 fetal liver cells in experiments essentially similar to those described in the previous section except that anti-CD4, anti-THY1.2, and anti NK cell antibodies were not administered.
  • TABLE II THY/LIV graft growth and T cell maturation does not require extensive antibody treatment.
  • the following procedure introduces donor thymic tissue and donor stem cells into the recipient and thus can be used to restore or induce immune function or to lengthen the time an implanted organ (a xenograft) survives in a xenogeneic host prior to rejection.
  • the organ can be any organ, e.g., a liver, e.g., a kidney, e.g., a heart.
  • the two main strategies are elimination of natural antibodies by organ perfusion, and transplantation of tolerance-inducing bone marrow.
  • Preparation of the recipient includes any or all of the following steps. Preferably they are carried out in the following sequence.
  • NK and T cells Elimination of NK and T cells.
  • ATG horse anti-human thymocyte globulin
  • the antibody preparation eliminates mature T cells and natural killer cells. If not eliminated, mature T cells would promote rejection of both the bone marrow transplant and, after sensitization, the xenograft itself of equal importance, the ATG preparation also eliminates natural killer (NK) cells. NK cells probably have no effect on the implanted organ, but would act immediately to reject the newly introduced bone marrow.
  • Anti-human ATG obtained from any ma host can also be used, e.g., ATG produced in pigs, although thus far preparations of pig ATG have been of lower titer than horse-derived ATG.
  • ATG is superior to anti-NK monoclonal antibodies, as the latter are generally not lytic to all host NK cells, while the polyclonal mixture in ATG is capable of lysing all host NK cells.
  • Anti-NK monoclonal antibodies can, however, be used.
  • Thymic tissue transplant In cases where the procedure is to restore or induce immunocompetence donor thymic tissue (preferably fetal or neonatal thymic tissue) is implanted in the recipient so that donor T cells (and recipient T cells if the are present and functional) can mature. Fetal or neonatal liver or spleen tissue can be implanted with the thymic tissue.
  • donor thymic tissue preferably fetal or neonatal thymic tissue
  • Fetal or neonatal liver or spleen tissue can be implanted with the thymic tissue.
  • the presence of donor antigen in the thymus during the time when host T cells are regenerating post-transplant is critical for tolerizing host T cells. If donor hematopoietic stem cells are not able to become established in the host thymus and induce tolerance before host T cells regenerate repeated doses of anti-recipient T cell antibodies may be necessary throughout the non-myeloablative regimen. Continuous depletion of host T cells may be required for several weeks. Alternatively, e.g., if this approach is not successful, and tolerance (as measured by donor skin graft acceptance, specific cellular hyporesponsiveness in vitro, and humoral tolerance) is not induced in these animals, the approach can be modified to include host thymectomy.
  • thymectomized recipients host T cells do not have an opportunity to differentiate in a host thymus, but must differentiate in the donor thymus. Immunocompetence can be measured by the ability to reject a non-donor type allogeneic donor skin graft, and to survive in a pathogen-containing environment.
  • a sublethal dose e.g., a dose about equal to 100, or more than 100 and less than about 400, rads, whole body radiation, plus 700 rads of local thymic radiation, has been found effective for this purpose.
  • Natural antibody elimination Natural antibodies are adsorbed from the recipient's blood by hemoperfusion of a liver of the donor species. Pre-formed natural antibodies-(nAb) are the primary agents of graft rejection. Natural antibodies bind to xenogeneic endothelial cells and are primarily of the IgM class. These antibodies are independent of any known previous exposure to antigens of the xenogeneic donor. B cells that produce these natural antibodies tend to be T cell-independent, and are normally tolerized to self antigen by exposure to these antigens during development. The mechanism by which newly developing B cells are tolerized is unknown. The liver is a more effective adsorber of natural antibodies than the kidney.
  • the next step in the non-myeloablative procedure is to implant donor stromal tissue, preferably obtained from fetal liver, thymus, and/or fetal spleen, into the recipient, preferably in the kidney capsule.
  • donor stromal tissue preferably obtained from fetal liver, thymus, and/or fetal spleen
  • Stem cell engraftment and hematopoiesis across disparate species barriers is enhanced by providing a hematopoietic stromal environment from the donor species.
  • the stromal matrix supplies species-specific factors that are required for interactions between hematopoietic cells and their stromal environment, such as hematopoietic growth factors, adhesion molecules, and their ligands.
  • Each organ includes an organ specific stromal matrix that can support differentiation of the respective undifferentiated stem cells implanted into the host.
  • adult thymus may be used, fetal tissue obtained sufficiently early in gestation is preferred because it is free from mature T lymphocytes which can cause GVHD. Fetal tissues also tend to survive better than adult tissues when transplanted.
  • thymic stromal tissue can be irradiated prior to transplantation, e.g., irradiated at 1000 rads.
  • fetal liver cells can be administered in fluid suspension.
  • bone marrow cells or another source of hematopoietic stem cells, e.g., a fetal liver suspension, or cord blood stem cells, of the donor are injected into the recipient.
  • Donor stem cells home to appropriate sites of the recipient and grow contiguously with remaining host cells and proliferate, forming a chimeric lymphohematopoietic population.
  • BMC bone marrow cells
  • hematopoietic stem cell e.g., BMC
  • engraftment has been achieved.
  • Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • the donor of the implant and the individual that supplies either the tolerance-inducing hematopoietic cells or the liver to be perfused should be the same individual or should be as closely related as possible.
  • zero time is defined as the moment that the arterial and venous cannulas of the recipient are connected to the liver to be perfused.
  • a commercial preparation (Upjohn) of horse anti-human anti-thymocyte globulin (ATG) is injected into the recipient.
  • ATG eliminates mature T cells and natural killer cells that would otherwise cause rejection of the bone marrow cells used to induce tolerance.
  • the recipient is anesthetized, an IV catheter is inserted into the recipient, and 6 ml of heparinized whole blood are removed before injection.
  • the ATG preparation is then injected (50 mg/kg) intravenously. Six ml samples of heparinized whole blood are drawn for testing at time points of 30 min., 24 hrs and 48 hrs.
  • Blood samples are analyzed for the effect of antibody treatment on natural killer cell activity (testing on K562 targets) and by FACS analysis for lymphocyte subpopulations, including CD4, CD8, CD3, CD11b, and CD16.
  • Preliminary data from both assays indicate that both groups of cells are eliminated by the administration of ATG. If mature T cells and NK cells are not eliminated, ATG can be re-administered at later times in the procedure, both before and after organ transplantation.
  • Sublethal irradiation is administered to the recipient between days ⁇ 1 and ⁇ 8. Irradiation is necessary to eliminate enough of the recipient's endogenous BMC to stimulate hematopoiesis of the newly introduced foreign BMC. Sublethal total body irradiation is sufficient to permit engraftment with minimal toxic effects to the recipient.
  • Whole body radiation 150 Rads
  • TRBC bilateral cobalt teletherapy unit at 10 Rads/min.
  • Local irradiation of the thymus 700 Rads was also employed in order to facilitate engraftment.
  • Natural antibodies are a primary cause of organ rejection.
  • an operative adsorption of natural antibodies is performed, using a miniature swine liver, as follows. At ⁇ 90 minutes the swine donor is anesthetized, and the liver prepared for removal by standard operative procedures. At ⁇ 60 minutes the recipient monkey is anesthetized. A peripheral IV catheter is inserted, and a 6 ml sample of whole blood is drawn. Through mid-line incision, the abdominal aorta and the vena cava are isolated. Silastic cannulas containing side ports for blood sampling are inserted into the blood vessels.
  • the liver is perfused in situ until it turns pale, and then removed from the swine donor and placed into cold Ringers Lactate. The liver is kept cold until just prior to reperfusion in the monkey. A liver biopsy is taken. At ⁇ 10 minutes the liver is perfused with warm albumin solution until the liver is warm (37 degrees).
  • liver biopsies are taken at 30 minutes and 60 minutes, respectively.
  • Samples of recipient blood are also drawn for serum at 30 minutes and 60 minutes respectively.
  • the liver is disconnected from the cannulas and the recipient's large blood vessels are repaired.
  • the liver having served its function of adsorbing harmful natural antibodies from the recipient monkey, is discarded.
  • Additional blood samples for serum are drawn from the recipient at 2, 24, and 48 hours. When this procedure was performed on two sequential perfusions of swine livers, the second liver showed no evidence of mild ischemic changes during perfusion.
  • the second liver looked grossly normal and appeared to be functioning, as evidenced by a darkening of the venous outflow blood compared to the arterial inflow blood in the two adjacent cannulas.
  • Tissue sections from the livers were normal, but immunofluorescent stains showed IgM on endothelial cells. Serum samples showed a decrease in natural antibodies.
  • donor bone marrow cells are administered to the recipient to form chimeric bone marrow.
  • the presence of donor antigens in the bone marrow allows newly developing B cells, and newly sensitized T cells, to recognize antigens of the donor as self, and thereby induces tolerance for the implanted organ from the donor.
  • donor stromal tissue in the form of tissue slices of fetal liver, thymus, and/or fetal spleen are transplanted under the kidney capsule of the recipient.
  • Stromal tissue is preferably implanted simultaneously with, or prior to, administration of hematopoietic stem cells, e.g., BMC, or a fetal liver cell suspension.
  • This assay uses monoclonal antibodies to distinguish between donor class I major histocompatibility antigens and leukocyte common antigens versus recipient class I major histocompatibility antigens.
  • BMC can in turn be injected either simultaneously with, or preceding, organ transplant.
  • Bone marrow is harvested and injected intravenously (7.5 ⁇ 10 8 /kg) as previously described (Pennington et al., 1988, Transplantation 45:21-26). Should natural antibodies be found to recur before tolerance is induced, and should these antibodies cause damage to the graft, the protocol can be modified to permit sufficient time following BMT for humoral tolerance to be established prior to organ grafting.
  • the approaches described above are designed to synergistically prevent the problem of transplant rejection.
  • a kidney is implanted into a cynomolgus monkey following liver adsorption of natural antibodies, without use of bone marrow transplantation to induce tolerance, renal functions continued for 1-2 days before rejection of the kidney.
  • four steps of the procedure were performed (adsorption of natural antibodies by liver perfusion, administration of ATG, sublethal irradiation and bone marrow infusion, followed by implant of a porcine kidney into a primate recipient), the kidney survived 7 days before rejection. Despite rejection of the transplanted organ, the recipient remained healthy.
  • implanted grafts may consist of organs such as liver, kidney, heart; body parts such as bone or skeletal matrix; tissue such as skin, intestines, endocrine glands; or progenitor stem cells of various types.
  • the methods of the invention may be employed with other mammalian recipients (e.g., rhesus monkeys, humans) and may use other mammalian donors (e.g., primates, swine, sheep, dogs).
  • mammalian recipients e.g., rhesus monkeys, humans
  • mammalian donors e.g., primates, swine, sheep, dogs
  • the methods of the invention may be employed in combination, as described, or in part.
  • the method of introducing bone marrow cells may be altered, particularly by (1) increasing the time interval between injecting hematopoietic stem cells and implanting the graft; (2) increasing or decreasing the amount of hematopoietic stem cells injected; (3) varying the number of hematopoietic stem cell injections; (4) varying the method of delivery of hematopoietic stem cells; (5) varying the tissue source of hematopoietic stem cells, e.g., a fetal liver cell suspension may be used; or (6) varying the donor source of hematopoietic stem cells.
  • hematopoietic stem cells derived from the graft donor are preferable, hematopoietic stem cells may be obtained from other individuals or species, or from genetically-engineered completely or partially inbred donor strains, or from in vitro cell culture.
  • Methods of preparing the recipient for transplant of hematopoietic stem cells may be varied.
  • the recipient may undergo a splenectomy or a thymectomy.
  • the latter would preferably by administered prior to the non-myeloablative regimen, e.g., at day ⁇ 14.
  • Hemoperfusion of natural antibodies may: (1) make use of other vascular organs, e.g., liver, kidney, intestines; (2) make use of multiple sequential organs; (3) make use of varying the length of time each organ is perfused; (4) make use of varying the donor of the perfused organ.
  • Irradiation of the recipient may make use of: (1) of varying the adsorbed dose of whole body radiation below the sublethal range; (2) of targeting different body parts (e.g., thymus, spleen); (3) varying the rate of irradiation (e.g., 10 rads/min, 15 rads/min); or (4) varying the time interval between irradiation and transplant of hematopoietic stem cells; any time interval between 1 and 14 days can be used, and certain advantages may flow from use of a time interval of 4-7 days.
  • Antibodies introduced prior to hematopoietic cell transplant may be varied by: (1) using monoclonal antibodies to T cell subsets or NK cells (e.g., anti-NKH1 A , as described by U.S. Pat. No. 4,772,552 to Hercend, et al., hereby incorporated by reference); (2) preparing anti-human ATG in other mammalian hosts (e.g., monkey, pig, rabbit, dog); or (3) using anti-monkey ATG prepared in any of the above mentioned hosts.
  • monoclonal antibodies to T cell subsets or NK cells e.g., anti-NKH1 A , as described by U.S. Pat. No. 4,772,552 to Hercend, et al., hereby incorporated by reference
  • preparing anti-human ATG in other mammalian hosts e.g., monkey, pig, rabbit, dog
  • anti-monkey ATG prepared in any of the above mentioned hosts.
  • host antibodies can be depleted by administration of an excess of hematopoietic cells.
  • Stromal tissue introduced prior to hematopoietic cell transplant may be varied by: (1) administering the fetal liver and thymus tissue as a fluid cell suspension; (2) administering fetal liver or thymus stromal tissue but not both; (3) placing a stromal implant into other encapsulated, well-vascularized sites; or (4) using adult thymus or fetal spleen as a source of stromal tissue.
  • the inventor has discovered that it is possible to induce mixed chimerism with less radiation toxicity by fractionating the radiation dose, i.e., by delivering the radiation in two or more exposures or sessions.
  • the radiation can either be delivered in a single exposure, or more preferably, can be fractionated into two or more exposures or sessions.
  • the sum of the fractionated dosages is preferably equal, e.g., in rads or Gy, to the radiation dosage which can result in mixed chimerism when given in a single exposure.
  • the fractions are preferably approximately equal in dosage.
  • a single dose of 700 rads can be replaced with, e.g., two fractions of 350 rads, or seven fractions of 100 rads.
  • Hyperfractionation of the radiation dose can also be used in methods of the invention.
  • the fractions can be delivered on the same day, or can be separated by intervals of one, two, three, four, five, or more days. Whole body irradiation, thymic irradiation, or both, can be fractionated.
  • Methods of the invention can include recipient splenectomy.
  • hemoperfusion e.g., hemoperfusion with a donor organ
  • Other methods for depleting or otherwise inactivating natural antibodies can be used with any of the methods described herein.
  • drugs which deplete or inactivate natural antibodies e.g., deoxyspergualin (DSG) (Bristol), or anti-IgM antibodies
  • DSG deoxyspergualin
  • anti-IgM antibodies can be administered to the recipient of an allograft or a xenograft.
  • DSG or similar drugs
  • anti-IgM antibodies, and hemoperfusion can be used to deplete or otherwise inactivate recipient natural antibodies in methods of the invention.
  • DSG at a concentration of 6 mg/kg/day, i.v. has been found useful in suppressing natural antibody function in pig to cynomolgus kidney transplants.
  • Some of the methods described herein use irradiation to create hematopoietic space, and thereby prepare a recipient for the administration of allogeneic, xenogeneic, syngeneic, or genetically engineered autologous, stem cells.
  • the use of sublethal levels of bone marrow depletion allows the generation of mixed chimerism in the recipient.
  • Mixed chimerism is generally preferable to total or lethal ablation of the recipient bone marrow followed by complete reconstitution of the recipient with administered stem cells.
  • Xenogeneic thymic tissue is easier to obtain and in general is less likely to harbor human pathogens. Thus, xenogeneic thymic tissue is preferred in methods for restoring or inducing immunocompetence. Allogeneic thymic tissue can however be used in these methods.
  • thymic irradiation e.g., to inactivate host thymic-T cells or to otherwise diminish the host's thymic-T cell mediated responses to donor antigens. It has been discovered that the thymic irradiation called for in allogeneic or xenogeneic methods of the invention can be supplemented with, or replaced by, other treatments which diminish (e.g., by depleting thymic-T cells and/or down modulating one or more of the T cell receptor (TCR), CD4 co-receptor, or CD8 co-receptor) the host's thymus function, e.g., the host's thymic-T cell mediated response.
  • TCR T cell receptor
  • CD4 co-receptor CD4 co-receptor
  • CD8 co-receptor CD8 co-receptor
  • thymic irradiation can be supplemented with, or replaced by, anti-T cell antibodies (e.g., anti-CD4 and/or anti-CD8 monoclonal antibodies) administered a sufficient number of times, in sufficient dosage, for a sufficient period of time, to diminish the host's thymic-T cell mediated response.
  • anti-T cell antibodies e.g., anti-CD4 and/or anti-CD8 monoclonal antibodies
  • anti-T cell antibodies should be administered repeatedly.
  • anti-T cell antibodies can be administered one, two, three, or more times prior to donor thymus or bone marrow transplantation.
  • a pre-thymus or bone marrow transplantation dose of antibodies will be given to the patient about 5 days prior to thymus or bone marrow transplantation. Additional, earlier doses 6, 7, or 8 days prior to thymus or bone marrow transplantation can also be given. It may be desirable to administer a first treatment then to repeat pre-thymus or bone marrow administrations every 1-5 days until the patient shows excess antibodies in the serum and about 99% depletion of peripheral T cells and then to perform the bone marrow transplantation.
  • Anti-T cell antibodies can also be administered one, two, three, or more times after thymus or donor bone marrow transplantation.
  • a post-thymus or bone marrow transplant treatment will be given about 2-14 days after bone marrow transplantation.
  • the post thymus or bone marrow administration can be repeated as many times as needed. If more than one administration is given the administrations can be spaced about 1 week apart. Additional doses can be given if the patient appears to undergo early or unwanted T cell recovery.
  • anti-T cell antibodies are administered at least once (and preferably two, three, or more times) prior to donor thymus or bone marrow transplantation and at least once (and preferably two, three, or more times) after donor thymus or bone marrow transplantation.
  • a recipient e.g., an allograft or xenograft recipient
  • a recipient e.g., an allograft or xenograft recipient
  • transplantation of tolerizing stem cells and/or the graft This is particularly useful in the case of humans receiving grafts from cadavers.
  • the treatment(s) can be administered, within a few days, preferably within 72, 48, or 24 hours, of transplantation of the stem cells and/or the graft.
  • primate e.g., human
  • recipients of allografts can be given any or all of treatments to inactivate or deplete host antibodies, treatments to inactivate host T cells or NK cells, or irradiation, within a few days, preferably within 72, 48, or 24 hours, of transplantation of stem cells and/or the graft.
  • treatment to deplete recipient T cells and/or NK cells e.g., administration of ATG
  • WBI thymic irradiation
  • stem cell e.g., bone marrow stem cells, administered on day 0.
  • the graft e.g., a renal allograft, is transplanted on day 0).
  • window there is a permissible time period (“window”) for hematopoietic stem cell engraftment following the creation of space (e.g., by whole body irradiation) for the donor hematopoietic stem cells in a recipient. It has further been discovered that space created for hematopoietic stem cell engraftment can be monitored over time by monitoring peripheral white blood cell levels in a recipient.
  • the myelosuppressive treatment sufficient to create hematopoietic space generally results in a reduction in white blood cell (WBC) levels (as revealed, e.g., by WBC counts) and the WBC reduction serves as a marker for the presence of hematopoietic space.
  • WBC white blood cell
  • the marker is a conservative one since WBC counts may recover at a time when space is still present in an animal.
  • transplantation can be performed during the permissible window for engraftment following creation of space for the hematopoietic stem cells.
  • white blood cell levels can be followed to monitor space for the donor hematopoietic stem cells (i.e., to assess the permissible window for engraftment).
  • Examples of procedures involving hematopoietic stem cell transplantation include: 1) conditioning of a recipient for an allo- or xenograft in which hematopoietic stem cell transplantation is performed in conjunction with transplantation of another allo- or xenograft; 2) treatment of various hematopoietic disorders, including leukemias, lymphomas and other hematopoietic malignancies and genetic hematopoietic disorders (e.g., adenosine deaminase deficiency, bare lymphocyte syndrome and other congenital immunodeficiency diseases) in which hematopoietic stem cell transplantation is performed therapeutically; and 3) transplantation of genetically modified hematopoietic stem cells (e.g., genetically modified autologous hematopoietic stem cells) to deliver a gene product to a recipient (e.g., as gene therapy).
  • genetically modified hematopoietic stem cells e.g., genetically modified
  • methods of the invention include a method of determining if a myelosuppressive or hematopoietic-space inducing treatment is sufficient to create hematopoietic space.
  • the method includes administering a myelosuppressive treatment to a recipient, and determining the level of white blood cells in the recipient, e.g., by determining the WBC count of the recipient, a depression in the level of white blood cells being indicative of the presence or induction of hematopoietic space.
  • the engraftment of exogenously supplied hematopoietic stem cells can be promoted by treating the recipient of the cells so as to induce hematopoietic space in the recipient.
  • Hematopoietic space is commonly induced by radiation, but other procedures can replace or reduce the need for WBI.
  • space can be created by treating the recipient with a monoclonal antibody against MHC class I antigens expressed by the recipient (see e.g., Voralia, M. et al. (1987) Transplantation 44:487) or space can be created by treating the recipient with myelosuppressive drugs (see e.g., Lapidot, T. et al. (1990) Proc. Natl. Acad. Sci. USA 87:4595).
  • embodiments of the invention include methods of promoting the acceptance a graft (e.g., by prolonging the acceptance the graft) by a recipient, by introducing into the recipient, donor antigen.
  • the graft can be an allograft, e.g., a graft from a primate e.g., a human, which is introduced into a primate of the same species.
  • the graft can be a concordant or discordant xenograft.
  • the graft can be a miniature swine graft introduced into a second species, e.g., a primate, e.g., a human.
  • the invention provides several methods of inducing tolerance to foreign antigens, e.g., to antigens on allogeneic or xenogeneic tissue or organ grafts. These methods can be used individually or in combination. For example, it has been discovered that short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA) significantly prolongs graft acceptance. (Preferably the help reduction regimen of the invention substantially eliminates the initial burst of IL-2 which accompanies the first recognition of an antigen but does not eliminate mature T cells. This is distinct from anti-T cell antibody treatments which eliminate mature T cells.)
  • a help reducing agent e.g., a short high dose course of cyclosporine A (CsA) significantly prolongs graft acceptance.
  • CsA cyclosporine A
  • the help reduction regimen of the invention substantially eliminates the initial burst of IL-2 which accompanies the first recognition of an antigen
  • an immunosuppressant e.g., cyclosporine
  • section I Experiments which show the effect of cyclosporine-induced tolerance on renal allografts in primates are described in section I below.
  • the help suppression methods of the invention can be combined with other methods for prolonging graft acceptance.
  • Section II below discusses implantation of retrovirally transformed bone marrow cells to induce tolerance to MHC disparity. This method can be combined with help suppression methods of the invention, e.g., a short course of high dose of cyclosporine to induce tolerance to class I and other minor disparities.
  • Section III discusses implantation of bone marrow cells to induce tolerance to MHC disparity. This method can be combined with a short course of high dose cyclosporine administration to induce tolerance to class I and other minor disparities. A short course of cyclosporine, to eliminate T cells, can also be combined with bone marrow transplant.
  • Renal transplants were performed between cynomolgus monkeys with or without a brief course of T cell-help-eliminating immunosuppression in the form of a short course of high dose cyclosporine. This regimen significantly prolonged acceptance of the grafts. Monkeys which received a post-transplant course of high dose cyclosporine (without Prednisone) were tolerant to kidney grafts from MLC (mixed lymphocyte culture assay) nonreactive class I and minor antigen mismatched donors for over 65 days, see below. Monkeys which did not receive post-transplant cyclosporine rejected grafts of the same disparity in less than 20 days. This work is discussed in more detail below.
  • Cynomolgus monkeys were purchased from Charles River Research Laboratories. The animals were quarantined, tested for a full battery of pathogens, and then housed in environmentally controlled rooms in strict conformance to the N.I.H. Guide for Care and use of Laboratory Animals, in an AAALAC accredited facility.
  • CsA Immunosuppression.
  • An intravenous preparation of CsA (Sandimmune, i.v.) was obtained from Sandoz Pharmaceuticals Corporation, Hanover, N.J. Monkeys received 12 doses of about 10 mg/kg with the first dose given 4 hours prior to graft revascularization.
  • the CsA was diluted in 250 ml of normal saline and infused intravenously over 1 hour.
  • the CsA was administered without other immunosuppressants.
  • the duration of therapy was 12 days.
  • the suitable dosage in pigs is about 15 mg/kg delivered intramuscularly.
  • the dosage in either animal should be such that a blood level of about 500-1,000 ng/ml is maintained.
  • Renal Function was followed by creatinine and BUN levels in serum. Pathology was by biopsy. Biopsies were performed at day 7, then weekly for 2 months, then monthly.
  • mice (no cyclosporine) rejected transplanted kidneys in less than 15 days.
  • the results with 6 cyclosporine treated animals were as follows: animal 1, died on day 65 (i.e., 65 days after transplant), the transplanted was rejected, (it should be noted that the blood cyclosporine level of this animal was below 500 ng/ml during the first 7 days after transplant); animal 2, died on day 65 from bleeding from a biopsy, the transplanted kidney was normal at the time of death; animal 3 died on day 82, some rejection was apparent on day 55; animal 4 was still normal at day 70 (at which time the experiment was still in progress); animal 5 was still normal at day 40 (at which time the experiment was still in progress); and animal 6 was still normal at day 26 (at which time the experiment was still in progress).
  • Retroviral transformation allows the reconstitution of a graft recipient's bone marrow with transgenic bone marrow cells, preferably autologous bone marrow cells, expressing allogeneic or xenogeneic MHC genes. Expression of the transgenic MHC genes confers tolerance to grafts which exhibit the products of these or closely related MHC genes. Thus, these methods provide for the induction of specific transplantation tolerance by somatic transfer of MHC genes. Retroviral introduction of MHC genes can be used alone or combined with the T cell help reducing methods described herein. This approach is discussed in detail below.
  • MHC genes for a variety of species are well studied.
  • HLA genes in man see, e.g., Hansen et al., 1989, The Major Histocompatibility Complex, In Fundamental Immunology 2d ed., W. E. Paul, ed., Raven Press Ltd., New York, hereby incorporated by reference
  • SLA genes in swine see e.g., Sachs et al., 1988, Immunogenetics 28:22-29, hereby incorporated by reference, have been cloned and characterized.
  • a gene encoding a MHC antigen can be used in methods of the invention to confer tolerance to a graft which displays that or a closely related MHC antigen.
  • Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a human.
  • the individual which supplies the MHC genes should be as genetically similar as possible, particularly in terms of the MHC genes, to the individual which supplies the graft.
  • MHC probes e.g., a probe from a third person or a synthetic consensus probe, can be used to isolate DNA encoding the MHC gene or genes of the implant donor individual. This allows the closest match between the genes used to confer tolerance and the genes which express MHC antigens on the graft.
  • the implant donor individual and the individual which supplies the tolerance conferring DNA should be the same individual or should be as closely related as possible.
  • implant tissue from a colony of donors which is highly inbred and, more preferably, which is homozygous for the MHC genes. This allows the single cloned MHC sequence to be used for many graft recipients.
  • MHC genes can be introduced into bone marrow cells by any methods which allows expression of these genes at a level and for a period sufficient to confer tolerance. These methods include e.g., transfection, electroporation, particle gun bombardment, and transduction by viral vectors, e.g., by retroviruses.
  • Retroviruses are a preferred delivery system. They have been developed extensively over the past few years as vehicles for gene transfer, see e.g., Eglitis et al., 1988, Adv. Exp. Med. Biol. 241:19.
  • the most straightforward retroviral vector construct is one in which the structural genes of the virus are replaced by a single gene which is then transcribed under the control of regulatory elements contained in the viral long terminal repeat (LTR).
  • LTR viral long terminal repeat
  • a variety of single-gene-vector backbones have been used, including the Moloney murine leukemia virus (MoMuLV).
  • Retroviral vectors which permit multiple insertions of different genes such as a gene for a selectable marker and a second gene of interest, under the control of an internal promoter can be derived from this type of backbone, see e.g., Gilboa, 1988, Adv. Exp. Med. Biol. 241:29.
  • Murine retroviral vectors have been useful for transferring genes efficiently into murine embryonic, see e.g., Wagner et al., 1985, EMBO J. 4:663; Griedley et al., 1987 Trends Genet. 3:162, and hematopoietic stem cells, see e.g., Lemischka et al., 1986, Cell 45:917-927; Dick et al., 1986, Trends in Genetics 2:165-170.
  • a recent improvement in retroviral technology which permits attainment of much higher viral titers than were previously possible involves amplification by consecutive transfer between ecotropic and amphotropic packaging cell lines, the so-called “ping-pong” method, see e.g., Kozak et al., 1990, J. Virol. 64:3500-3508; Bodine et al., 1989, Prog. Clin. Biol. Res. 319: 589-600.
  • Transduction efficiencies can be enhanced by pre-selection of infected marrow prior to introduction into recipients, enriching for those bone marrow cells expressing high levels of the selectable gene, see e.g., Dick et al., 1985, Cell 42:71-79; Keller et al., 1985, Nature 318:149-154.
  • recent techniques for increasing viral titers permit the use of virus-containing supernatants rather than direct incubation with virus-producing cell lines to attain efficient transduction, see e.g., Bodine et al., 1989, Prog. Clin. Biol. Res. 319:589-600.
  • N2A or other Moloney-based vectors are preferred retroviral vectors for transducing human bone marrow cells.
  • the preparative regimens necessary to permit engraftment of modified autologous hematopoietic stem cells may be much less toxic than those needed for allogeneic bone marrow transplantation—preferably requiring only depletion of mature T cells with monoclonal antibodies, as has been recently demonstrated in a mouse model, see Sharabi et al., 1989, J. Exp. Med. 169:493-502. It is possible that transient expression may be sufficient to induce tolerance, which may then be maintained by the transplant even if expression on hematopoietic cells is lost, as has been observed for heart transplants in a mixed xenogeneic bone marrow transplant model, Ildstad et al., 1985, Transplant. Proc. 17: 535-538.
  • the help reducing methods described above can be administered in conjunction with transplantation of the graft, as is described above.
  • Cocultivation of murine bone marrow with high-titer producer lines leads to the transduction of 40% of granulocyte/macrophage colony-forming units (CFU-GM) as determined by the frequency of colony formation under G418 selection.
  • CFU-GM granulocyte/macrophage colony-forming units
  • virus-exposed marrow still contained G418-resistant CFU-GM at a frequency of 25%.
  • virtually all of the transduced and selected colonies contained DRB-specific transcripts.
  • FIG. 3 Details of retroviral constructs are given in FIG. 3 .
  • the diagram in FIG. 3 depicts the GS4.5 retroviral construct.
  • the arrows in FIG. 3 indicate the directions of transcription.
  • GS4.5 the orientation of DRB cDNA transcription is the same as viral transcription.
  • GS4.4 (not shown), the TK promoter and the DRB cDNA were inserted into the 3′ LTR of N2A in the reverse orientation of transcription with respect to viral transcription and the simian virus 40 3′ RNA processing signal was added.
  • pBSt refers to Bluescript vector sequence (Stratagene).
  • TK thymidine kinase
  • the thymidine kinase (TK) promoter was contained within the 215-base-pair (bp) Pvu II-Pst I fragment from the herpes simplex virus TK gene, McKnight, 1980 Nucleic Acids Res. 8:5949-5964.
  • the simian virus 40 3′ RNA processing signal was contained within the 142-bp Hpa I-Sma I fragment from the pBLCAT3 plasmid, Luckow et al., (1987) Nucleic Acids Res. 15:5490-5497, (see FIG. 3 ). Sequence analysis of the junctions of the promoter, the class II cDNA, and the vector sequences confirmed that the elements of the constructs were properly ligated.
  • retroviral constructs were transfected into the amphotropic packaging cell line PA317, and transfectants were selected in G418-containing medium.
  • 8 and 12 clones were found, respectively, to be positive for DRB, although the DRB signal was consistently weaker for the GS4.4-derived clones.
  • Analysis of genomic and spliced transcripts from GS4.5 cells by dot-blot analysis of PEG-precipitated particles revealed heterogeneity among viral transcripts in various clones transfected by GS4.5.
  • G418-resistance (G418 r ) titer determination of supernatants from DRB-positive clones confirmed that the average titer produced by GS4.5-transfected clones (10 3 -10 4 CFU/ml) was significantly higher than that of the GS4.4-transfected clones (10 2 -10 3 CFU/ml). Further transduction experiments were, therefore, conducted with the best clone, named GS4.5 C4, which produced an initial G418 r titer of 3 ⁇ 10 4 CFU/ml.
  • DMEM Dulbecco's modified Eagle's medium
  • fetal bovine serum CELLect Silver; Flow Laboratories
  • nonessential amino acids Woodtaker Bioproducts
  • antibiotics penicillin 5 units/ml
  • streptomycin 5 ⁇ g/ml
  • Amplification of virus titer was achieved by the ping-pong technique. Since there is evidence that psiCRIP packaging cells are less prone to produce helper virus than PA317 when using certain types of vectors, Miller, 1990, Hum. Gene Therapy 1:5-14, DRB recombinant virions were prepared using the psiCRIP/GP-E-86 producer combination. Titer values>1 ⁇ 10 7 CFU/ml with no detectable amphotropic helper viruses were obtained, confirming that this strategy produced safe viral particles suitable for in vivo experiments.
  • RNA species corresponding to the full-length viral genome, the spliced Neo transcript, and the DRB transcription unit were observed with additional RNA species.
  • High molecular size species observed in these experiments may constitute a read-through transcript starting from the TK promoter and ending in the other long terminal repeat (LTR).
  • LTR long terminal repeat
  • Retroviral titers were determined as follows. Replication-defective retroviral particles were produced from packaging cell lines initially transfected with recombinant construct using the standard calcium phosphate precipitation method, Wigler et al., 1978, Cell 14:725-733. Retrovirus production was estimated by the drug-resistance titer (G418-resistant colony-forming units/ml, CFU/ml) as described, Bodine et al., 1990, Proc. Natl. Acad. Sci. USA 87:3738-3742. Except for the psiCRIP line, G418 (GIBCO) selection was carried out in active component at 500 ⁇ g/ml for 10-12 days.
  • G418 G418
  • Hygromycin B selection was applied to psiCRIP-derived packaging clones in medium containing active drug at 50 ⁇ g/ml for 10 days. Replication-competent helper virus titer was assayed on PG4 feline cells by the S + L ⁇ method, Bassen et al., 1971, Nature 229:564-566.
  • PEG precipitation of viral particles was performed as follows. Virions contained in 1 ml of culture supernatant were precipitated with 0.5 ml of 30% (wt/vol) polyethylene glycol (PEG) for 30 min. at 4° C. After centrifugation, the pellets were treated with a mixture of RNase inhibitors (vanadyl ribonuclease complex, BRL), phenol/chloroform-extracted, and ethanol-precipitated. Pellets were then resuspended in 15.7% (vol/vol) formaldehyde and serial dilutions were dotted onto nitrocellulose membrane.
  • RNase inhibitors vanadyl ribonuclease complex
  • FIG. 4 depicts the structure of the provirus genome and the expected sizes of transcripts initiated from either the viral LTR or the TK promoters.
  • the class II transduction assay was performed as diagrammed in FIG. 6 .
  • NIH 3T3 cells were transfected with the SLA-DRA d cDNA inserted in a plasmid expression vector, Okayama et al., 1982, Mol. Cell. Biol. 2:161-170.
  • Approximately 3 ⁇ 10 4 cells of a stable DRA transfectant (clone 11/12.2F) that expressed a high level of DRA mRNA were then transduced overnight with 1 ml of DRB-containing retroviral supernatant.
  • Cells were subsequently cultivated in fresh DMEM supplemented with 10% fetal bovine serum and antibiotics for 2 additional days and examined for cell surface expression of the DR antigen by FCM analysis.
  • the class II transduction assay described here provides a fast and simple method to test both the expression and functional titer of retroviral constructs.
  • DRA deoxyribonucleic acid
  • the need for lengthy double selection after transduction by two separated vectors Yang et al., 1987, Mol. Cell Biol. 1:3923-3928; Korman et al., 1987, Proc. Natl. Acad. Sci. USA 84:2150-2154, is obviated.
  • Cell-surface expression of DR heterodimers was demonstrated by FCM analysis 3 days after transduction, providing direct evidence that the transferred sequences were sufficient to produce significant level of DR ⁇ chain. More importantly, this test allows determination of “functional” titers based on the expression of the gene of interest rather than on that of the independently regulated drug-resistance marker.
  • the SLA-DRB probe was an EcoRI cDNA fragment containing the complete coding sequence of the DR ⁇ chain, Gustafsson et al., 1990, Proc. Natl. Acad Sci. USA 87:9798-9802.
  • the neomycin phosphotransferase gene (Neo) probe was the Bcl I-Xho I fragment of the N2A retroviral plasmid, Hantzopoulos et al., 1989, Proc. Natl. Acad Sci. USA 86:3519-3523.
  • DRB-specific transcripts were also detected in the bulk population of drug-resistant colonies and in producer cells but were not detected in controls such as a bulk population of untransduced colonies, fibroblasts used to provide carrier RNA, and a bulk population of transduced colonies processed as above but without reverse transcriptase. These latter data demonstrate that the PCR signal was dependent on the synthesis of cDNA, excluding the possibility that provirus, rather than viral message, was responsible for the amplified fragment.
  • Bone marrow cells were obtained from the femora of 6- to 12-week-old female C57BL/10 mice and were prepared as described, Ildstad et al., 1984, Nature 307:168-170. Methylcellulose colony assays for granulocyte/macrophage colony-forming units (CFU-GM), Eaves et al., 1978, Blood 52:1196-1210, were performed as described using 5% (vol/vol) murine interleukin 3 culture supplement (Collaborative Research). Long-term Dexter-type bone marrow cultures were initiated in 60-mm culture dishes with 2 ⁇ 10 7 nucleated cells, Eaves et al., 1987, CRC Crit. Rev. Oncol. Hematol. 7:125-138.
  • CFU-GM granulocyte/macrophage colony-forming units
  • Bone marrow cells were transduced essentially as described, Bodine et al., 1989, Proc. Natl. Acad. Sci. USA 86:8897-8901. Briefly, bone marrow was harvested for 6-12-week-old female C57BL/10 donors that had been treated 2 days with 5-fluorouracil (150 mg/kg). Prestimulation was performed by incubating 1 ⁇ 10 6 cells per ml for 2 days in long-term Dexter-type bone marrow culture medium to which was added 7.5% interleukin 3 culture supplement and recombinant human interleukin 6 (200 units/ml; gift from J. Jule, National Institutes of Health, Bethesda, Md.). Marrow cells were transduced for 48 hr by adding 5 ⁇ 10 6 cells per 10-cm plate containing nearly confluent virus-producers, Polybrene (8 mg/ml), and the cytokines described above.
  • First-strand cDNA was prepared from 20 ⁇ g of total RNA using the Invitrogen Red Module kit.
  • cDNA was then subjected to 50 cycles of PCR amplification in the presence of the SLA DRB-specific oligonucleotides 04 (5′-CCACAGGCCTGATCCCTAATGG) (Seq. I.D. No. 1) and 17 (5′-AGCATAGCAGGAGCCTTCTCATG) (Seq. I.D. No. 2) using the Cetus GeneAmp kit as recommended (Perkin-Elmer/Cetus). Reaction products were visualized after electrophoresis on a 3% NuSieve agarose gel (FMC) by staining with ethidium bromide.
  • SLA DRB-specific oligonucleotides 04 5′-CCACAGGCCTGATCCCTAATGG
  • 17 5′-AGCATAGCAGGAGCCTTCTCATG
  • FCM analysis was performed with a FAC-Scan II fluorescence-activated cell sorter (Becton Dickenson) on cells stained with the anti-DR monoclonal antibody 40D, Pierres et al., 1980, Eur. J. Immunol. 10:950-957, an anti-H-2 d allo antiserum, or the anti-porcine CD8 monoclonal antibody 76-2-11, Pescovitz et al., 1984, J. Exp. Med. 160:1495-1505, followed by fluorescein isothiocyanate-labeled goat anti-mouse antibodies (Boehringer Mannheim).
  • a MHC gene was transferred into clonogenic progenitor cells from swine using a recombinant retroviral vector (GS4.5) and a transduction protocol designed to be applicable in vivo. Both the selectable drug resistance gene and the allogeneic class II cDNA transferred by this vector were expressed in the progeny of these transduced progenitors. Expression of the Neo gene was monitored functionally by colony formation under G418 selection, while the presence of class II transcripts was detected by PCR analysis. With this latter method, the transcriptional expression of both endogenous and virally derived DRB genes in transduced and selected colonies were demonstrated.
  • GS4.5 containing virions could transduce swine myelopoietic progenitor cells a colony assay adapted for swine CFU-GM was used. Transductions were carried out by incubating bone marrow from a donor of the SLAC haplotype in high titer viral supernatant. Comparisons of the number of colonies which formed in the presence and absence of G418 for a total of 5 independent experiments indicated that 5% to 14% of CFU-GM were transduced.
  • Colonies of cells originating from transduced CFU-GM were examined for the presence of DRB-specific transcripts by converting RNA into cDNA, and then performing PCR amplification. Utilizing a polymorphic Sau3AI restriction site absent from the endogenous DRB c gene, the presence of DRB d -specific transcripts was unambiguously demonstrated. Gel electrophoresis of the PCR product demonstrated that a 183/177 bp doublet indicative of the vector-derived DRB d transcript was amplified in samples derived not only from pools of transduced and selected CFU-GM progeny, but also from at least 4 out of 6 individual colonies tested.
  • Controls were as follows: template synthesized either in the presence or absence of reverse transcriptase; template derived from cells producing GS4.5-containing virions, from PBL isolated from SLA c or SLA d donors, and from untransduced producer cells used as carrier RNA.
  • Transduction of bone marrow was performed as follows. Swine bone marrow was harvested as previously described (Pennington et al., 1988, Transplantation 45:21-26) and transductions were carried out by incubating marrow cells in high titer viral supernatants at an m.o.i. of 3-5 in the presence of 8 ⁇ g of polybrene per ml at 37° C. for 5 hr. Myeloid progenitors were assayed by colony formation in methylcellulose cultures using PHA-stimulated swine lymphocyte conditioned medium as a source of growth factors. Selective medium contained 1.2 mg/ml active G418.
  • Transduced bone marrow was administered to a lethally irradiated miniature swine.
  • peripheral blood lymphocytes were analyzed by Southern, northern, and cell-surface FACS analyses. By all of these test there was evidence of presence of the transduced allogeneic class II gene in these cells and for expression of the product of this gene.
  • northern analysis showed bands characteristic of the transcribed cDNA, and FACS analysis with a combination of alloantisera and monoclonal antibodies to DR showed presence of the transduced allele of DR beta on the surface of peripheral lymphocytes.
  • FACS analysis for cell-surface expression was variable, with PBL from most H2 animals showing only a slight shift of the entire peak for staining with anti-K b , as compared to PBL from A2 animals stained with the same antibody, or as compared to PBL from H2 animals stained with the non-reactive HOPC antibody.
  • Allogeneic grafts On day 40 skin from B10.MBR (K b specific) and B10.BR (control, third party class I disparate) donors was grafted onto all animals. Graft survivals were scored daily by a blinded observer (i.e., readings were made without knowledge of which graft was from which donor strain) until rejection was complete. The survival times are shown in FIG. 9 , and indicate marked specific prolongation of survival of the B10.MBR skin grafts on the recipients of K b -transduced BMC ( FIG. 9B ), but not on recipients of control marrow ( FIG. 9A ).
  • FIGS. 10 shows FACS analysis on thymocytes from skin graft rejecter ( FIGS. 10A , B) and skin graft acceptor ( FIGS. 10C , D). Staining with control HOPC1 antibody ( FIGS. 10A , C) and with specific anti-K b antibody ( FIGS. 10B , D). A similar comparison of staining patterns on bone marrow cells showed the presence of low level K b expression on a cell population in the marrow of the tolerant mouse, but not of the mouse which had rejected its skin graft.
  • B10.AKM and B10.MBR congenic mouse strains are identical except in the MHC class I region.
  • a recombinant retrovirus containing the class I gene from the B10.MBR stain (H-2K b ) linked to a B19 parvovirus promoter (B19-H2K b ) and a neomycin resistance (neo r ) gene was introduced into B10.AKM (H-2K k ) marrow cells.
  • neo r neomycin resistance
  • the transduced marrow was injected into lethally irradiated AKM recipients pre-treated with an anti-CD8 monoclonal antibody. Twelve weeks post BMT, quantitative PCR was used to show that the B19-H-2K b proviral sequences were present in 5%-30% of peripheral blood cells in all recipient animals. Reverse transcriptase PCR was used to demonstrate the B19-H-2K b mRNA in RNA isolated from bone marrow and spleen of a subset of recipient animals.
  • the retroviral vectors used the Maloney murine leukemia virus based vector N2, Armentano et al., 1987, J. Virol. 61:1647-1650. The coding regions within this virus were deleted during its construction, and replaced with the selectable marker gene, neomycin phosphotransferase (Neo), which is transcribed from the viral LTR promoter, and provides drug resistance to G418.
  • This conventional N2 virus was then further modified by insertion of a parvovirus-derived promoter, B19, Liu et al., 1991, J. Virol.
  • P PstI
  • X XhoI
  • H HinDIII
  • E EcoRI
  • B BamHI.
  • This latter cDNA was derived by Waneck et al. during the construction of an H-2 b cDNA library for their purposes, Waneck et al., 1987, J. Exp. Med. 165:1358-1370.
  • Viral producer cell lines were developed using the packaging cell lines for amphotropic (psi-Crip), Danos et al., 1988, Proc. Natl. Acad. Sci 85:6460-6464, and ecotropic (psi-2), Sambrook et al., 1989, Molecular cloning: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, viral production. These cell lines have been specially designed to produce structural viral proteins for the recombinant defective virus to be produced. Viral production was achieved by transfecting psi-Crip with N2-B19-H2b. Both amphotropic and ecotropic producer cell lines were then co-cultivated allowing multiple integration events and high expression [i.e.
  • transduced 3T3 cells were stained with a monoclonal antibody specific to this antigen and analyzed by flow microfluorometry. These experiments clearly demonstrated high level expression of virally derived K b .
  • Bone marrow transplantation was performed as follows. Techniques for bone marrow transplantation in mice are known to those skilled in the art, see e.g., Sykes et al., 1988, J. Immunol. 140:2903-2911. Briefly, recipient B10.AKM mice aged 12 to 16 weeks are lethally irradiated (1025R, 137Cs source, 110R/min) and reconstituted within 8 hours with 2.5 ⁇ 10 6 bone marrow cells, obtained from the tibiae and femora of sex-matched donors aged 6-14 weeks. Animals are housed in sterilized microisolator cages, and receive autoclaved food and autoclaved acidified drinking water.
  • Donors will be treated with 5-Fluorouracil, 150 mg/kg i.v. on day ⁇ 7 in order to induce pluripotent stem cells to cycle.
  • Marrow will be harvested from donors on day ⁇ 5, and T cell depleted with mAbs and complement.
  • Marrow will than be cultured for 5 days in supernatant from an ecotropic packaging cell line (B17H2Kb-18) which produces a high titer of non-infectious retroviral particles containing the K b gene (see below).
  • IL-3 and IL-6 will be added to the cultures.
  • recipient B10.AKM mice will be lethally irradiated (10.25 Gy), and will be reconstituted with 2.5 ⁇ 10 6 BMC transduced with the K b gene.
  • Control animals will be similarly treated, except that they will receive marrow exposed to supernatant from a similar ecotropic packaging line not exposed to a K b -containing vector.
  • the recipient may also be pre-treated with anti-CD8 monoclonal antibody.
  • CM Cell-Mediated Lympholysis
  • Wells containing 10 5 (24 wells), 3 ⁇ 10 4 (24 wells), 10 4 (30 wells), 3000 (30 wells), 1000 (30 wells), 300 (30 wells), and 100 (30 wells) responder cells are prepared. Three thousand 51 Cr-labeled con A blasts are added to each well on day 7, and 4 hour 51 Cr release is measured. Wells are considered positive if 51 Cr release is 3 standard deviations greater than the mean 51 Cr release in 24 wells containing stimulator cells only plus similar numbers of target cells. The Poisson distribution is used to determine the frequency of precursor CTL's which recognize each target, and statistical analysis is performed by the Chi square method of Taswell, Taswell, 1981, J. Immunol. 126:1614.
  • Flow microfluorometry One-color and two-color flow cytometry will be performed, and percentages of cells expressing a particular phenotype will be determined from 2-color data, as previously described in detail Sykes, 1990, J. Immunol. 145:3209-3215.
  • the Lysis II software program (Becton Dickinson) will be used for distinguishing granulocytes from lymphocytes by gating on the basis of forward angle and 90° light scatter.
  • Cell sorting will be performed on a Coulter Epics Elite cell sorter.
  • Cell suspensions for flow cytometry PBL, BMC, thymocyte, splenocyte, and lymph node suspensions will be prepared as previously described, Sykes, M. et al., 1988, J.
  • FITC-labeled rat-anti-mouse granulocyte antibody Gr1 is purchased from Zymed; FITC-labeled rat-anti-mouse Thy1.2 mAb will be purchased from Becton-Dickinson; FITC-labeled mouse-anti-human CD3 mAb Leu4 (Becton Dickenson) is used as a directly FITC labeled negative control antibody.
  • Thymic tissue immunofluorescence The tissue is incubated in L15 medium for 24 hours to reduce background staining, and is then cut and embedded in O.C.T. compound for freezing in Isopentane. Frozen sections are prepared (thickness 4 ⁇ m) on a cryostat, dried, fixed in acetone, then washed in PBS. The first antibody incubation (with 28-8-6) is performed in the presence of 2% normal mouse serum, in order to saturate Fc receptors. After 45 minutes, the slides are washed 4 times, and FITC-conjugated secondary reagent (monoclonal rat-anti-mouse IgG2a-FITC, purchased from Pandex) is added. After 45 minutes' incubation with the secondary reagent, four washes are performed and the tissue is mounted. Sections are examined under a fluorescence microscope by an observer who is unaware of the group of animals from which the tissue was obtained.
  • Bone marrow stem cells The methodology used for transduction of bone marrow stem cells has been described previously, Karlsson et al., 1988, Proc. Natl. Acad. Sci. 85:6062-6066. Bone marrow is harvested from 6-12 week old female B10.AKM donors treated 2 days previously with 150 mg/kg 5-FU. Following T cell depletion (see above), the marrow is divided and 10 7 cells per 10 cm plate are cultured for 5 days in the presence of 8 ⁇ g of Polybrene per ml, 10% FCS, 0.6% IL-3-containing supernatant, 0.6% IL-6-containing supernatant, and fresh supernatants from B19H2K b or N2 cells.
  • IL-3- and IL-6-containing supernatant is 48 hour supernatant of COS 7 cells transfected with the murine rIL-3 gene-containing plasmid pCD-IL-3 or with the murine rIL-6 gene-containing plasmid pCD-IL-6, respectively (both plasmids provided by Dr. Frank Lee, DNAX Corp.).
  • IL-3-containing supernatants are tittered by testing proliferation of the IL-3-dependent cell line 32D in the presence of dilutions of these supernatants, and IL-6 is tittered in a similar manner using the IL-6-dependent line T1165 as the indicator cell line.
  • the virus-containing supernatants are refreshed on a daily basis by harvesting the non-adherent layer of each plate, pelleting the cells, and resuspending in freshly harvested filtered virus-containing B19H2K b or N2 supernatant with additives. After 5 days, the non-adherent and adherent BMC are harvested, washed, and resuspended at 2.5 ⁇ 10 6 /ml in Medium 199 with Hepes buffer and Gentamycin plus Heparin 10 U/Ml. One ml. of this suspension is injected i.v. to irradiated mice.
  • Murine CFU-GM assay To test for the bone marrow progenitor cells known as CFU-GM (colony forming unit-granulocyte/macrophage), bone marrow cells are suspended in plating medium consisting of IMDM medium containing 30% defined fetal bovine serum (FBS) (HyClone, Logan, Utah), 10 ⁇ 4 M ⁇ -mercaptoethanol, antibiotics, 5% v/v murine IL-3 culture supplement (Collaborative Research Inc., Bedford, Mass.) and 0.8% methylcellulose (achieved by adding 36% v/v of a commercially prepared solution purchased from the Terry Fox Laboratory, Vancouver).
  • FBS fetal bovine serum
  • N2-B19-H2b vector This vector was constructed staring from the original retroviral vector N2, Eglitis et al., 1985, Science 230:1395-1398, as modified by Shimada to include an additional BamHI site immediately 3′ of the XhoI site. It includes the K b cDNA previously cloned in the vector pBG367, as described by G. Waneck, Waneck et al., 1987, J. Exp. Med. 165:1358-1370. This gene has been placed under control of the B19 promoter, a highly efficient parvo virus derived promoter, Liu et al., 1991, J. Virol. In Press: ] to produce the N2-B19-H2b construct.
  • Southern blot analysis can be performed on DNA extracted from PBL, thymocyte, BMC, splenocyte or lymph node cell suspensions using standard methods, Ausubel et al., 1989, Current protocols in molecular biology. John Wiley & Sons, New York, and probing will be performed with the fragment of K b cDNA released from pBG367 by EcoRI. The genomic DNA will be cut with enzymes capable of distinguishing the transduced K b from other class I genes of the B10.AKM strain.
  • PCR analysis of DNA can be performed using primers previously shown to be effective in our preliminary studies (see FIG. 6 ): (Seq. ID No. 3) 5′ primer: 5′-GGCCCACACTCGCTGAGGTATTTCGTC-3′ (covers 5′ end of ⁇ 1 exon) (Seq. ID No. 4) 3′ primer: 5′-GCCAGAGATCACCTGAATAGTGTGA-3′ (covers 5′ end of ⁇ 2 exon)
  • DNA is subjected to 25 cycles of PCR amplification using these specific oligonucleotides and the Cetus GeneAmp kit (Perkin Elmer Cetus, Norwalk, Conn.) according to the manufacturer's directions.
  • [ 32 ]PdCTP will be included in the PCR reaction in order to visualize products by autoradiography following electrophoresis.
  • RNA can be isolated from 5 ⁇ 10 6 to 5 ⁇ 10 7 cells using the guanidine isothiocyanate and CsCl methods, Chirgwin et al., 1979, Biochem. 18:5294-5308, and will be used for northern analyses, RNase protection analyses, and for PCR analyses of products formed by reverse transcriptase. For situations in which less then 5 ⁇ 10 6 cells are available, for example following tail bleedings of individual mice, we will utilize the QuickPrep mRNA Purification Kit (Amgen) as miniaturized RNA preparation procedure.
  • Amgen QuickPrep mRNA Purification Kit
  • RNAse protection analyses are more sensitive than standard northern blots, yet still quantitative. Procedures based on published methods, Sambrook et al., 1989, Molecular cloning: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, will be used to derive riboprobes. Briefly, the Kb cDNA will be cloned into a plasmid vector containing the T3 and T7 RNA polymerase promoter sequences (bluescript or Bluescribe plasmids from Stratagene). Using appropriate polymerase and 32 P-nucleotides, transcription of the insert will be initiated and the radioactive K b RNA will be purified. This probe will then be incubated with various RNA preparations followed by treatment with ribonuclease. Presence of RNA will be assessed by electrophoresis on a sequencing gel.
  • RNA will be prepared by the GuSCN/CsCl method and first strand cDNA will be prepared from 5 ug of total RNA using the SuperScript preamplification system (BRL/Life Technologies, Inc., Gaithersburg, Md.). PCR amplifications will be conducted for 50 cycles, Hansen et al., J. Immunol.
  • mice After lethal irradiation (10 Gy in two fractions 24 hours apart) in days ⁇ 1 and 0, animals were transplanted with 0.4 to 1.3 ⁇ 10 8 cells/kg on day 0. The effectiveness of the gene transfer was tested using a colony forming unit assay for granulocyte/macrophage progenitors (CFU-GM) in the presence of G418 to select for neomycin resistance.
  • CFU-GM colony forming unit assay for granulocyte/macrophage progenitors
  • the frequency of G418 resistant CFU-GM varied significantly between animals (6.5% to 25.9%) immediately after transduction and dimnished slowly with time. All animals regained their responsiveness to allogeneic stimuli by the third month post-bone marrow transplantation, as tested by MLR.
  • mAbs specific for DQ and DR molecules of the class II MHC were used for “blocking” MLR studies, to separate the effects of recognition of DQ and DR.
  • assays using cells from recipients of bone marrow transplantation transduced with the allogeneic DRB gene the DR portion of the response to the gene-donor type cells was strongly diminished, demonstrating the effectiveness of the transduced gene at inducing DR-specific unresponsiveness. This effect was observed in all experimental animals, although more pronounced in the DRB d to cc combination than in DRB a to gg direction.
  • cytokines Panes 321 (Pixy is a human-GM-CSF/IL3 fusion protein) 100 Units/ml; mouse stem cell growth factor 20 Units/ml; although these cytokines were used, cytokines from the same species as the cell being transformed can also be used) were included in the culture medium during transduction with the allogeneic DRB retroviral expression vector.
  • the cytokines may have led to the transduction of multilineage pluripotent hematopoietic stem cells, including the precursors of dendritic cells which ultimately induced DRB-specific hyporesponsiveness.
  • Xenografts The following procedure was designed to lengthen the time an implanted organ (a xenograft) survives in a xenogeneic host prior to rejection.
  • the organ can be any organ, e.g., a liver, e.g., a kidney, e.g., a heart.
  • the main strategies are elimination of natural antibodies by organ perfusion, transplantation of tolerance-inducing bone marrow, optionally, the implantation of donor stromal tissue, and, optionally, the administration of a short course of a help reducing agent at about the time of introduction of the graft, as described above preparation of the recipient for transplantation includes any or all of these steps. Preferably they are carried out in the following sequence.
  • a preparation of horse anti-human thymocyte globulin is intravenously injected into the recipient.
  • the antibody preparation eliminates mature T cells and natural killer cells. If not eliminated, mature T cells would promote rejection of both the bone marrow transplant and, after sensitization, the xenograft itself. Of equal importance, the ATG preparation also eliminates natural killer (NK) cells. NK cells probably have no effect on the implanted organ, but would act immediately to reject the newly introduced bone marrow.
  • Anti-human ATG obtained from any mammalian host can also be used, e.g., ATG produced in pigs, although thus far preparations of pig ATG have been of lower titer than horse-derived ATG.
  • ATG is superior to anti-NK monoclonal Antibodies, as the latter are generally not lytic to all host NK cells, while the polyclonal mixture in ATG is capable of lysing all host NK cells.
  • Anti-NK monoclonal antibodies can, however, be used.
  • the presence of donor antigen in the host thymus during the time when host T cells are regenerating post-transplant is critical for tolerizing host T cells. If donor hematopoietic stem cells are not able to become established in the host thymus and induce tolerance before host T cells regenerate repeated doses of anti-recipient T cell antibodies may be necessary throughout the non-myeloablative regimen. Continuous depletion of host T cells may be required for several weeks. Alternatively, e.g. if this approach is not successful, and tolerance (as measured by donor skin graft acceptance, specific cellular hyporesponsiveness in vitro, and humoral tolerance) is not induced in these animals, the approach can be modified to include host thymectomy.
  • thymectomized recipients host T cells do not have an opportunity to differentiate in a host thymus, but must differentiate in the donor thymus. If this is not possible, then the animal has to rely on donor T cells developing in the donor thymus for immunocompetence. Immunocompetence can be measured by the ability to reject a non-donor type allogeneic donor skin graft, and to survive in a pathogen-containing environment.
  • the recipient is administered low dose radiation in order to make room for newly injected bone marrow cells.
  • natural antibodies are adsorbed from the recipient's blood by hemoperfusion of a liver of the donor species.
  • Pre-formed natural antibodies are the primary agents of graft rejection.
  • Natural antibodies bind to xenogeneic endothelial cells and are primarily of the IgM class. These antibodies are independent of any known previous exposure to antigens of the xenogeneic donor.
  • B cells that produce these natural antibodies tend to be T cell-independent, and are normally tolerized to self antigen by exposure to these antigens during development. The mechanism by which newly developing B cells are tolerized is unknown.
  • the liver is a more effective adsorber of natural antibodies than the kidney.
  • the fourth step in the non-myeloablative procedure is to implant donor stromal tissue, preferably obtained from fetal liver, thymus, and/or fetal spleen, into the recipient, preferably in the kidney capsule.
  • donor stromal tissue preferably obtained from fetal liver, thymus, and/or fetal spleen
  • Stem cell engraftment and hematopoiesis across disparate species barriers is enhanced by providing a hematopoietic stromal environment from the donor species.
  • the stromal matrix supplies species-specific factors that are required for interactions between hematopoietic cells and their stromal environment, such as hematopoietic growth factors, adhesion molecules, and their ligands.
  • fetal liver can also serve as an alternative to bone marrow as a source of hematopoietic stem cells.
  • the thymus is the major site of T cell maturation.
  • Each organ includes an organ specific stromal matrix that can support differentiation of the respective undifferentiated stem cells implanted into the host.
  • adult thymus may be used, fetal tissue obtained sufficiently early in gestation is preferred because it is free from mature T lymphocytes which can cause GVHD. Fetal tissues also tend to survive better than adult tissues when transplanted.
  • thymic stromal tissue can be irradiated prior to transplantation, e.g., irradiated at 1000 rads.
  • fetal liver cells can be administered in fluid suspension.
  • BMC bone marrow cells
  • another source of hematopoietic stem cells e.g., a fetal liver suspension
  • Donor BMC home to appropriate sites of the recipient and grow contiguously with remaining host cells and proliferate, forming a chimeric lymphohematopoietic population.
  • BMC bone marrow cells
  • newly forming B cells (and the antibodies they produce) are exposed to donor antigens, so that the transplant will be recognized as self.
  • Tolerance to the donor is also observed at the T cell level in animals in which hematopoietic stem cell, e.g., BMC, engraftment has been achieved.
  • a short course of a help reducing agent e.g., a short course of high dose CsA is administered to the recipient.
  • the course is begun at about the time of implantation, or a little before, and is continued for a time about equal to the time it takes for a mature T cell to be stimulated and initiate rejection. While any of these procedures may aid the survival of an implanted organ, best results are achieved when all steps are used in combination.
  • Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • the donor of the implant and the individual that supplies either the tolerance-inducing hematopoietic cells or the liver to be perfused should be the same individual or should be as closely related as possible.
  • zero time is defined as the moment that the arterial and venous cannulas of the recipient are connected to the liver to be perfused.
  • a commercial preparation (Upjohn) of horse anti-human anti-thymocyte globulin (ATG) is injected into the recipient.
  • ATG eliminates mature T cells and natural killer cells that would otherwise cause rejection of the bone marrow cells used to induce tolerance.
  • the recipient is anesthetized, an IV catheter is inserted into the recipient, and 6 ml of heparinized whole blood are removed before infection.
  • the ATG preparation is then injected (50 mg/kg) intravenously. Six ml samples of heparinized whole blood are drawn for testing at time points of 30 min., 24 hours and 48 hours.
  • Blood samples are analyzed for the effect of antibody treatment on natural killer cell activity (testing on K562 targets) and by FACS analysis for lymphocyte subpopulations, including CD4, CD8, CD3, CDllb, and CD16.
  • Preliminary data from both assays indicate that both groups of cells are eliminated by the administration of ATG. If mature T cells and NK cells are not eliminated, ATG can be re-administered at later times in the procedure, both before and after organ transplantation.
  • Sublethal irradiation is administered to the recipient between days ⁇ 1 and ⁇ 8. Irradiation is necessary to eliminate enough of the recipient's endogenous BMC to stimulate hematopoiesis of the newly introduced foreign BMC. Sublethal total body irradiation is sufficient to permit engraftment with minimal toxic effects to the recipient.
  • Whole body radiation 150 Rads
  • TRBC bilateral cobalt teletherapy unit at 10 Rads/min.
  • Local irradiation of the thymus 700 Rads was also employed in order to facilitate engraftment.
  • Natural antibodies are a primary cause of organ rejection.
  • an operative adsorption of natural antibodies is performed, using a miniature swine liver, as follows. At ⁇ 90 minutes the swine donor is anesthetized, And the liver prepared for removal by standard operative procedures. At ⁇ 60 minutes the recipient monkey is anesthetized. A peripheral IV catheter is inserted, and a 6 ml sample of whole blood is drawn. Through mid-line incision, the abdominal aorta and the vena cava are isolated. Silastic cannulas containing side ports for blood sampling are inserted into the blood vessels.
  • the liver is perfused in situ until it turns pale, and then removed from the swine donor and placed into cold Ringers Lactate. The liver is kept cold until just prior to reperfusion in the monkey. A liver biopsy is taken. At ⁇ 10 minutes the liver is perfused with warm albumin solution until the liver is warm (37 degrees).
  • liver biopsies are taken at 30 minutes and 60 minutes, respectively.
  • Samples of recipient blood are also drawn for serum at 30 minutes and 60 minutes respectively.
  • the liver is disconnected from the cannulas and the recipient's large blood vessels are repaired.
  • the liver having served its function of adsorbing harmful natural antibodies from the recipient monkey, is discarded.
  • Additional blood samples for serum are drawn from the recipient at 2, 24, and 48 hours. When this procedure was performed on two sequential perfusions of swine livers, the second liver showed no evidence of mild ischemic changes during perfusion.
  • the second liver looked grossly normal and appeared to be functioning, as evidenced by darkening of the venous outflow blood compared to the arterial inflow blood in the two adjacent cannulas.
  • Tissue sections from the livers were normal, but immunofluorescent stains showed IgM on endothelial cells. Serum samples showed a decrease in natural antibodies.
  • donor bone marrow cells are administered to the recipient to form chimeric bone marrow.
  • the presence of donor antigens in the bone marrow allows newly developing B cells, and newly sensitized T cells, to recognize antigens of the donor as self, and thereby induces tolerance for the implanted organ from the donor.
  • donor stromal tissue in the form of tissue slices of fetal liver, thymus, and/or fetal spleen are transplanted under the kidney capsule of the recipient.
  • Stromal tissue is preferably implanted simultaneously with, or prior to, administration of hematopoietic stem cells, e.g., BMC, or a fetal liver cell suspension.
  • BMC can in turn be injected either simultaneously with, or preceding, organ transplant. Bone marrow is harvested and injected intravenously (7.5 ⁇ 10 8 /kg) as previously described (Pennington et al., 1988, Transplantation 45:21-26). Should natural antibodies be found to recur before tolerance is induced, and should these antibodies cause damage to the graft, the protocol can be modified to permit sufficient time following BMT for humoral tolerance to be established prior to organ grafting.
  • the approaches described above are designed to synergistically prevent the problem of transplant rejection.
  • a kidney is implanted into a cynomolgus monkey following liver adsorption of natural antibodies, without use of bone marrow transplantation to induce tolerance, renal functions continued for 1-2 days before rejection of the kidney.
  • four steps of the procedure were performed (adsorption of natural antibodies by liver perfusion, administration of ATG, sublethal irradiation and bone marrow infusion, followed by implant of a porcine kidney into primate recipient), the kidney survived 7 days before rejection. Despite rejection of the transplanted organ, the recipient remained healthy.
  • the methods of the invention may be employed in combination, as described, or in part.
  • the method of introducing bone marrow cells may be altered, particularly by (1) increasing the time interval between injecting hematopoietic stem cells and implanting the graft; (2) increasing or decreasing the amount of hematopoietic stem cells injected; (3) varying the number of hematopoietic stem cell injections; (4) varying the method of delivery of hematopoietic stem cells; (5) varying the tissue source of hematopoietic stem cells, e.g., a fetal liver cell suspension may be used; or (6) varying the donor source of hematopoietic stem, cells.
  • tissue source of hematopoietic stem cells e.g., a fetal liver cell suspension may be used
  • hematopoietic stem cells may be obtained from other individuals or species, or from genetically-engineered inbred donor strains, or from in vitro cell culture.
  • Methods of preparing the recipient for transplant of hematopoietic stem cells may be varied. For instance, recipient may undergo a splenectomy or a thymectomy. The latter would preferably be administered prior to the non-myeloablative regimen, e.g., at day ⁇ 14.
  • Hemoperfusion of natural antibodies may: (1) make use of other vascular organs, e.g., liver, kidney, intestines; (2) make use of multiple sequential organs; (3) vary the length of time each organ is perfused; (4) vary the donor of the perfused organ.
  • Irradiation of the recipient may make use of: (1) varying the absorbed dose of whole body radiation below the sublethal range; (2) targeting different body parts (e.g., thymus, spleen); (3) varying the rate of irradiation (e.g., 10 rads/min, 15 rads/min); or (4) varying the time interval between irradiation and transplant of hematopoietic stem cells; any time interval between 1 and 14 days can be used, and certain advantages may flow from use of a time interval of 4-7 days.
  • Antibodies introduced prior to hematopoietic cell transplant may be varied by: (1) using monoclonal antibodies to T cell subsets or NK cells (e.g., anti-NKH1 A , as described by U.S. Pat. No. 4,772,552 to Hercend, et al., hereby incorporated by reference); (2) preparing anti-human ATG in other mammalian hosts (e.g., monkey, pig, rabbit, dog); or (3) using anti-monkey ATG prepared in any of the above mentioned hosts.
  • monoclonal antibodies to T cell subsets or NK cells e.g., anti-NKH1 A , as described by U.S. Pat. No. 4,772,552 to Hercend, et al., hereby incorporated by reference
  • preparing anti-human ATG in other mammalian hosts e.g., monkey, pig, rabbit, dog
  • anti-monkey ATG prepared in any of the above mentioned hosts.
  • the methods of the invention may be employed with other mammalian recipients (e.g., rhesus monkeys) and may use other mammalian donors (e.g., primates, sheep, or dogs).
  • host antibodies can be depleted by administration of an excess of hematopoietic cells.
  • Stromal tissue introduced prior to hematopoietic cell transplant may be varied by: (1) administering the fetal liver and thymus tissue as a fluid cell suspension; (2) administering fetal liver or thymus stromal tissue but not both; (3) placing a stromal implant into other encapsulated, well-vascularized sites, or (4) using adult thymus or fetal spleen as a source of stromal tissue.
  • MHC major histocompatibility complex
  • MA minor antigens
  • BMT Bone marrow transplantation
  • kidney transplants that are class II matched to the BMT donor.
  • Long-term unresponsiveness to kidney transplants that are fully mismatched to both the recipient and BMT donor can be achieved in some cases, apparently dependent on the degree of disparity at multiple loci (compare with the difference between inbred and outbred donors).
  • mixed chimerism obtained during a non-myeloablative protocol to achieve engraftment, is capable of producing multilineage lymphohematopoietic chimerism and long-term tolerance to renal allografts between fully MHC mismatched cynomolgus monkeys.
  • Complete ablation of host lymphohematopoietic elements is neither necessary nor desirable when bone marrow transplantation is utilized as a tolerance-inducing regimen. Instead, it is advantageous to achieve a state of mixed chimerism, in which the presence of certain donor-derived elements induce specific tolerance, while host-type antigen presenting cells maintain normal immunocompetence.
  • the basic protocol for nonlethal preparative regimen was as follows: Cynomolgus monkeys weighing 6 to 10 kg. (Charles River Primates, Wilmington, Mass.) were treated with 300 Rads of WBI either as a single dose (#M393) on day ⁇ 6 or as two fractions of 150 Rads each on days ⁇ 6 and ⁇ 5 (#M3093 and #M3293). 700 Rads of thymic irradiation was administered on day ⁇ 1. Horse anti-human thymocyte globulin (ATG) (Upjohn) was administered at 50 mg/kg i.m. on days ⁇ 2, ⁇ 1 and 0.
  • ATG horse anti-human thymocyte globulin
  • Orthotopic kidney transplantation was performed on day 0 through a midline incision using end to side anastamoses of the donor renal artery and renal vein into the recipient aorta and vena cava, respectively, and using a ureteroureteral anastomosis for urinary drainage.
  • Bone marrow was harvested from two donor ribs, prepared as a single cell suspension, and infused i.v. into the recipient at the end of the renal transplant.
  • Treatment with cyclosporine (Sandimmune®, 15 mg/kg/day, suspended in olive oil) i.m. was begun on day 0 and continued for 27 days.
  • Monkey #393 became pancytopenic on day 8, and required three blood transfusions with blood group matched, irradiated whole blood over the next two weeks. However, peripheral blood components recovered gradually thereafter, and were normal by day 30. Renal function has remained normal for over 250 days, and a biopsy on day 215 showed a normal kidney.
  • FCM Sequential flow cytometric analyses were performed on this animal utilizing a monoclonal anti-class I antibody previously determined to distinguish donor from host, and analyzing lymphoid, monocytic and neutrophil populations as determined by scatter profiles. Clear evidence for chimerism in all three subpopulations was detected first on day 10, and persisted at similarly high levels until cyclosporine treatment was discontinued on day 27. Thereafter, the levels of chimerism detected in each subpopulation decreased, but chimerism was still detectable by FCM among lymphocytes (1.5%) and monocytes (29%) as late as day 203, the last day tested. In addition, a bone marrow aspirate on day 203 showed 11.2% donor cells by FCM.
  • the preparative regimen for these xenografts included: 1) conditioning with non-myeloablative whole body irradiation (WBI) and thymic irradiation; 2) removal of preformed mAbs by perfusion of monkey blood through a pig liver; 3) splenectomy; 4) T cell depletion with ATG and/or mAbs; and 5) postoperative immunosuppression with cyclosporine and in some animals anti-IgM mAbs.
  • WBI non-myeloablative whole body irradiation
  • thymic irradiation thymic irradiation
  • the infiltrate contained approximately 20% T cells, as determined by staining with mAbs to CD3, CD4 and CD8.
  • IgM natural antibodies were effectively removed during liver perfusion in this animal, and strikingly, they did not appear in the serum thereafter, IgG levels started to rise on day 7, correlating with the beginning of renal dysfunction.
  • Stromal tissue introduced prior to hematopoietic cell transplant may be varied by: (1) administering the fetal liver and thymus tissue as a fluid cell suspension; (2) administering fetal liver or thymus stromal tissue but not both; (3) placing a stromal implant into other encapsulated, well-vascularized sites, or (4) using adult thymus or fetal spleen as a source of stromal tissue.
  • the methods described herein for inducing tolerance to, or promoting the acceptance of, an allogeneic antigen or allogeneic graft can be used where, as between the donor and recipient, there is any degree of mismatch at MHC loci or other loci which influence graft rejection.
  • class I and class II MHC loci the donor and recipient can be: matched at class I and mismatched at class II; mismatched at class I and matched at class II; mismatched at class I and mismatched at class II; matched at class I, matched at class II.
  • mismatched at MHC class I means mismatched for one or more MHC class I loci, e.g., in the case of humans, mismatched at one or more of HLA-A, HLA-B, or HLA-C, or in the case of swine, mismatch at one or more SLA class I loci, e.g., the swine A or B loci.
  • Mismatched at MHC class II means mismatched at one or more MHC class II loci, e.g., in the case of humans, mismatched at one or more of a DP ⁇ , a DP ⁇ , a DQ ⁇ , a DQ ⁇ , a DR ⁇ , or a DR ⁇ , or in the case of swine, mismatch at one or SLA class II loci, e.g., mismatch at DQ ⁇ or ⁇ , or DR ⁇ or ⁇ .
  • the methods described herein for inducing tolerance to an allogeneic antigen or allogeneic graft can be used where, as between the donor and recipient, there is any degree of reactivity in a mixed lymphocyte assay, e.g., wherein there is no, low, intermediate, or high mixed lymphocyte reactivity between the donor and the recipient.
  • mixed lymphocyte reactivity is used to define mismatch for class II, and the invention includes methods for performing allogeneic grafts between individuals with any degree of mismatch at class II as defined by a mixed lymphocyte assay.
  • Serological tests can be used to determine mismatch at class I or II loci and the invention includes methods for performing allogeneic grafts between individuals with any degree of mismatch at class I and or II as measured with serological methods.
  • the invention features methods for performing allogeneic grafts between individuals which, as determined by serological and or mixed lymphocyte reactivity assay, are mismatched at both class I and class II.
  • the methods of the invention are particularly useful for replacing a tissue or organ afflicted with a neoplastic disorder, particularly a disorder which is resistant to normal modes of therapy, e.g., chemotherapy or radiation therapy.
  • Methods of the invention can be used for inducing tolerance to a graft, e.g., an allograft, e.g., an allograft from a donor which is mismatched at one or more class I loci at one or more class II loci or at one or more loci at each of class I and class II.
  • the graft includes tissue from the digestive tract or gut, e.g., tissue from the stomach, or bowel tissue, e.g., small intestine, large intestine, or colon; the graft replaces a portion of the recipient's digestive system e.g., all or part of any of the digestive tract or gut, e.g., the stomach, bowel, e.g., small intestine, large intestine, or colon.
  • Tolerance refers not only to complete immunologic tolerance to an antigen, but to partial immunologic tolerance, i.e., a degree of tolerance to an antigen which is greater than what would be seen if a method of the invention were not employed.
  • the inventor has discovered that it is possible to induce mixed chimerism with less radiation toxicity by fractionating the radiation dose, i.e., by delivering the radiation in two or more exposures or sessions.
  • the radiation can either be delivered in a single exposure, or more preferably, can be fractionated into two or more exposures or sessions.
  • the sum of the fractionated dosages is preferably equal, e.g., in rads or Gy, to the radiation dosage which can result in mixed chimerism when given in a single exposure.
  • the fractions are preferably approximately equal in dosage.
  • a single dose of 700 rads can be replaced with, e.g., two fractions of 350 rads, or seven fractions of 100 rads.
  • Hyperfractionation of the radiation dose can also be used in methods of the invention.
  • the fractions can be delivered on the same day, or can be separated by intervals of one, two, three, four, five, or more days. Whole body irradiation, thymic irradiation, or both, can be fractionated.
  • the inventor has also discovered that much or all of the preparative regimen can be delivered or administered to a recipient, e.g., an allograft or xenograft recipient, within a few days, preferably within 72, 48, or 24 hours, of transplantation of tolerizing stem cells and/or the graft. This is particularly useful in the case of humans receiving grafts from cadavers.
  • the treatment(s) can be administered, within a few days, preferably within 72, 48, or 24 hours, of transplantation of the stem cells and/or the graft.
  • primate e.g., human
  • recipients of allografts can be given any or all of treatments to inactivate or deplete host antibodies, treatments to inactivate host T cells or NK cells, or irradiation, within a few days, preferably within 72, 48, or 24 hours, of transplantation of stem cells and/or the graft.
  • treatment to deplete recipient T cells and/or NK cells e.g., administration of ATG
  • WBI thymic irradiation
  • stem cell e.g., bone marrow stem cells, administered on day 0.
  • the graft e.g., a renal allograft, is transplanted on day 0).
  • Methods of the invention can include recipient splenectomy.
  • hemoperfusion e.g., hemoperfusion with a donor organ
  • Other methods for depleting or otherwise inactivating natural antibodies can be used with any of the methods described herein.
  • drugs which deplete or inactivate natural antibodies e.g., deoxyspergualin (DSG) (Bristol), or anti-IgM antibodies
  • DSG deoxyspergualin
  • anti-IgM antibodies can be administered to the recipient of an allograft or a xenograft.
  • DSG or similar drugs
  • anti-IgM antibodies, and hemoperfusion can be used to deplete or otherwise inactivate recipient natural antibodies in-methods of the invention.
  • DSG at a concentration of 6 mg/kg/day, i.v. has been found useful in suppressing natural antibody function in pig to cynomolgus kidney transplants.
  • Some of the methods described herein use lethal irradiation to create hematopoietic space, and thereby prepare a recipient for the administration of allogeneic, xenogeneic, syngeneic, or genetically engineered autologous, stem cells.
  • the use of sublethal levels of bone marrow depletion allows the generation of mixed chimerism in the recipient.
  • Mixed chimerism is generally preferable to total or lethal ablation of the recipient bone marrow followed by complete reconstitution of the recipient with administered stem cells.
  • thymic T cells are also included in embodiments of the invention. Some of the methods described herein include the administration of thymic irradiation to inactivate host thymic-T cells or to otherwise diminish the host's thymic-T cell mediated responses to donor antigens.
  • thymic irradiation called for in allogeneic or xenogeneic methods of the invention can be supplemented with, or replaced by, other treatments which diminish (e.g., by depleting thymic-T cells and/or down modulating one or more of the T cell receptor (TCR), CD4 co-receptor, or CD8 co-receptor) the host's thymic-T cell mediated response.
  • TCR T cell receptor
  • CD4 co-receptor CD8 co-receptor
  • thymic irradiation can be supplemented with, or replaced by, anti-T cell antibodies (e.g., anti-CD4 and/or anti-CD8 monoclonal antibodies) administered a sufficient number of times, in sufficient dosage, for a sufficient period of time, to diminish the host's thymic-T cell mediated response.
  • anti-T cell antibodies e.g., anti-CD4 and/or anti-CD8 monoclonal antibodies
  • anti-T cell antibodies should be administered repeatedly.
  • anti-T cell antibodies can be administered one, two, three, or more times prior to donor bone marrow transplantation.
  • a pre-bone marrow transplantation dose of antibodies will be given to the patient about 5 days prior to bone marrow transplantation. Additional, earlier doses 6, 7, or 8 days prior to bone marrow transplantation can also be given. It may be desirable to administer a first treatment then to repeat pre-bone marrow administrations every 1-5 days until the patient shows excess antibodies in the serum and about 99% depletion of peripheral T cells and then to perform the bone marrow transplantation.
  • Anti-T cell antibodies can also be administered one, two, three, or more times after donor bone marrow transplantation.
  • a post-bone marrow transplant treatment will be given about 2-14 days after bone marrow transplantation.
  • the post bone marrow administration can be repeated as many times as needed. If more than one administration is given the administrations can be spaced about 1 week apart. Additional doses can be given if the patient appears to undergo early or unwanted T cell recovery.
  • anti-T cell antibodies are administered at least once (and preferably two, three, or more times) prior to donor bone marrow transplantation and at least once (and preferably two, three, or more times) after donor bone marrow transplantation.
  • T cell-depleting antibodies can replace thymic irradiation in a non-myeloablative conditioning regimen and allow allogeneic bone marrow engraftment and donor-specific tolerance induction.
  • a low toxicity, non-myeloablative conditioning regimen that allows allogeneic bone marrow engraftment and donor-specific tolerance induction in mice has been previously described.
  • a regimen which includes pre-treatment with depleting doses of anti-CD4 and anti-CD8 monoclonal antibodies on day ⁇ 5, administration of 3 Gy whole body irradiation and 7 Gy of thymic irradiation on day 0 followed by administration of fully MHC-mismatched donor bone marrow cells, allows the induction of permanent mixed chimerism and skin graft tolerance.
  • the thymic irradiation step in this protocol was replaced with additional anti-CD4 and anti-CD8 monoclonal antibody treatment.
  • Multilineage chimerism was compared in B10 (H-2 b ) mice receiving allogeneic (B10.A, H-2 a ) bone marrow transplantation on day 0 following 3 Gy whole body irradiation with or without thymic irradiation, and treatment with monoclonal antibodies by a variety of schedules pre and post-bone marrow transplantation. Most (50 of 52) animals that either received thymic irradiation or that received at least two pre-bone marrow transplantation monoclonal antibody treatments demonstrated long-term multilineage peripheral blood mixed allogeneic chimerism (as demonstrated by flow cytometric analysis).
  • the monoclonal antibody treatments were spaced about 5 days apart with the final treatment 1 day prior to bone marrow transplantation.
  • levels of donor T cell reconstitution were highest in animals receiving thymic irradiation or receiving additional anti-T cell monoclonal antibody treatments following bone marrow transplantation.
  • Eleven of 20 mice receiving two pre-bone marrow transplantation monoclonal antibody treatments (the monoclonal antibody treatments were spaced about 5 days apart with the final treatment 1 day prior to bone marrow transplantation) and no thymic irradiation showed relatively low levels of donor T cell reconstitution (less than 20% donor, more than 80% host) at 6 weeks, and 9 of these showed a marked loss of donor cells in all lineages by 20 weeks.
  • mice receiving 1 or 2 additional post-bone marrow transplantation monoclonal antibody treatments showed high levels of donor T cell reconstitution at 6 weeks (mean 86 ⁇ 12% donor), and high levels of donor reconstitution persisted in all lineages at 20 weeks.
  • a second dose of pre-bone marrow transplantation T cell-depleting monoclonal antibodies can replace thymic irradiation and allow tolerance induction in our regimen, but additional monoclonal antibodies administered at one and two weeks post-bone marrow transplantation may increase the ability to reliably induce durable mixed chimerism and tolerance.
  • the capacity of repeated anti-T cell monoclonal antibody treatments to replace thymic irradiation in this regimen most likely reflects their ability to deplete host thymocytes that escape depletion by the initial monoclonal antibody treatment.
  • These monoclonal antibodies deplete most host T cells and induce down-modulation of both TCR and CD4 and CD8 co-receptors on the few remaining cells.
  • early migration of donor bone marrow-derived cells to the host thymus is associated with complete clonal deletion of mature host-type thymocytes with TCR that recognize donor antigens.
  • a small population of host T cells with such TCR persists in the spleens of chimeras, these cells are anergic to stimulation through their TCR.
  • this relatively non-toxic regimen achieves pluripotent hematopoietic stem cell engraftment and specific tolerance by ablating most of the existing T cell repertoire and allowing new T cell development in the presence of intrathymic donor antigen, and by inducing anergy among the few remaining host T cells in the periphery.
  • Some of the methods herein include the administration of hematopoietic stem cells to a recipient.
  • hematopoietic stem cells are administered prior to or at the time of the implantation of a graft (an allograft or a xenograft), the primary purpose of the administration of hematopoietic stem cells being the induction of tolerance to the graft.
  • a graft an allograft or a xenograft
  • the inventors have found that one or more subsequent administrations (e.g., a second, third, fourth, fifth, or further subsequent administration) of hematopoietic stem cells can be desirable in the creation and/or maintenance of tolerance.
  • the invention also includes methods in which hematopoietic stem cells are administered to a recipient, e.g., a primate, e.g., a human, which has previously been administered hematopoietic stem cells as part of any of the methods referred to herein.
  • a recipient e.g., a primate, e.g., a human
  • any method referred to herein which includes the administration of hematopoietic stem cells can further include multiple administrations of stem cells.
  • a first and a second administration of stem cells are provided prior to the implantation of a graft; a first administration of stem cells is provided prior to the implantation of a graft and a second administration of stem cells is provided at the time of implantation of the graft.
  • a first administration of stem cells is provided prior to or at the time of implantation of a graft and a second administration of stem cells is provided subsequent to the implantation of a graft.
  • the period between administrations of hematopoietic stem cells can be varied.
  • a subsequent administration of hematopoietic stem cell is provided: at least two days, one week, one month, or six months after the previous administration of stem cells; at least two days, one week, one month, or six months after the implantation of the graft.
  • the method can further include the step of administering a second or subsequent dose of hematopoietic stem cells: when the recipient begins to show signs of rejection, e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte response to donor antigen; when the level of chimerism decreases; when the level of chimerism falls below a predetermined value; when the level of chimerism reaches or falls below a level where staining with a monoclonal antibody specific for a donor PBMC antigen is equal to or falls below staining with an isotype control which does not bind to PBMC's, e.g.
  • method of the invention can be modified to include a further step of determining if a subject which has received a one or more administrations of hematopoietic stem cells is in need of a subsequent administration of hematopoietic stem cells, and if so, administering a subsequent dose of hematopoietic stem cells to the recipient.
  • any of the methods referred to herein can include the administration of agents, e.g., 15-deoxyspergualin, mycophenolate mofetil, brequinar sodium, or similar agents, which inhibit the production, levels, or activity of antibodies in the recipient.
  • agents e.g., 15-deoxyspergualin, mycophenolate mofetil, brequinar sodium, or similar agents, which inhibit the production, levels, or activity of antibodies in the recipient.
  • agents e.g., 15-deoxyspergualin, mycophenolate mofetil, brequinar sodium, or similar agents, which inhibit the production, levels, or activity of antibodies in the recipient.
  • agents e.g., 15-deoxyspergualin, mycophenolate mofetil, brequinar sodium, or similar agents, which inhibit the production, levels, or activity of antibodies in the recipient.
  • One or more of these agents can be administered: prior to the implantation of donor tissue, e.g., one, two, or three
  • the administration of the agent can be initiated: when the recipient begins to show signs of rejection, e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte response to donor antigen; when the level of chimerism decreases; when the level of chimerism falls below a predetermined value; when the level of chimerism reaches or falls below a level where staining with a monoclonal antibody specific for a donor PBMC antigen is equal to or falls below staining with an isotype control which does not bind to PBMC's, e.g. when the donor specific monoclonal stains less than 1-2% of the cells; or generally, as is needed to maintain tolerance or otherwise prolong the acceptance of a graft.
  • signs of rejection e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte
  • the period over which the agent is administered can be long term, e.g., for six months or more or a year or more, or short term, e.g., for less than a year, more preferably six months or less, more preferably one month or less, and more preferably two weeks or less.
  • the period will generally be at least about one week and preferably at least about two weeks in duration. In preferred embodiments the period is two or three weeks long.
  • Preferred embodiments include administration of 15-deoxyspergualin (6 mg/kg/day) for about two weeks beginning on the day of graft implantation.
  • Some of the methods referred to herein include steps in which antibodies, e.g., preformed natural antibodies, are removed from the blood of a recipient. For example, in some methods antibodies are removed by hemoperfusion of an organ from the donor species.
  • an ⁇ 1-3 galactose linkage epitope-affinity matrix e.g., in the form of an affinity column, is useful for removing antibodies from the recipient's blood.
  • an ⁇ 1-3 galactose linkage epitope-affinity matrix e.g., matrix bound linear B type VI carbohydrate
  • an antibody perfusion or removal technique e.g., organ perfusion
  • Some of the methods referred to herein include the administration of hematopoietic stem cells to a recipient.
  • hematopoietic stem cells are administered prior to or at the time of the administration of a graft (an allograft or a xenograft), the primary purpose of the administration of hematopoietic stem cells being the induction of tolerance to the graft.
  • the inventors have found that administration of one or more cytokines, preferably a cytokine from the species from which the stem cells are derived, can promote tolerance or otherwise prolong acceptance of a graft.
  • the invention also includes methods in a subject which has previously been administered donor hematopoietic stem cells, is administered one or more cytokine, e.g., a donor-species cytokine.
  • any method referred to herein which includes the administration of hematopoietic stem cells can further include the-administration of a cytokine, e.g., SCF, IL-3, or GM-CSF.
  • a cytokine e.g., SCF, IL-3, or GM-CSF.
  • the cytokine one which is species specific in its interaction with target cells.
  • Administration of a cytokine can begin prior to, at, or after the implantation of a graft or the implantation of stem cells.
  • the method can further include the step of administering a first or subsequent dose of a cytokine to the recipient: when the recipient begins to show signs of rejection, e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte response to donor antigen; when the level of chimerism decreases; when the level of chimerism falls below a predetermined value; when the level of chimerism reaches or falls below a level where staining with a monoclonal antibody specific for a donor PBMC antigen is equal to or falls below staining with an isotype control which does not bind to PBMC's, e.g.
  • method of the invention can be modified to include a further step of determining if a subject is in need of cytokine therapy and if so, administering a cytokine.
  • the period over which the cytokine(s) is administered can be long term, e.g., for six months of more or a year or more, or short term, e.g., for a year or less, more preferably six months or less, more preferably one month or less, and more preferably two weeks or less.
  • the period will generally be at least about one week and preferably at least about two weeks in duration.
  • the recipient is a primate, e.g., a human
  • the donor is from a different species
  • the donor is a pig and: pig SCF is administered; pig IL-3 is administered; a combination of pig SCF and pig IL-3 is administered; a pig specific hematopoiesis enhancing factor, e.g., pig GM-SCF, is administered, e.g., after the implantation of stem cells, e.g., about a month after the implantation of stem cells.
  • a particularly preferred embodiment combines a short course, e.g., about a month, of cyclosporine or a similar agent, a short course, e.g., about two weeks, of 15-deoxyspergualin or a similar agent, and a short course, e.g., about two weeks, of donor specific cytokines, e.g., SCF and IL-3.
  • Cynomolgus monkeys receiving pig grafts and pig stem cells treatment which included the combination of cyclosporine (15 mg/kg/day for 28 days), 15-deoxyspergualin (6 mg/kg/day for two weeks), and recombinant pig cytokines (SCF and IL-3, each at 10 ⁇ g/kg/day, i.v., for two weeks) was found to be useful.
  • Administration began at the time of graft implant. (The monkeys were also given a preparative regimen consisting of 3 ⁇ 100 cGy total body irradiation on day ⁇ 6, and ⁇ 5 and hemoperfusion with a pig liver just prior to stem cell administration.)
  • An anti-CD2 antibody preferably a monoclonal, e.g., BTI-322, or a monoclonal directed at a similar or overlapping epitope, can be used in addition to or in place of any anti-T cell antibodies (e.g., ATG) in any method referred to herein.
  • a monoclonal e.g., BTI-322

Abstract

The invention provides methods for restoring or inducing imnmunocompetence, the methods including the step of introducing donor thymic tissue into the recipient. The invention also provides methods for inducing tolerance in a recipient including introducing donor thymic tissue into the recipient. The invention further provides methods of inducing tolerance including administering to the recipient a short course of help reducing treatment or administering a short course and methods of prolonging the acceptance of a graft by administering a short course of an immunosuppressant.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. Ser. No. 09/126,704, filed on Jul. 30, 1998, which is a continuation of U.S. Ser. No. 08/458,720, filed on Jun. 1, 1995, now U.S. Pat. No. 5,876,708, which is a continuation-in-part of: U.S. Ser. No. 08/266,427, filed on Jun. 27, 1994 now U.S. Pat. No. 5,614,187; U.S. Ser. No. 08/451,210, filed on May 26, 1995, which is a file wrapper continuation of U.S. Ser. No. 07/838,595, filed on Feb. 19, 1992, now abandoned; U.S. Ser. No. 08/220,371, filed on Mar. 29, 1994, now abandoned; PCT/US94/05527 filed on May 16, 1994; U.S. Ser. No. 08/08/243,653, filed on May 16, 1994, now U.S. Pat. No. 5,658,564; U.S. Ser. No. 08/114,072, filed on Aug. 30, 1993, now U.S. Pat. No. 5,624,823; U.S. Ser. No. 08/150,739, filed on Nov. 10, 1993, now abandoned; U.S. Ser. No. 08/212,228, filed on Mar. 14, 1994, now abandoned; and PCT/US94/01616 filed on Feb. 14, 1994. All of the above-referenced applications are hereby incorporated by reference.
  • BACKGROUND OF THE INVENTION
  • This invention relates to the replacement of thymus function and to the induction or restoration of immunological tolerance. The invention further relates to tissue and organ transplantation.
  • The thymus is the central organ for the development of mature, self-tolerant T cells that recognize peptide antigens in the context of self major histocompatibility (MHC) antigens. The requirement for self MHC molecules to present antigen is termed MHC restriction. Athymic individuals do not have an organ in which to generate normal numbers of MHC restricted T cells and are therefore immunoincompetent.
  • SUMMARY OF THE INVENTION
  • It has been discovered that host T cells of an athymic T cell depleted host which has received a thymic graft, e.g., a xenogeneic thymic graft, can mature in the donor thymic tissue, e.g., in xenogeneic thymic tissue. Host T cells which mature in the implanted xenogeneic thymic tissue are immunocompetent.
  • Accordingly, the invention features, in one aspect, a method of restoring or inducing immunocompetence (or restoring or promoting the thymus-dependent ability for T cell progenitors to mature or develop into functional mature T cells) in a host or recipient, e.g., a primate host or recipient, e.g., a human, which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells for a normal immune response. The invention includes the step of introducing into the primate host, donor thymic tissue, e.g., xenogeneic thymic tissue, preferably fetal or neonatal thymic tissue, so that host T cells can mature in the implanted thymic tissue.
  • In preferred embodiments the donor of the thymic tissue is a xenogeneic species and: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the host is a primate, e.g., a human, and the thymic tissue is swine, e.g., miniature swine, thymic tissue, or primate thymic tissue.
  • The method can include other steps which facilitate acceptance of the donor tissue, or otherwise optimize the method. In preferred embodiments the thymic tissue is xenogeneic and: liver or spleen tissue, preferably fetal or neonatal liver or spleen tissue, is implanted with the thymic tissue; donor hematopoietic cells, e.g., cord blood stem cells or fetal or neonatal liver or spleen cells, are administered to the recipient, e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously; the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • In other preferred embodiments the method includes: (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient natural killer (NK) cells, e.g., by introducing into the recipient an antibody capable of binding to NK cells of the recipient, to prevent NK mediated rejection of the thymic tissue; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host T cell function, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host CD4+ cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4+ cells of the recipient.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, or the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient (preferably prior to thymic tissue transplantation).
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the host with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the host's blood, e.g., by contacting the host's blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • In preferred embodiments the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • In preferred embodiments the method further includes the step of identifying a host or recipient which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells for a normal immune response.
  • In other preferred embodiments, a graft which is obtained from a different organ than is the thymic tissue is implanted in the recipient; and the recipient does not receive hematopoietic stem cells, e.g., bone marrow cells, from the donor or the donor species.
  • Other methods can be combined with the methods disclosed herein to promote the acceptance of the graft by the recipient. For example, tolerance to the donor tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient. For example, human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and a class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue. When inserted into a recipient primate, e.g., a human, expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor. These methods, and other methods which can be combined with the methods disclosed herein, are discussed in Sachs, U.S. Ser. No. 08/126, 122, filed Sep. 23, 1993, hereby incorporated by reference and in Sachs, U.S. Ser. No. 08/129,608, filed Sep. 29, 1993, hereby incorporated by reference.
  • Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi, and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, hereby incorporated by reference, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance may also be used to promote acceptance of the donor tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994, hereby incorporated by reference.
  • Other methods of promoting tolerance or promoting the acceptance of grafts, e.g., by altering levels of cytokine activity, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072, filed Aug. 30, 1993, hereby incorporated by reference.
  • It has also been discovered that xenogeneic thymic tissue can be used to induce tolerance to a xenogeneic graft in a recipient.
  • Accordingly, in another aspect, the invention features, a method of inducing tolerance in a recipient mammal, e.g., a primate, e.g., a human, of a first species to a graft obtained from a mammal of a second species, e.g., a discordant species. The method includes: prior to or simultaneous with transplantation of the graft, introducing into the recipient mammal thymic tissue, e.g., thymic epithelium, preferably fetal or neonatal thymic tissue, of the second species; and (optionally) implanting the graft in the recipient. The thymic tissue prepares the recipient for the graft that follows, by inducing immunological tolerance at the T-cell level.
  • In preferred embodiments: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the recipient is a human and the thymic tissue is swine, e.g., miniature swine, thymic tissue, or primate thymic tissue.
  • Preferred embodiments include other steps to promote acceptance of the graft thymus and the induction of immunological tolerance or to otherwise optimize the procedure. In preferred embodiments: liver or spleen tissue, preferably fetal or neonatal liver or spleen tissue, is implanted with the thymic tissue; donor hematopoietic cells, e.g., cord blood stem cells or fetal or neonatal liver or spleen cells, are administered to the recipient, e.g., a suspension of fetal liver cells administered intraperitoneally or intravenously; the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • In other preferred embodiments the method includes (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting host CD4+ cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4+ cells of the recipient. An anti-mature T cell antibody which lyses T cells as well as NK cells can be administered. Lysing T cells is advantageous for both thymic tissue and xenograft survival. Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable. Monoclonal preparations can be used in the methods of the invention.
  • Other preferred embodiments include those in which: the recipient does not receive hematopoietic cells from the donor or the donor species: the same mammal of the second species is the donor of both the graft and the thymic tissue; the donor mammal is a swine, e.g., a miniature swine; an anti-human thymocyte polyclonal anti-serum, obtained, e.g., from a horse or pig is administered to the recipient.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • In preferred embodiments the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • In preferred embodiments the method further includes the step of identifying a host or recipient which is in need of a graft.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • Other methods can be combined with the methods disclosed herein to promote the acceptance of the graft by the recipient. For example, tolerance to the xenogeneic thymic tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient. For example, human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue. When inserted into a recipient primate, e.g., a human, expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor. These methods, and other methods which can be combined with the methods disclosed herein, are discussed in Sachs, U.S. Ser. No. 08/126,122, filed Sep. 23, 1993, and in Sachs, U.S. Ser. No. 08/129,608, filed Sep. 29, 1993.
  • Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi, and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance may also be used to promote acceptance of the xenogeneic thymic tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Other methods of promoting tolerance or promoting the acceptance of grafts, e.g., by altering levels of cytokine activity, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072 filed Aug. 30, 1993.
  • In another aspect, the invention features, a method of restoring or inducing immunocompetence in a recipient, e.g., a primate recipient, e.g., a human, at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells to provide a normal immune response. The invention includes the steps of introducing into the primate recipient, donor thymic tissue, e.g., xenogeneic thymic tissue, so that recipient T cells can mature in the implanted donor thymic tissue. The thymic tissue is preferably fetal or neonatal thymic tissue.
  • In preferred embodiments the thymic tissue is xenogeneic and: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the recipient is a human and the thymic tissue is vertebrate, e.g., swine, e.g., miniature swine, thymic tissue, or primate thymic tissue.
  • Acceptance of a graft, especially a xenogeneic graft, will depend on the stage of the immune disorder. Generally, the more advanced the disorder the more compromised the recipient immune system and the easier it is to induce acceptance of donor thymic tissue. In some cases, the tolerizing effect of the graft itself will be sufficient to provide for acceptance of xenogeneic thymus. In other cases, additional measures will be needed. Thus, the method can include other steps which facilitate acceptance of the donor tissue or otherwise optimize the method. In preferred embodiments: liver or spleen tissue, preferably fetal or neonatal liver or spleen tissue, is implanted with the thymic tissue; donor hematopoietic cells, e.g., cord blood stem cells or fetal or neonatal liver or spleen cells, are administered to the recipient, e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously; the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • In preferred embodiments: the method includes (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; the method includes (preferably prior to or at the time of introducing the thymic tissue into the recipient), depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host CD4+ cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4+ cells of the recipient.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • In preferred embodiments the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • In preferred embodiments the method further includes the step of identifying a host or recipient which is at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), which is capable of producing T cell progenitors but which is thymus-function deficient and thus unable to produce a sufficient number of mature functional T cells to provide a normal immune response.
  • Other methods can be combined with the methods disclosed herein to promote the acceptance of the thymic graft by the recipient. For example, tolerance to donor tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient. For example, human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and a class II MHC gene, and the cells implanted in a human recipient who will receive swine thymic tissue. When inserted into a recipient primate, e.g., a human, expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor. These methods, and other methods which can be combined with the methods disclosed herein, are discussed in Sachs, U.S. Ser. No. 08/126, 122, filed Sep. 23, 1993, hereby incorporated by reference and in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi, and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance may also be used to promote acceptance of the donor thymic tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Other methods of promoting tolerance or promoting the acceptance of grafts, e.g., by altering levels of cytokine activity, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072, filed Aug. 30, 1993.
  • In another aspect, the invention features, a method of restoring or inducing immunocompetence in a recipient, e.g., a primate recipient, e.g., a human, at risk for an acquired immune disorder, (e.g., a human at risk for AIDS) which is unable to produce a normal or sufficient number of mature functional T cells to provide normal immune function. The invention includes the steps of introducing into the primate recipient, donor hematopoietic stem cells, so that donor T cells can mature in the recipient thymus.
  • In preferred embodiments the donor stem cells are from a xenogeneic donor and: the xenograft hematopoietic stem cells are from a discordant species; the hematopoietic stem cells are from a concordant species; the recipient is a human and the hematopoietic stem cells are vertebrate, e.g., swine, e.g., miniature swine, hematopoietic stem cells, or primate hematopoietic stem cells.
  • Acceptance of the donor cells will depend on the stage of the immune disorder. Generally, the more advanced the disorder the more compromised the recipient immune system and the easier it is to induce acceptance of donor, especially xenogeneic donor, tissue. In some cases, the tolerizing effect of the stem cells themselves will be sufficient to provide for acceptance. In other cases, additional measures will be needed. Thus, the method can include other steps which facilitate acceptance of the donor cells or otherwise optimize the method. In preferred embodiments: liver or spleen tissue, preferably fetal or neonatal liver or spleen tissue, is implanted with the donor hematopoietic cells, e.g., cord blood stem cells or fetal or neonatal liver or spleen cells, are administered to the recipient, e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously.
  • In preferred embodiments: the method includes, (preferably prior to or at the time of introducing the donor cells into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; the method includes (preferably prior to or at the time of introducing the thymic tissue into the recipient), depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host CD4+ cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4+ cells of the recipient.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient mamma i with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • In preferred embodiments the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • In preferred embodiments the method further includes the step of identifying a host or recipient which is at risk for an acquired immune disorder, (e.g., a human at risk for AIDS) and which is unable to produce a normal or sufficient number of mature functional T cells to provide normal immune function.
  • Other methods can be combined with the methods disclosed herein to promote the acceptance of the transplanted stem cells by the recipient. For example, tolerance to donor tissue can be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient. For example, human recipient stem cells can be engineered to express a swine MHC gene, e.g., a swine class I or class II MHC gene, or both a class I and a class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue. When inserted into a recipient primate, e.g., a human, expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to tissue from the donor. These methods, and other methods which can be combined with the methods disclosed herein, are discussed in Sachs, U.S. Ser. No. 08/126, 122, filed Sep. 23, 1993, and in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance can be combined with the methods disclosed herein to promote acceptance of donor tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Other methods of promoting tolerance or promoting the acceptance of grafts, e.g., by altering levels of cytokine activity, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072, filed Aug. 30, 1993.
  • In another aspect, the invention features, a method of restoring or inducing immunocompetence in a recipient, e.g., a primate recipient, e.g., a human, at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), which is thymus-function deficient and thus unable to produce a normal number of mature functional T cells or a sufficient number of mature functional T cells for a normal immune response. The invention includes the steps of introducing into the primate recipient, donor thymic tissue, preferably, xenogeneic thymic tissue, and donor hematopoietic stem cells, preferably xenogeneic hematopoietic stem cells, so that donor T cells can mature in the implanted donor thymic tissue. The thymic tissue is preferably fetal or neonatal thymic tissue.
  • In preferred embodiments the thymic graft is a xenograft and: the thymic xenograft is a discordant xenograft; the thymic xenograft is a concordant xenograft; the recipient is a human and the thymic tissue is vertebrate, e.g., swine, e.g., immature swine, thymic tissue, or primate thymic tissue.
  • In preferred embodiments: the xenograft hematopoietic stem cells are from a discordant species; the hematopoietic stem cells are a concordant species; the recipient is a human and the hematopoietic stem cells are vertebrate, e.g., swine, e.g., miniature swine, hematopoietic stem cells, or primate hematopoietic stem cells.
  • In preferred embodiments the donor of the thymic graft and the donor of the stem cells are: the same organism; from the same species; syngeneic; matched at least one MHC locus; matched at least one class I MHC locus; matched at least one class II MHC locus; sufficiently MHC matched that one will not reject a graft from the other; miniature swine from a herd which is completely or partially inbred.
  • Acceptance of donor tissue, especially xenogeneic tissue, will depend on the stage of the immune disorder. Generally, the more advanced the disorder the more compromised the recipient immune system and the easier it is to induce acceptance of donor tissue. In some cases, the tolerizing effect of the donor tissue itself will be sufficient to provide for acceptance. In other cases, additional measures will be needed. Thus, the method can include other steps which facilitate acceptance of donor tissue or otherwise optimize the method. In preferred embodiments: liver or spleen tissue, preferably fetal or neonatal liver or spleen tissue, is implanted with the thymic tissue; donor hematopoietic cells, e.g., cord blood cells or fetal or neonatal liver or spleen cells, are administered to the recipient, e.g., a suspension of fetal liver cells is administered intraperitoneally or intravenously; the recipient is thymectomized, preferably before or at the time the xenograft thymic tissue is introduced.
  • In preferred embodiments: the method includes, (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient an antibody capable of binding to natural killer (NK) cells of the recipient, to prevent NK mediated rejection of the thymic tissue; the method includes, (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient mammal; (preferably prior to or at the time of introducing the thymic tissue or stem cells into the recipient) depleting, inactivating or inhibiting host CD4+ cell function, e.g., by introducing into the recipient an antibody capable of binding to CD4, or CD4+ cells of the recipient.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • In preferred embodiments the host or recipient is a post-natal individual, e.g., an adult, or a child.
  • In preferred embodiments the method further includes the step of identifying a host or recipient which is at risk for an acquired immune disorder, (e.g., a human at risk for AIDS), and which is thymus-function deficient and thus unable to produce a normal number of mature functional T cells or a sufficient number of mature functional T cells for a normal immune response.
  • Other methods can be combined with the methods disclosed herein to promote the acceptance of donor tissue by the recipient. For example, tolerance to donor tissue can be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient. For example, human recipient stem cells can be engineered to express a swine MHC gene, e.g. a swine class I or class II MHC gene, or both a class I and a class II gene, and the cells implanted in a human recipient who will receive swine thymic tissue. When inserted into a recipient primate, e.g., a human, expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to tissue from the donor. These methods and other methods which can be combined with the methods disclosed herein are discussed in Sachs, U.S. Ser. No. 08/126, 122, filed Sep. 23, 1993, and in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi, and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance can be combined with the methods disclosed herein to promote acceptance of donor tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Other methods of promoting tolerance or promoting the acceptance of donor tissue, e.g., by altering levels of cytokine activity, or inhibiting Graft-versus-recipient-disease, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072, filed Aug. 30, 1993. It has also been discovered that hematopoietic cells can be use to induce tolerance to a graft.
  • Accordingly, in another aspect, the invention features, a method of inducing immunological tolerance in a recipient mammal, e.g., a primate, e.g., a human, of a first species to a graft obtained from a donor mammal of a second species, e.g., a discordant species e.g., a discordant primate species. The method includes: prior to or simultaneous with transplantation of the graft, introducing into the recipient mammal hematopoietic stem cells, e.g., bone marrow cells, or fetal liver or spleen cells, of the second species; (preferably, the hematopoietic stem cells home to a site in the recipient mammal); optionally, (preferably prior to introducing the hematopoietic stem cells into the recipient mammal), depleting, inactivating or inhibiting recipient NK cells, e.g., by introducing into the recipient mammal an antibody capable of binding to natural killer (NK) cells of the recipient mammal, to prevent NK mediated rejection of the hematopoietic cells; and (optionally) implanting the graft in the recipient. As will be explained in more detail below, the hematopoietic cells prepare the recipient for the graft that follows, by inducing tolerance at both the B-cell and T-cell levels. Preferably, hematopoietic cells are fetal liver or spleen, or bone marrow cells, including immature cells (i.e., undifferentiated hematopoietic stem cells; these desired cells can be separated out of the bone marrow prior to administration), or a complex bone marrow sample including such cells can be used.
  • One source of anti-NK antibody is anti-human thymocyte polyclonal anti-serum. A second, anti-mature T cell antibody can be administered as well, which lyses T cells as well as NK cells. Lysing T cells is advantageous for both bone marrow and xenograft survival. Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable. Monoclonal preparations can be used in the methods of the invention.
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species-specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus; and the step of prior to hematopoietic stem cell transplantation, introducing into the recipient mammal an antibody capable of binding to mature T cells of the recipient mammal.
  • Preferred embodiments include those in which: the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells; the hematopoietic stem cells are introduced simultaneously with, or prior to, the antibody; the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells, and the hematopoietic stem cells are introduced simultaneously with, or prior to, the antibody.
  • Preferred embodiments include those in which: the same mammal of the second species is the donor of both the graft and the hematopoietic cells; the donor mammal is a swine, e.g., a miniature swine; the introduction is by intravenous injection; and an anti-human thymocyte polyclonal anti-serum; obtained, e.g. from a horse or pig is administered.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • Preferred embodiments include: (preferably prior to hematopoietic stem cell transplantation) depleting, inactivating, or inhibiting recipient T cells, e.g., by introducing into the recipient an antibody capable of binding to mature T cells of the recipient.
  • Preferably the graft is obtained from a different organ than the hematopoietic stem cells.
  • Preferred embodiments include those in which: the primate is a cynomolgus monkey; the primate is a human; the stromal tissue is fetal or neonatal liver; the stromal tissue is fetal or neonatal thymus; the mammal is a swine; e.g., a miniature swine; the graft is a liver; the graft is a kidney.
  • Other methods can be combined with the methods disclosed herein to promote the acceptance of the graft by the recipient. For example, tolerance to the xenogeneic thymic tissue can also be induced by inserting a nucleic acid which expresses a donor antigen, e.g., a donor MHC gene, into a cell of the recipient, e.g., a hematopoietic stem cell, and introducing the genetically engineered cell into the recipient. For example, human recipient stem cells can be engineered to express a swine class I or class II MHC gene, or both a class I and II gene, and the cells implanted in a human recipient who will receive swine thymic tissue. When inserted into a recipient primate, e.g., a human, expression of the donor MHC gene results in tolerance to subsequent exposure to donor antigen, and can thus induce tolerance to thymic tissue from the donor. These methods, and other methods which can be combined with the methods disclosed herein, are discussed in Sachs, U.S. Ser. No. 08/126, 122, filed Sep. 23, 1993, and in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi, and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance may also be used to promote acceptance of the xenogeneic thymic tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994, hereby. incorporated by reference.
  • Other methods of promoting tolerance or promoting the acceptance of grafts, e.g., by altering levels of cytokine activity, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072, filed Aug. 30, 1993.
  • In another aspect, the invention features a method of inducing immunological tolerance in a recipient mammal, e.g., a primate, e.g., a human to a graft obtained from a donor mammal of the same species. The method includes the following: (preferably prior to or simultaneous with transplantation of the graft) introducing into the recipient mammal hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, obtained from a mammal (preferably, the hematopoietic stem cells home to a site in the recipient mammal); and, preferably, introducing the graft into the recipient.
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species-specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus; and prior to hematopoietic stem cell transplantation, depleting, inactivating or inhibiting recipient T cells, e.g., by introducing into the recipient mammal an antibody capable of binding to mature T cells of the recipient mammal.
  • Other preferred embodiments include the step of (preferably prior to thymic tissue or hematopoietic stem cell transplantation) creating hematopoietic space, e.g., by one or more of: irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation, the administration of a myelosuppressive drug, the administration of a hematopoietic stem cell inactivating or depleting antibody, to deplete or partially deplete the bone marrow of the recipient.
  • Other preferred embodiments include (preferably prior to thymic tissue or hematopoietic stem cell transplantation) inactivating thymic T cells by one or more of: irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, administering to the recipient one or more doses of an anti T cell antibody, e.g., an anti-CD4 and/or an anti-CD8 monoclonal antibody, or administering to the recipient a short course of an immunosuppressant, as is described in U.S. Ser. No. 08/220,371 and further described below.
  • Other preferred embodiments include depleting or otherwise inactivating natural antibodies, e.g., by one or more of: the administration of a drug which depletes or inactivates natural antibodies, e.g., deoxyspergualin; the administration of an anti-IgM antibody; or the adsorption of natural antibodies from the recipient's blood, e.g., by contacting the hosts blood with donor antigen, e.g., by hemoperfusion of a donor organ, e.g., a kidney or a liver, from the donor species.
  • In other preferred embodiments; the method includes: (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting recipient natural killer (NK) cells, e.g., by introducing into the recipient an antibody capable of binding to NK cells of the recipient, to prevent NK mediated rejection of the thymic tissue; (preferably prior to or at the time of introducing the thymic tissue into the recipient) depleting, inactivating or inhibiting host T cell function, e.g., by introducing into the recipient an antibody capable of binding to T cells of the recipient.
  • Other methods can be combined with-the methods disclosed herein to promote tolerance to a graft. Methods of inducing tolerance, e.g., by the implantation of hematopoietic stem cells, disclosed in Sachs, Cosimi, and Sykes, U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, can also be combined with the methods disclosed herein.
  • Other methods of inducing tolerance may also be used to promote acceptance of donor tissue. For example, suppression of T cell help, which can be induced, e.g., by the administration of a short course of high dose immunosuppressant, e.g., cyclosporine, has been found to induce tolerance. In these methods, T cell help is suppressed for a comparatively short period just subsequent to implantation of a graft, and does not require or include chronic immunosuppression. These methods, as well as other methods which can be combined with the methods disclosed herein, are described in Sachs, U.S. Ser. No. 08/220,371, filed Mar. 29, 1994.
  • Other methods of promoting tolerance or promoting the acceptance of grafts, e.g., by altering levels of cytokine activity, are disclosed in Sachs, LeGuern, Sykes, and Blancho, U.S. Ser. No. 08/114,072, filed Aug. 30, 1993.
  • The invention further provides several methods of inducing tolerance to foreign antigens, e.g., to antigens on allogeneic or xenogeneic tissue or organ grafts. These methods can be used individually or in combination with one another. For example, it has been found that the short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA), can significantly prolong graft acceptance. The short term help reduction-methods of the invention can be combined with one or more other methods for prolonging graft acceptance. For example, a short course of high dose cyclosporine treatment to induce tolerance to unmatched donor class I and other minor unmatched donor antigens can be combined with implantation of retrovirally transformed bone marrow cells to induce tolerance to unmatched donor class II. A short course of high dose cyclosporine administered to induce tolerance to unmatched donor class I and other minor antigens can also be combined with implantation of donor bone marrow cells to induce tolerance to unmatched donor class II.
  • Accordingly, the invention features, in one aspect, a method of inducing tolerance in a recipient mammal, e.g., a primate, e.g., a human, to an allograft from a donor primate including: implanting the graft in the recipient; and administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine. The short course of help reducing treatment is generally administered at about the time the graft is introduced into the recipient.
  • Preferably, the recipient is mismatched at a first locus which affects graft rejection, e.g., an MHC class I or II locus, or a minor antigen locus, and matched, or tolerant of a mismatch, at a second locus which affects graft rejection, e.g., an MHC class I or II locus, or a minor antigen locus. Matching at the second locus can be achieved by selection of a recipient or donor of the appropriate genotype. The recipient can be rendered tolerant of a mismatch at the second locus by any method of tolerance induction, e.g., by administering donor bone marrow tissue to the recipient to induce tolerance to donor antigens expressed on the donor bone marrow, by expressing an MHC antigen of the donor from a stem cell of the recipient to induce tolerance to the donor antigen, or by altering the immunological properties of the graft, e.g., by masking, cleaving, or otherwise modifying cell surface molecules on the graft. In preferred embodiments, any of the methods which can be used to match or induce tolerance to the second locus can be used to match or induce tolerance to a third locus which affects graft rejection, e.g., an MHC class I or II locus, or a minor antigen locus.
  • In preferred embodiments, the recipient and donor are matched at a class II locus and the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on the graft. In preferred embodiments, tolerance to a class II antigen is induced by a method other than a short course of a help reducing treatment, and the short course of help reducing treatment induces tolerance to unmatched class I and minor antigens on the graft.
  • In preferred embodiments, the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen (in humans this is usually 8-12 days, preferably about 10 days); in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for mature T cells of the recipient to initiate rejection of an antigen after first being stimulated by the antigen.
  • In other preferred embodiments, the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g. in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient. In preferred embodiments, the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone.
  • In preferred embodiments: the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative, or the short course is postoperative; or the donor and recipient are class I matched.
  • Methods of inducing tolerance by a short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA), can be combined with other methods for inducing tolerance, e.g., methods for the implantation of transduced bone marrow cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 008/126,122, filed on Sep. 23, 1993.
  • Accordingly, in another aspect, the invention features a method of inducing tolerance in a recipient mammal, e.g., a primate, e.g., a human, of a first species to a graft from a mammal, e.g., a swine, e.g., a miniature swine, of a second species, which graft preferably expresses a major histocompatibility complex (MHC) antigen. The method includes inserting DNA encoding an MHC antigen of the second species into a hematopoietic stem cell, e.g., a bone marrow hematopoietic stem cell, of the recipient mammal; allowing the MHC antigen encoding DNA to be expressed in the recipient; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine treatment. The short course of help reducing treatment is generally administered at about the time the graft is introduced into the recipient.
  • In preferred embodiments, the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on a graft which is introduced into the recipient subsequent to expression of the MHC antigen.
  • In preferred embodiments, the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • In other preferred embodiments, the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient. In preferred embodiments, the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone.
  • In preferred embodiments: the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative; or the short course is postoperative.
  • Preferred embodiments include those in which: the cell is removed from the recipient mammal prior to the DNA insertion and returned to the recipient mammal after the DNA insertion; the DNA is obtained from the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is syngeneic with the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is MHC matched, and preferably identical, with the individual mammal from which the graft is obtained; the DNA includes an MHC class I gene; the DNA includes an MHC class II gene; the DNA is inserted into the cell by transduction, e.g., by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient for at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of: prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the step of, prior to implantation of a graft, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • In other preferred embodiments: the method further includes, prior to hematopoietic stem cell transplantation, introducing into the recipient an antibody capable of binding to mature T cells of said recipient mammal.
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a liver or a kidney.
  • In another aspect, the invention features a method of inducing tolerance in a recipient mammal, preferably a primate, e.g., a human, to a graft obtained from a donor of the same species, which graft preferably expresses an MHC antigen. The method includes: inserting DNA encoding an MHC antigen of the donor into a hematopoietic stem cell, e.g., bone marrow hematopoietic stem cell, of the recipient; allowing the MHC antigen encoding DNA to be expressed in the recipient; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine. The short course of help reducing treatment is generally administered at about the time the graft is introduced into the recipient.
  • In preferred embodiments, the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on a graft which is introduced into the recipient subsequent to expression of the MHC antigen.
  • In preferred embodiments, the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • In other preferred embodiments, the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient. In preferred embodiments, the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone
  • In preferred embodiments: the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative, or the short course is postoperative; or the donor and recipient are class I matched.
  • Preferred embodiments include those in which: the cell is removed from the recipient prior to the DNA insertion and returned to the recipient after the DNA insertion; the DNA includes a MHC class I gene; the DNA includes a MHC class II gene; the DNA is inserted into the cell by transduction, e.g. by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • In other preferred embodiments: the method further includes, prior to hematopoietic stem cell transplantation, introducing into the recipient an antibody capable of binding to mature T cells of said recipient mammal.
  • In preferred embodiments the graft is a liver or a kidney.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of: prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the step of, prior to implantation of a graft, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • Methods of inducing tolerance with a short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA), can be combined with other methods for inducing tolerance, e.g., methods of inducing tolerance which use the implantation of donor stem cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 07/838,595, filed Feb. 19, 1992.
  • Accordingly, in another aspect, the invention features a method of inducing tolerance in a recipient mammal of a first species, e.g., a primate, e.g., a human, to a graft obtained from a mammal of a second, preferably discordant species, e.g., a swine, e.g., a miniature swine, or a discordant primate species. The method includes: preferably prior to or simultaneous with transplantation of the graft, introducing, e.g., by intravenous injection, into the recipient mammal, hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, of the second species (preferably the hematopoietic stem cells home to a site in the recipient mammal); (optionally) inactivating the natural killer cells of the recipient mammal, e.g., by prior to introducing the hematopoietic stem cells into the recipient mammal, introducing into the recipient mammal an antibody capable of binding to natural killer cells of said recipient mammal; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine. The short course of help reducing treatment is generally administered at the time at the graft is introduced into the recipient.
  • In preferred embodiments, the short course of help reducing treatment induces tolerance to unmatched class I and/or minor antigens on the graft which is introduced into the recipient.
  • In preferred embodiments, the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times, the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • In other preferred embodiments, the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient. In preferred embodiments, the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone.
  • In preferred embodiments: the help reducing treatment is begun before or at about the time the graft is introduced; or the short course is perioperative, the short course is postoperative.
  • As will be explained in more detail below, the hematopoietic cells prepare the recipient for the graft that follows, by inducing tolerance at both the B-cell and T-cell levels. Preferably, hematopoietic cells are fetal liver or spleen, or bone marrow cells, including immature cells (i.e., undifferentiated hematopoietic stem cells; these desired cells can be separated out of the bone marrow prior to administration), or a complex bone marrow sample including such cells can be used.
  • One source of anti-NK antibody is anti-human thymocyte polyclonal anti-serum. As is discussed below, preferably, a second anti-mature T cell antibody can be administered as well, which lyses T cells as well as NK cells. Lysing T cells is advantageous for both bone marrow and xenograft survival. Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable. Monoclonal preparations can be used in the methods of the invention.
  • Other preferred embodiments include: the step of introducing into the recipient mammal, donor species-specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus. In preferred embodiments: the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells; the hematopoietic stem cells are introduced simultaneously with, or prior to, the antibody.
  • Other preferred embodiments include those in which: the same mammal of the second species is the donor of one or both the graft and the hematopoietic cells; and the antibody is an anti-human thymocyte polyclonal anti-serum, obtained, e.g., from a horse or pig.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of: prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • In other preferred embodiments: the method further includes, prior to hematopoietic stem cell transplantation, introducing into the recipient an antibody capable of binding to mature T cells of said recipient mammal.
  • In other preferred embodiments: the method further includes inactivating T cells of the recipient, e.g., by, prior to introducing the hematopoietic stem cells into the recipient, introducing into the recipient an antibody capable of binding to T cells of the recipient.
  • In preferred embodiments, the method includes the step of introducing into the recipient a graft obtained from the donor which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • In another aspect, the invention features a method of inducing tolerance in a recipient mammal, preferably a primate, e.g., a human, to a graft obtained from a donor, e.g., of the same species. The method includes: preferably prior to or simultaneous with transplantation of the graft, introducing, e.g., by intravenous injection, into the recipient, hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, of a mammal, preferably the donor (preferably the hematopoietic stem cells home to a site in the recipient); (optionally), inactivating T cells of the recipient, e.g., by, prior to introducing the hematopoietic stem cells into the recipient, introducing into the recipient an antibody capable of binding to T cells of the recipient; preferably, implanting the graft in the recipient; and, preferably, administering to the recipient a short course of help reducing treatment, e.g., a short course of high dose cyclosporine. The short course of help reducing treatment is generally administered at the time the graft is introduced into the recipient.
  • In preferred embodiments, the short course of help reducing treatment induces tolerance to unmatched class I and minor antigens on the graft which is introduced into the recipient.
  • In preferred embodiments, the duration of the short course of help reducing treatment is approximately equal to or is less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen; in more preferred embodiments, the duration is approximately equal to or is less than two, three, four, five, or ten times the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen.
  • In other preferred embodiments, the short course of help reducing treatment is administered in the absence of a treatment which stimulates the release of a cytokine by mature T cells in the recipient, e.g., in the absence of a steroid drug in a sufficient concentration to counteract the desired effect of the help reducing treatment, e.g., in the absence of Prednisone (17,21-dihydroxypregna-1,4-diene-3,11,20-trione) at a concentration which stimulates the release of a cytokine by mature T cells in the recipient. In preferred embodiments, the short course of help reducing treatment is administered in the absence of a steroid drug, e.g., in the absence of Prednisone
  • In preferred embodiments: the help reducing treatment is begun before or at about the time the graft is introduced; the short course is perioperative, the short course is postoperative; the donor and recipient are class I matched.
  • In preferred embodiments, the hematopoietic stem cells are introduced simultaneously with, or prior to administration of the antibody; the antibody is an antihuman thymocyte polyclonal anti-serum; and the anti-serum is obtained from a horse or pig.
  • Other preferred embodiments include: the further step of, prior to hematopoietic stem cell transplantation, inactivating or depleting NK cells of the recipient, e.g., by introducing into the recipient mammal an antibody capable of binding to NK cells of the recipient mammal; and those in which the same individual is the donor of both the graft and the bone marrow.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient; inactivating thymic T cells by one or more of, prior to hematopoietic stem cell transplantation, irradiating the recipient mammal with, e.g., about 700 rads of thymic irradiation, or administering to the recipient a short course of an immunosuppressant, as is described herein.
  • Other preferred embodiments include: the further step of, prior to bone marrow transplantation, adsorbing natural antibodies from the blood of the recipient by hemoperfusing an organ, e.g., the liver, or a kidney, obtained from the donor.
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • In preferred embodiments, the method includes the step of introducing into the recipient, a graft which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • Methods of inducing tolerance with short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA), can be combined with yet other methods for inducing tolerance, e.g., with: methods which use the implantation of a xenogeneic thymic graft to induce tolerance, e.g., the methods described in U.S. Ser. No. 08/163, 912 filed on Dec. 7, 1993; methods of increasing the level of the activity of a tolerance promoting or GVHD inhibiting cytokine or decreasing the level of activity of a tolerance inhibiting or GVHD promoting cytokine, e.g., the methods described in U.S. Ser. No. 08/114,072, filed Aug. 30, 1993; methods of using cord blood cells to induce tolerance, e.g., the methods described in U.S. Ser. No. 08/150,739 filed Nov. 10, 1993; and the methods for inducing tolerance disclosed in Sykes and Sachs, PCT/US94/01616, filed Feb. 14, 1994.
  • It has also been discovered that a short course of an immunosuppressant, e.g., cyclosporine, can be used to diminish or inhibit T cell activity which would otherwise promote the rejection of an allograft or xenograft.
  • Accordingly, in another aspect, the invention features a method of diminishing or inhibiting T cell activity, preferably the activity of thymic or lymph node T cells, in a recipient mammal, e.g., a primate, e.g., a human, which receives a graft from a donor mammal. The method includes, inducing tolerance to the graft; administering to the recipient a short course of an immunosuppressive agent, e.g., cyclosporine, sufficient to inactivate T cells, preferably thymic or lymph node T cells; and preferably transplanting the graft into the recipient.
  • Tolerance to the graft can be induced by any method, e.g., by any of the methods discussed herein. For example, tolerance can be induced by the administration of donor allogeneic or xenogeneic hematopoietic stem cells, the administration of genetically engineered autologous stem cells, by the administration of a short course of a help reducing agent, or by altering the immunological properties of the graft, e.g., by masking, cleaving, or otherwise modifying cell surface molecules of the graft.
  • In preferred embodiments the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period.
  • In preferred embodiments: the short course is begun before or at about the time the treatment to induce tolerance is begun, e.g., at about the time, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient; the short course begins on the day the treatment to induce tolerance is begun, e.g., on the day, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the treatment to induce tolerance is begun, e.g., within 1, 2, 4, 6, 8, or 10 days before or after xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient.
  • In other preferred embodiments: the short course of an immunosuppressive is administered in conjunction with an anti-T cell antibody; the short course of an immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG antibody, or similar, preparations.
  • In preferred embodiments: the recipient mammal is other than a mouse or rat.
  • Methods of inactivating T cells, preferably thymic or lymph node T cells, of the invention can be combined with methods of inducing tolerance in which the inactivation of T cells is desirable. The anti-T cell methods of the invention can be used in place of, or in addition to, methods for the inactivation of T cells called for, or useful in such methods of inducing tolerance. For example, anti-thymic or lymph node T cell methods of the invention can be used with methods for the implantation of transduced bone marrow cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 008/126,122, filed on Sep. 23, 1993.
  • Accordingly, in another aspect, the invention features a method of promoting, in a recipient mammal of a first species, the acceptance of a graft from a donor mammal of a second species, which graft, preferably, expresses a major histocompatibility complex (MHC) antigen. The method includes inserting DNA encoding an MHC antigen of the second species into a hematopoietic stem cell, e.g., a bone marrow hematopoietic stem cell, of the recipient mammal; allowing the MHC antigen encoding DNA to be expressed in the recipient; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells. (Thymic or lymph node T cells might otherwise inhibit the survival of the graft or engineered cells.)
  • In preferred embodiments, the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period.
  • In preferred embodiments: the recipient mammal is a primate, e.g., a human, and the donor mammal is a swine, e.g., a miniature swine.
  • In preferred embodiments: the short course is begun before or at about the time genetically engineered stem cells are introduced into the recipient; the short course begins on the day the genetically engineered stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the genetically engineered stem cells are introduced into the recipient.
  • In other preferred embodiments: the short course of an immunosuppressive agent is administered in conjunction with an anti-T cell antibody; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG, or similar, antibody preparations.
  • Preferred embodiments include those in which: the cell is removed from the recipient mammal prior to the DNA insertion and returned to the recipient mammal after the DNA insertion; the DNA is obtained from the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is syngeneic with the individual mammal from which the graft is obtained; the DNA is obtained from an individual mammal which is MHC matched, and preferably identical, with the individual mammal from which the graft is obtained; the DNA includes an MHC class I gene; the DNA includes an MHC class II gene; the DNA is inserted into the cell by transduction, e.g., by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient for at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell implantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • In preferred embodiments, the method further includes the administration of thymic irradiation to the recipient, e.g., 700 rads of thymic irradiation.
  • Other preferred embodiments include: the step of depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a liver or a kidney.
  • In another aspect, the invention features a method of promoting, in a recipient mammal, preferably a primate, e.g., a human, acceptance of a graft obtained from a donor of the same species, which graft expresses an MHC antigen. The method includes: inserting DNA encoding an MHC antigen of the donor into a hematopoietic stem cell, e.g., a bone marrow hematopoietic stem cell, of the recipient; allowing the MHC antigen encoding DNA to be expressed in the recipient; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells. (Thymic or lymph node T cells might otherwise inhibit the survival of the graft or engineered cells.)
  • In preferred embodiments, the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period.
  • In preferred embodiments: the short course is begun before or at about the time genetically engineered stem cells are introduced into the recipient; the short course begins on the day the genetically engineered stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the genetically engineered stem cells are introduced into the recipient.
  • In other preferred embodiments: the short course of an immunosuppressive agent is administered in conjunction with an anti-T cell antibody; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG, or similar, antibody preparations.
  • Preferred embodiments include those in which: the cell is removed from the recipient prior to the DNA insertion and returned to the recipient after the DNA insertion; the DNA includes a MHC class I gene; the DNA includes a MHC class II gene; the DNA is inserted into the cell by transduction, e.g. by a retrovirus, e.g., by a Moloney-based retrovirus; and the DNA is expressed in bone marrow cells and/or peripheral blood cells of the recipient at least 14, preferably 30, more preferably 60, and most preferably 120 days, after the DNA is introduced into the recipient.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell implantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • In preferred embodiments, the method further includes the administration of thymic irradiation to the recipient, e.g., 700 rads of thymic irradiation.
  • Other preferred embodiments include: the step of depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a liver or a kidney.
  • Methods of inactivating T cells, preferably thymic or lymph node T cells, of the invention can be combined with methods of inducing tolerance which use the implantation of donor stem cells to induce tolerance to an antigen, e.g., the methods described in U.S. Ser. No. 07/838,595, filed Feb. 19, 1992, hereby incorporated by reference.
  • Accordingly, in another aspect, the invention features a method of promoting, in a recipient mammal of a first species, e.g., a primate, e.g., a human, acceptance of a graft obtained from a mammal of a second, preferably discordant species, e.g., a swine, e.g., a miniature swine, or a discordant primate species. The method includes: introducing, e.g., by intravenous injection, into the recipient mammal, hematopoietic stem cells, e.g., bone marrow cells or fetal liver or spleen cells, of the second species (preferably the hematopoietic stem cells home to a site in the recipient mammal); (optionally) inactivating natural killer cells of the recipient mammal, e.g., by, prior to introducing the hematopoietic stem cells into the recipient mammal, introducing into the recipient mammal an antibody capable of binding to natural killer cells of said recipient mammal; (optionally) inactivating T cells of the recipient mammal, e.g., by, prior to introducing the hematopoietic stem cells into the recipient mammal, introducing into the recipient mammal an antibody capable of binding to T cells of the recipient mammal; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells. (Thymic or lymph node T cells might otherwise inhibit the engraftment or survival of the engineered cells.)
  • In preferred embodiments, the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period mentioned above.
  • In preferred embodiments: the short course is begun before or at about the time stem cells are introduced into the recipient; the short course begins on the day the stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the stem cells are introduced into the recipient.
  • In other preferred embodiments: the short course of an immunosuppressive agent is administered in conjunction with one or both of an anti-T cell antibody, or thymic irradiation, e.g., 700 rads of thymic irradiation; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG antibody preparations.
  • As will be explained in more detail below, the hematopoietic cells prepare the recipient for the graft that follows, by inducing tolerance at both the B-cell and T-cell levels. Preferably, hematopoietic cells are fetal liver or spleen, or bone marrow cells, including immature cells (i.e., undifferentiated hematopoietic stem cells; these desired cells can be separated out of the bone marrow prior to administration), or a complex bone marrow sample including such cells can be used.
  • One source of anti-NK antibody is anti-human thymocyte polyclonal anti-serum. As is discussed below, preferably, a second anti-mature T cell antibody can be administered as well, which lyses T cells as well as NK cells. Lysing T cells is advantageous for both bone marrow and xenograft survival. Anti-T cell antibodies are present, along with anti-NK antibodies, in anti-thymocyte anti-serum. Repeated doses of anti-NK or anti-T cell antibody may be preferable. Monoclonal preparations can be used in the methods of the invention.
  • Other preferred embodiments include: the step of introducing into the recipient mammal, donor species-specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus. In preferred embodiments: the stromal tissue is introduced simultaneously with, or prior to, the hematopoietic stem cells; the hematopoietic stem cells are introduced simultaneously with, or prior to, an anti-NK or T cell antibody.
  • Other preferred embodiments include those in which: the same mammal of the second species is the donor of one or both the graft and the hematopoietic cells; and the anti-T or anti-NK cell antibody is an anti-human thymocyte polyclonal anti-serum, obtained, e.g., from a horse or pig.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • In preferred embodiments, the method further includes the administration of thymic irradiation to the recipient, e.g., 300 to 700 rads of thymic irradiation.
  • Other preferred embodiments include: the step of prior to hematopoietic stem cell transplantation, depleting natural antibodies from the blood of the recipient mammal, e.g., by hemoperfusing an organ, e.g., a liver or a kidney, obtained from a mammal of the second species. (In organ hemoperfusion antibodies in the blood bind to antigens on the cell surfaces of the organ and are thus removed from the blood.)
  • Other preferred embodiments further include the step of introducing into the recipient a graft obtained from the donor, e.g., a graft which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • In preferred embodiments the stem cells are introduced into the recipient prior to or simultaneous with transplantation of the graft.
  • In another aspect, the invention features a method of promoting, in a recipient mammal, preferably a primate, e.g., a human, acceptance of a graft obtained from a donor of the same species. The method includes: introducing, e.g., by intravenous injection into the recipient, hematopoietic stem cells, e.g. bone marrow cells or fetal liver or spleen cells, of a mammal, preferably the donor (preferably the hematopoietic stem cells home to a site in the recipient); (optionally) inactivating T cells of the recipient, e.g., by, prior to introducing the hematopoietic stem cells into the recipient, introducing into the recipient an antibody capable of binding to T cells of the recipient; and, preferably, administering to the recipient a short course of an immunosuppressive agent, e.g., a short course of cyclosporine treatment, sufficient to inactivate recipient T cells, preferably thymic or lymph node T cells. (Thymic or lymph node T cells might otherwise inhibit the engraftment or survival of the engineered cells.)
  • In preferred embodiments, the duration of the short course of immunosuppressive agent is: approximately equal to 30 days; approximately equal to or less than 8-12 days, preferably about 10 days; approximately equal to or less than two, three, four, five, or ten times the 8-12 or 10 day period mentioned above.
  • In preferred embodiments: the short course is begun before or at about the time stem cells are introduced into the recipient; the short course begins on the day the stem cells are introduced into the recipient; the short course begins within 1, 2, 4, 6, 8, or 10 days before or after the stem cells are introduced into the recipient.
  • In other preferred embodiments: the short course of an immunosuppressive agent is administered in conjunction with one or both of an anti-T cell antibody, or thymic irradiation, e.g., 700 rads of thymic irradiation; the short course of immunosuppressive is sufficient to inactivate T cells, e.g., thymic or lymph node T cells, which would not be inactivated by antibody-based inactivation of T cells, e.g., inactivation by intravenous administrations of ATG antibody preparations.
  • In preferred embodiments, the anti-T cell or NK cell antibody is an antihuman thymocyte polyclonal anti-serum; and the anti-serum is obtained from a horse or pig.
  • Other preferred embodiments include: the further step of, prior to hematopoietic stem cell transplantation, inactivating recipient NK cells, e.g., by introducing into the recipient mammal an antibody capable of binding to NK cells of the recipient mammal; and those in which the same individual is the donor of both the graft and the bone marrow.
  • Other preferred embodiments include: the step of, prior to hematopoietic stem cell transplantation, creating hematopoietic space in the recipient so as to promote engraftment and survival of the implanted stem cells, e.g., by irradiating the recipient mammal with low dose, e.g., between about 100 and 400 rads, whole body irradiation to deplete or partially deplete the bone marrow of the recipient.
  • In preferred embodiments the method further includes administering thymic irradiation to the recipient, e.g., 700 rads of thymic irradiation.
  • Other preferred embodiments include: the further step of, prior to bone marrow transplantation, adsorbing natural antibodies from the blood of the recipient by hemoperfusing an organ, e.g., the liver, or a kidney, obtained from the donor.
  • Preferred embodiments include: the step of introducing into the recipient mammal, donor species specific stromal tissue, preferably hematopoietic stromal tissue, e.g., fetal liver or thymus.
  • Other preferred embodiments further include the step of introducing into the recipient, a graft obtained from the donor, e.g., a graft which is obtained from a different organ than the hematopoietic stem cells, e.g., a liver or a kidney.
  • In preferred embodiments, the stem cells are introduced into the recipient prior to or simultaneous with transplantation of the graft.
  • Methods of inactivating T cells, preferably thymic or lymph node T cells, of the invention can be used with yet other methods of inducing tolerance in which the inactivation of thymic or lymph node T cells is desirable. For example, anti-thymic or lymph node T cell methods of the invention can be used with: methods which use the implantation of a xenogeneic thymic graft to induce tolerance, e.g., the methods described in U.S. Ser. No. 08/163, 912 filed on Dec. 7, 1993; methods of increasing the level of the activity of a tolerance promoting or GVHD inhibiting cytokine or decreasing the level of activity of a tolerance inhibiting or GVHD promoting cytokine, e.g., the methods described in U.S. Ser. No. 08/114,072, filed Aug. 30, 1993; methods of using cord blood cells to induce tolerance, e.g., the methods described in U.S. Ser. No. 08/150,739; and the methods for inducing tolerance disclosed in Sykes and Sachs, PCT/US94/01616, filed Feb. 14, 1994.
  • “Thymus-function deficient”, as used herein, refers to a condition in which the ability of an individual's thymus to support the maturation of T cells is impaired as compared with a normal individual. Thymus deficient conditions include those in which the thymus or thymus function is essentially absent.
  • “Tolerance”, as used herein, refers to the inhibition of a graft recipient's ability to mount an immune response, e.g., to a donor antigen, which would otherwise occur, e.g., in response to the introduction of a non self MHC antigen into the recipient. Tolerance can involve humoral, cellular, or both humoral and cellular responses. The concept of tolerance includes both complete and partial tolerance. In other words, as used herein, tolerance include any degree of inhibition of a graft recipient's ability to mount an immune response, e.g., to a donor antigen.
  • “A discordant species combination”, as used herein, refers to two species in which hyperacute rejection occurs when vascular organs are grafted. Generally, discordant species are from different orders, while non-discordant species are from the same order. For example, rats and mice are non-discordant species, i.e. their MHC antigens are substantially similar, and they are members of the same order, rodentia.
  • “Hematopoietic stem cell”, as used herein, refers to a cell that is capable of developing into mature myeloid and/or lymphoid cells. Preferably, a hematopoietic stem cell is capable of the long-term repopulation of the myeloid and/or lymphoid lineages. Stem cells derived from the cord blood of the recipient or the donor can be used in methods of the invention. See U.S. Pat. No. 5,192,553, hereby incorporated by reference, and U.S. Pat. No. 5,004,681, hereby incorporated by reference.
  • “Miniature swine”, as used herein, refers to completely or partially inbred miniature swine.
  • “Graft”, as used herein, refers to a body part, organ, tissue, cells, or portions thereof
  • “Stromal tissue”, as used herein, refers to the supporting tissue or matrix of an organ, as distinguished from its functional elements or parenchyma.
  • Restoring, inducing, or promoting immunocompetence, as used herein, means one or both of: (1) increasing the number of mature functional T cells in the recipient (over what would be seen in the absence of treatment with a method of the invention) by either or both, increasing the number of recipient-mature functional T cells or by providing mature functional donor-T cells, which have matured in the recipient; or (2) improving the immune-responsiveness of the recipient, e.g., as is measured by the ability to mount a skin response to a recall antigen, or improving the responsiveness of a of a T cell of the recipient, e.g., as measured by an in vitro test, e.g., by the improvement of a proliferative response to an antigen, e.g., the response to tetanus antigen or to an alloantigen.
  • A mature functional T cell, as used herein, is a T cell (of recipient or donor origin) which responds to microbial antigens and tolerant to recipient and donor tissue.
  • “Lymph node or thymic T cell”, as used herein, refers to T cells which are resistant to inactivation by traditional methods of T cell inactivation, e.g., inactivation by a single intravenous administration of anti-T cell antibodies, e.g., anti-bodies, e.g., ATG preparation.
  • Restoring or inducing the thymus-dependent ability for T cell progenitors to mature into mature T cells, as used herein, means either or both, increasing the number of functional mature T cells of recipient origin in a recipient, or providing mature functional donor T cells to a recipient, by providing donor thymic tissue in which T cells can mature. The increase can be partial, e.g., an increase which does not bring the level of mature functional T cells up to a level which results in an essentially normal immune response or partial, e.g., an increase which falls short of bringing the recipient's level of mature functional T cells up to a level which results in an essentially normal immune response.
  • Methods of the invention will allow the induction of immunocompetence in patients suffering from an immunodeficiency, e.g., a T cell deficiency, e.g., a thymic based immunodeficiency, e.g., a congenital immunodeficiency due to thymic aplasia or dysfunction, an acquired immune disorder, e.g., AIDS, immunoincompetence resulting form a neoplastic disease, or immunoincompetence resulting from a medical procedure, e.g., chemotherapy or radiation treatment. An acquired immune deficiency is one which is due primarily to other than genetic defects.
  • At risk for AIDS, as used herein, refers to being HIV positive or having AIDS.
  • “An immunosuppressive agent capable of inactivating thymic or lymph node T cells”, as used herein, is an agent, e.g., a chemical agent, e.g., a drug, which, when administered at an appropriate dosage, results in the inactivation of thymic or lymph node T cells. Examples of such agents are cyclosporine, FK-506, and rapamycin. Anti-T cell antibodies, because they are comparatively less effective at inactivating thymic or lymph node T cells, are not preferred for use as agents. An agent should be administered in sufficient dose to result in significant inactivation of thymic or lymph node T cells which are not inactivated by administration of an anti-T cell antibody, e.g., an anti-ATG preparation. Putative agents, and useful concentrations thereof, can be prescreened by in vitro or in vivo tests, e.g., by administering the putative agent to a test animal, removing a sample of thymus or lymph node tissue, and testing for the presence of active T cells in an in vitro or in vivo assay. Such prescreened putative agents can then be further tested in transplant assays.
  • “Short course of a immunosuppressive agent”, as used herein, means a transitory non-chronic course of treatment. The treatment should begin before or at about the time the treatment to induce tolerance is begun, e.g., at about the time, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient. e.g., the short course can begin on the day the treatment to induce tolerance is begun, e.g., on the day, xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient or the short course can begin within 1, 2, 4, 6, 8, or 10 days before or after the treatment to induce tolerance is begun, e.g., within 1, 2, 4, 6, 8, or 10 days before or after xenogeneic, allogeneic, genetically engineered syngeneic, or genetically engineered autologous stem cells are introduced into the recipient. The short course can last for: a period equal to or less than about 8-12 days, preferably about 10 days, or a time which is approximately equal to or is less than two, three, four, five, or ten times the 8-12 or 10 day period. Optimally, the short course lasts about 30 days. The dosage should be sufficient to maintain a blood level sufficient to inactivate thymic or lymph node T cells. A dosage of approximately 15 mg/kg/day has been found to be effective in primates.
  • “Help reduction”, as used herein, means the reduction of T cell help by the inhibition of the release of at least one cytokine, e.g., any of IL-2, IL-4, IL-6, gamma interferon, or TNF, from T cells of the recipient at the time of the first exposure to an antigen to which tolerance is desired. The inhibition induced in a recipient's T cell secretion of a cytokine must be sufficient such that the recipient is tolerized to an antigen which is administered during the reduction of help. Although not being bound by theory, it is believed that the level of reduction is one which substantially eliminates the initial burst of IL-2 which accompanies the first recognition of a foreign antigen but which does not eliminate all mature T cells, which cells may be important in educating and producing tolerance.
  • “A help reducing agent”, as used herein, is an agent, e.g., an immunosuppressive drug, which results in the reduction of cytokine release. Examples of help reducing agents are cyclosporine, FK-506, and rapamycin. Anti-T cell antibodies, because they can eliminate T cells, are not preferred for use as help reducing agents. A help reducing agent must be administered in sufficient dose to give the level of inhibition of cytokine release which will result in tolerance. The help reducing agent should be administered in the absence of treatments which promote cytokine, e.g., IL-2, release. Putative agents help reducing agents can be prescreened by in vitro or in vivo tests, e.g., by contacting the putative agent with T cells and determining the ability of the treated T cells to release a cytokine, e.g., IL-2. The inhibition of cytokine release is indicative of the putative agent's efficacy as a help reducing agent. Such prescreened putative agents can then be further tested in a kidney transplant assay. In a kidney transplant assay a putative help reducing agent is tested for efficacy by administering the putative agent to a recipient monkey and then implanting a kidney from a class II matched class I and minor antigen mismatched donor monkey into the recipient. Tolerance to the donor kidney (as indicated by prolonged acceptance of the graft) is indicative that the putative agent is, at the dosage tested, a help reducing agent.
  • “Short course of a help reducing agent”, as used herein, means a transitory non-chronic course of treatment. The treatment should begin before or at about the time of transplantation of the graft. Alternatively, the treatment can begin before or at about the time of the recipient's first exposure to donor antigens. Optimally, the treatment lasts for a time which is approximately equal to or less than the period required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen. The duration of the treatment can be extended to a time approximately equal to or less than two, three, four, five, or ten times, the period required for a mature T cell of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen. The duration will usually be at least equal to the time required for mature T cells of the recipient species to initiate rejection of an antigen after first being stimulated by the antigen. In pigs and monkeys, about 12 days of treatment is sufficient. Experiments with cyclosporine A (10 mg/kg) in pigs show that 6 days is not sufficient. Other experiments in monkeys show that IL-2 administered on day 8, 9, or 10 of cyclosporine A treatment will result in rejection of the transplanted tissue. Thus, 8, 9, or 10 days is probably not sufficient in pigs. In monkeys, a dose of 10 mg/kg cyclosporine with a blood level of about 500-1,000 ng/ml is sufficient to induce tolerance to class II matched class I and minor antigen mismatched kidneys. The same blood level, 500-1,000 ng/ml, is sufficient to induce tolerance in pigs. Long-term administration of 5 mg/kg prevents rejection (by long term immune suppression) but does not result in tolerance.
  • The help suppressing methods of the invention avoid the undesirable side effects of long-term or chronic administration of the broad spectrum immune suppressants often used in transplantation. Long-term or chronic administration of drugs such as Prednisone, Imuran, CyA, and, most recently FK506, have all had an important impact on the field of transplantation. However, all of these drugs cause nonspecific suppression of the immune system which must be titrated sufficiently to avoid rejection while not completely eliminating immune function. Patients who must stay on chronic immunosuppressive therapy for the remainder of their lives face major complications arising from too much or too little immunosuppression, causing infection and rejection, respectively. The help suppressing methods of the invention are based on the administration of a transitory short term high dose course of a help reducing treatment.
  • “MHC antigen”, as used herein, refers to a protein product of one or more MHC genes; the term includes fragments or analogs of products of MHC genes which can evoke an immune response in a recipient organism. Examples of MHC antigens include the products (and fragments or analogs thereof) of the human MHC genes, i.e., the HLA genes. MHC antigens in swine, e.g., miniature swine, include the products (and fragments and analogs thereof) of the SLA genes, e.g., the DRB gene.
  • Recipient thymic or lymph node T cells are responsible for significant resistance to implanted grafts, e.g., to transplanted hematopoietic cells or transplanted organs. It has been found that the usual methods of T cell depletion or inactivation, e.g., the administration of anti-T cell antibodies, often fall short of an optimum level of T cell depletion or inactivation. In particular, such methods fail to provide optimum levels of depletion or inactivation of thymic or lymph node T cells. Methods of the invention in which a short course of an immunosuppressant, e.g., cyclosporine, capable of inactivating recipient thymic or lymph node T cells is administered to the recipient, result in more thorough inactivation of thymic or lymph node T cells and thus in improved acceptance of graft tissue.
  • The retroviral methods of the invention allow the reconstitution of a graft recipient's bone marrow with transgenic autologous bone marrow cells expressing allogeneic or xenogeneic MHC genes. Expression of the transgenic MHC genes confers tolerance to grafts which exhibit the products of these or closely related MHC genes. Thus, these methods provide for the induction of specific transplantation tolerance by somatic transfer of MHC genes. Retroviral methods of the invention avoid the undesirable side effects of broad spectrum immune suppressants which are often used in transplantation.
  • Tolerance to transplantation antigens can be achieved through induction of lymphohematopoietic chimerism by bone marrow transplantation (BMT). BMT across MHC barriers presents two major risks: if mature T cells are not removed from the marrow inoculum the recipient may develop severe graft versus host disease (GVHD); removal of these cells often leads to failure of engraftment. Retroviral methods of the invention, which induce specific tolerance by reconstitution of the recipient's bone marrow with autologous (as opposed to allogeneic or heterologous) bone marrow cells, allow tolerance to be conferred with minimal risk of GVHD and with minimal need to remove T cells from the marrow inoculum.
  • Retroviral methods of the invention can be combined with the help suppression and T cell inactivation methods of the invention to prolong graft acceptance.
  • Hematopoietic cell transplant methods of the invention avoid the undesirable side effects of broad spectrum immune suppressants which are often used in transplantation. Hematopoietic cell transplant methods of the invention can be combined with the help suppression and T cell elimination methods of the invention to prolong graft acceptance.
  • Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a graph showing growth of fetal pig THY/LIV graft vs. time after transplantation in the presence of mature mouse T cells in the periphery.
  • FIG. 1B is a dot plot analysis of live peripheral white blood cells of a representative animal 16 weeks post-transplant. Upper left quadrant 0.5%; upper right quadrant 12.1%.
  • FIG. 2 is a graph of mouse anti-pig mixed lymphocyte reactions (MLR's) performed to determine whether or not mouse T cells which matured in pig thymus grafts were tolerant to pig antigens.
  • FIG. 3 is a diagram of the GS4.5 retroviral construct.
  • FIG. 4 is a diagram of the GS4.5 proviral genome and the expected transcripts.
  • FIGS. 5 a and 5 b are representations of flow cytometry profile of transduced cells.
  • FIG. 6 is a diagram of the transduction assay.
  • FIG. 7 is a diagram of genetic maps of the C57BL/10, B10.AKM, and B10.MBR strains.
  • FIG. 8 is a diagram of the FACS profile of spleen cells from a recipient of transduced bone marrow.
  • FIGS. 9 a and 9 b are graphs of survival versus time in skin graft experiments.
  • FIGS. 10 a-d are diagrams of FACS analysis of thymocytes from graft rejecters, and controls.
  • FIG. 11 is a diagram of the N2-B19-H2b vector.
  • DETAILED DESCRIPTION
  • Maturation of Host T Cells in a Xenogeneic Thymus and Induction of Tolerance to a Xenograft by Xenogeneic Thymic Tissue
  • The following procedure was designed to promote the acceptance of a xenograft thymus by a host and thusly to either or both: 1, lengthen the time an implanted organ (a xenograft) survives in a xenogeneic host prior to rejection; and 2, provide xenogeneic thymic tissue in which host T cells can mature.
  • In the case of an organ transplant, the organ can be any organ, e.g., a liver, e.g., a kidney, e.g., a heart. The two main strategies are elimination of natural antibodies and transplantation of thymic tissue to induce tolerance.
  • Preparation of the recipient for either organ transplantation or thymus replacement includes any or all of the following steps. Preferably they are carried out in the following sequence.
  • First, a preparation of horse anti-human thymocyte globulin (ATG) is intravenously injected into the recipient. The antibody preparation eliminates mature T cells and natural killer cells. If not eliminated, mature T cells might promote rejection of both the thymic transplant and, after sensitization, the xenograft organ. The ATG preparation also eliminates natural killer (NK) cells. NK cells probably have no effect on an implanted organ, but might act immediately to reject the newly introduced thymic tissue. Anti-human ATG obtained from any mammalian host can also be used, e.g., ATG produced in pigs, although thus far preparations of pig ATG have been of lower titer than horse-derived ATG. ATG is superior to anti-NK monoclonal antibodies, as the latter are generally not lytic to all host NK cells, while the polyclonal mixture in ATG is capable of lysing all host NK cells. Anti-NK monoclonal antibodies can, however, be used. In a relatively severely immunocompromised individual this step may not be necessary. As host (or donor) T cells mature in the xenogeneic thymus they will be tolerant of the xenogeneic thymic tissue. Alternatively, as the host immune system is progressively restored, it may be desirable to treat the host to induce tolerance to the xenogeneic thymic tissue.
  • Optimally, the recipient can be thymectomized. In thymectomized recipients, recipient T cells do not have an opportunity to differentiate in the recipient thymus, but must differentiate in the donor thymus. In some cases it may be necessary to splenectomize the recipient in order to avoid anemia.
  • Second, the recipient can be administered low dose radiation. Although this step is thought to be beneficial in bone marrow transplantation (by creating hematopoietic space for newly injected bone marrow cells), it is of less importance in thymic grafts which are not accompanied by bone marrow transplantation. However, a sublethal dose e.g., a dose about equal to 100, or more than 100 and less than about 400, rads, whole body radiation, plus 700 rads of local thymic radiation, can be used.
  • Third, natural antibodies can be adsorbed from the recipient's blood. (This is of more importance in organ grafts but can be used in thymus replacement procedures as well.) Antibody removal can be accomplished by exposing the recipient's blood to donor or donor species antigens, e.g., by hemoperfusion of a liver of the donor species to adsorb recipient-natural antibodies. Pre-formed natural antibodies (nAb) are the primary agents of graft rejection. Natural antibodies bind to xenogeneic endothelial cells and are primarily of the IgM class. These antibodies are independent of any known previous exposure to antigens of the xenogeneic donor. B cells that produce these natural antibodies tend to be T cell-independent, and are normally tolerized to self antigen by exposure to these antigens during development. The mechanism by which newly developing B cells are tolerized is unknown. The liver is a more effective adsorber of natural antibodies than the kidney. Again, this step may not be required, at least initially, in a relatively severely immunocompromised patient.
  • Donor thymic tissue, preferably fetal or neonatal thymic tissue is implanted in the recipient. Fetal or neonatal liver or spleen tissue can be included.
  • While any of these procedures may aid the survival of implanted thymic tissue or another xenogeneic organ, best results are achieved when all steps are used in combination.
  • Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • The donor of the implant and the individual that supplies the tolerance-inducing thymic graft should be the same individual or should be as closely related as possible. For example, it is preferable to derive implant tissue from a colony of donors that is highly or completely inbred. The donor of the organ used for perfusion need not be closely related to the donor of the implant or thymic tissue.
  • Xenograft Thymic Tissue Transplantation: Detailed Protocol
  • Immunocompetent C57BL/10 (B10) mice were used to test the ability of pig thymus to induce specific tolerance to discordant pig antigens. B10 mice were treated with a non-myeloablative conditioning regimen which has previously shown to permit induction of tolerance to rat xeno-antigens in mice, see e.g., Sharabi et al., 1990, J. Exp. Med. 172:195-202. Euthymic or thymectomized (ATX) mice received depleting doses of anti-T cell and anti-NK cell mAbs, 7 Gy mediastinal irradiation and 3 Gy whole body irradiation (WBI), and then received fetal swine thymus/liver (THY/LIV) transplants under the kidney capsule followed by administration of 108 fetal liver cells (FLC) i.p. Mice either received no further anti-T cell and anti-NK cell mAb treatments after 0 to 6 weeks post-tx, or were maintained on chronic mAb treatment for the duration of the experiment.
  • Swine THY/LIV grafts grew initially in treated euthymic mice, but stopped growing after T cell and natural killer (NK) cell-depleting monoclonal antibodies (mAbs) were discontinued, and these mice developed anti-pig IgG response. When euthymic mice were maintained on chronic mAb treatment, the grafts enlarged markedly and no anti-pig IgG response was observed. Pig thymopoiesis was supported for at least 32 weeks during chronic mAb administration, although no pig T cells were detected in the periphery by flow cytometry (FCM). Percentages of intra-graft CD4+/CD8, CD4/CD8+, CD4+/CD8+, and CD4/CD8 pig thymocyte subsets were similar to those in normal pig thymus.
  • In contrast, swine THY/LIV grafts grew markedly in adult thymectomized mice (ATX-THY/LIV) which received only a short (less than 6 weeks) course of mAb treatment post-transplant (post-tx). FCM analysis of peripheral WBC in these mice 6 weeks after discontinuing mAb treatment revealed the presence of mature (αβ-TCRhi) mouse T cells. Unlike T cells in euthymic grafted mice, these cells were tolerant to pig antigens, as evidenced by the growth of swine THY/LIV grafts, (FIG. 1), and the absence of anti-pig IgG antibody responses. The majority (more than 90%) of the αβ-TCRhi T cells were CD4+/CD8. FMC analyses 13 to 26 weeks post-tx demonstrated normal mouse thymocyte subsets in swine thymi. For example, 7.9% CD4+/CD8, 2.9% CD4/CD8+, 85.5% CD4+/CD8+, 3.7% CD4/CD8 and 11.6% αβ-TCRhi thymocytes were found in a swine THY/LIV graft by FCM 17 weeks post-tx compared to 3.5% CD4+/CD8, 3.2% CD4/CD8+, 87.8% CD4+/CD8+, 5.5% CD4/CD8 and 10.0% αβ-TCRhi thymocytes in a normal B10 thymus. Fetal swine liver grafted without a thymus fragment did not grow in control nAb-treated ATX-B10 mice and αβ-TCRhi T cells did not appear in the periphery. Thus, the pig thymus was required for the development of mature mouse T cells.
  • Mouse anti-pig mixed lymphocyte reactions (MLR's) were performed to determine whether or not mouse T cells which matured in pig thymus grafts were tolerant to pig antigens. ATX-THY/LIV B10 mice (H-2b) mounted no anti-B10 or anti-pig responses, but demonstrated normal allo-responses against a fully MHC-mismatched allogeneic stimulator, B10.BR (H-2k) (FIG. 2).
  • In order to determine if host bone marrow-derived cells were participating in negative selection of the developing mouse thymocytes, fetal pig THY/LIV grafts were transplanted into both I-E+ (BALB/c nude) and I-E (ATX B10) recipients. I-E+ mice delete Vβ11 T cells because of presentation in the thymus of an endogenous superantigen in association with I-E, whereas I-E mice do not delete this T cell family. The percentages of Vβ11 T cells were therefore compared between I-E+ and I-E recipients of fetal pig thymus grafts in which murine T cells developed in pig thymi. ATX B10 recipients were treated as described above. BALB/c nude mice were depleted of NK cells and irradiated with 3 Gy WBI prior to transplant. These mice also developed large numbers of mature CD4+ T cells that migrated to the periphery. Complete deletion of Vβ11 T cells was observed in the periphery of BALB/c nude recipients of fetal swine thymus grafts (Table I), indicating that mouse I-E also participated in negative selection of mouse T cells developing in pig thymi. Negative selection is most likely carried out by murine Ia+dendritic cells which were detected predominantly in the cortico-medullary junction of swine thymus grafts by immunoperoxidase staining. In the ATX B10 recipients of swine THY/LIV grafts, reduction in the percentage of Vβ11 T cells was observed compared to normal B10 mice (mean 2.8% of T cells±0.8 S.D., normal B10 5.2%, p<0.005) suggesting that the pig SLA DR class II, which shares significant homology with mouse I-E class II, may participate in negative selection of mouse T cells developing in the pig thymus graft (Table I).
    TABLE I
    N Strain Thy/Liv Graft % Vβ8.1/8.2 % Vβ11
    4 Normal C57BL/10 16.3 ± 2.2 5.2 ± 0.5
    4 Normal BALB/c 20.8 ± 0.3 0.2 ± 0.1
    4 C57BL/10 + 16.7 ± 3.0 2.8 ± 0.8
    5 BALB/c nude + 20.0 ± 3.7 0.4 ± 0.3

    Table I. Clonal deletion in mice transplanted with swine THY/LIV grafts. B10 recipients were treated as described below. Normal percentages of T cells staining with Vβ8.1/8.2 demonstrates normal positive selection in grafted mice of a VβT cell family which is not deleted in I-E+ or I-E mice. BALB/c nude mice were depleted of NK cells using rabbit anti-Asialo-Gm1 serum, and were given 3 Gy WBI, and fetal swine THY/LIV grafts implants under the kidney capsule, followed by injection of 108 FLC i.p. on Day 0. ACK-lysed splenocytes or peripheral white blood cells (red blood cells were removed by hypotonic shock) were collected 13 to 19 weeks post-tx and analyzed by FCM for Vβ11 T cell deletion. Murine FcR's were blocked using rat anti-mouse FcR mAb, 2.4G2, and then cells were stained with either fluoresceinated hamster anti-mouse Vβ8.1/8.2 TCR (Pharmingen) or rat anti-mouse Vβ11 TCR (Pharmingen) (green fluorescence) followed by phycoerythrin-conjugated rat anti-mouse CD4 and CD8 mAbs (Pharmingen) (orange fluorescence) and analyzed by two color FCM as described below. Fluoresceinated murine mAb HOPCI, with no known reactivity to mouse cells, or rat anti-mouse IgG1 (Zymed Laboratories, Inc.) was used as the negative control mAb in the green fluorescence. Phycoerythrin-conjugated mAb Leu4 (Becton-Dickinson), was used as the negative control mAb in the orange fluorescence. Approximately 5,000 gated CD4+ and CD8+ cells were usually collected for analysis of Vβ families. Non-viable cells were excluded using the vital nucleic acid stain, propidium iodide. Percentages of positive cells were determined as described below. Results are presented as the mean±SD of results obtained for individual mice. p value<0.005 for % Vβ11 in normal B10 mice compared to THY/LIV-grafted B10 mice. p value is >0.20 for % Vβ11 in normal BALB/c mnice compared to THY/LIV-grafted BALB/c nude mice.
  • These studies demonstrate that discordant xenogeneic thymic stroma is capable of supporting mouse thymopoiesis and that CD4+/CD8/αβ-TCRhi T cells which are released into the periphery are phenotypically normal, functional and tolerant to donor xeno-antigens, and to host antigens. The lack of CD4/CD8+/αβ-TCRhi repopulation in the periphery may be due to failure of mouse CD8 to interact with pig MHC class I molecules, as has been demonstrated for mouse anti-human responses, thereby preventing positive selection of CD8+ thymocytes by swine thymic epithelium. Since human CD8+ T cells are able to interact with pig MHC class I directly, human CD8+ T cells should mature effectively in swine fetal thymus grafts.
  • Presumably, tolerance to pig antigens is not induced in euthymic mice which receive swine THY/LIV grafts because mouse T cell progenitors mature in the host thymus, which lacks the pig cells necessary to tolerize developing mouse thymocytes. The non-myeloablative conditioning regimen used in this study permits engraftment of rat marrow and induction of donor-specific tolerance in murine recipients. Tolerance is thought to be induced in this model by rat dendritic cells detected at the cortico-medullary junction of the thymus of chimeric animals. In the present study, failure of pig hematopoietic stem cells, present in the FLC suspension administered on Day 0, to migrate to the mouse thymus may be due to failure of homing and differentiation, possibly reflecting species specificity of cytokines and adhesion molecules. In ATX recipients, on the other hand, mouse T cell progenitors home to the pig thymus graft and are tolerized pig antigens. No mouse TE is present in ATX hosts, but mouse dendritic cells are detectable in THY/LIV grafts and probably mediate the observed clonal deletion of cells reactive to host antigen. Although the decreased percentage of Vβ11 T cells in ATX B10 recipients of swine THY/LIV grafts suggests clonal deletion by swine cells, it is possible that there is a defect in the positive selection of this Vβ family on swine thymic stroma. However, the normal percentages of Vβ8.1/8.2 T cells in both ATX B10 and BALB/c nude recipients of swine THY/LIV grafts compared to those in normal B10 and BALB/c mice suggests that no defect in positive selection is present. The observed tolerance could be explained if the swine thymic stroma either clonally deletes or anergizes developing mouse thymocytes reactive to donor xeno-antigens.
  • Survival of swine THY/LIV grafts in euthymic and thymectomized B10 mice depleted of T cells and NK cells, was determined as follows. 6-12 week old euthymic or ATX C57BL/10 (B10) mice received i.p. injections of mAbs GK1.5 (anti-mouse CD4), 2.43 (anti-mouse CD8), 30-H12 (anti-mouse Thy1.2) and PK136 (anti-mouse NK1.1) in depleting doses, as described in Sharabi et al., on days −6 and −1 prior to transplantation. On either day −1 or day 0, 7 Gy localized thymic irradiation and 3 Gy whole body irradiation were administered to recipients, and second trimester (gestational day 36-72) fetal thymic and liver fragments, approximately 1 mm3 in size, were transplanted under the kidney capsule via a midline laparotomy incision. (Thymic irradiation was not found to be necessary for mouse T cells to mature in pig thymus grafts in subsequent experiments, and was therefore eliminated from the conditioning regimen.) After the abdomen was closed in two layers, 108 fetal liver cells (FLC) in suspension were injected i.p. Recipients were treated on a weekly basis post-tx with depleting doses of the same four mAbs for a period of 0-6 weeks. No difference in murine CD4+ T cell reconstitution or tolerance to pig antigens was observed in mice which were treated with no mAb post-tx compared to those which received 6 weeks mAb treatment post-tx. Some groups of control mice were maintained on chronic mAb treatment until the time of sacrifice.
  • As described above, an increase in fetal pig THY/LIV graft size was observed upon exploratory laparotomy performed at 5 and 19 weeks post-tx, despite the presence of mature CD4+/αβTCRhi T cells in the peripheral blood (shown 16 weeks after mAbs were discontinued). Growth of fetal pig THY/LIV graft in the presence of mature mouse T cells in the periphery was studied as follows. Peripheral WBC contained 12.1% CD4+/αβ-TCRhi T cells and 0.5% CD8+/αβ-TRC+ T cells. Control ATX mice which received fetal swine liver grafts without a thymus fragment did not maintain their grafts, and developed less than 5% αβ-TRC+ T cells in the periphery. ATX mice were conditioned as described above. Exploratory laparotomies were performed at 5-6 and 15-19 weeks post-transplant to measure graft size. Mice were tail bled at regular intervals post-tx to obtain peripheral WBC which were prepared by hypotonic shock to remove red blood cells. Cells were stained with a fluoresceinated rat anti-mouse CD4 mAb (Pharmigen) (green fluorescence) versus biotinylated hamster anti-mouse αβTRC mAb (Pharmigen) plus phycoerythrin streptavidin (orange fluorescence) and analyzed by two-color flow cytometry (FCM) using either a FACScan or FACSort flow cytometer (Becton-Dickinson). Murine mAb HOPC1, with no known reactivity to pig or mouse cells, was used as the negative control mAb in both the green and orange fluorescence. Percentages of positive cells were determined by subtracting the percentage of cells staining with the control mAb HOPC1 from the percentage of cells staining with the anti-mouse mAbs. A dot plot analysis of live peripheral white blood cells of a representative animal 16 weeks post-tx (16 weeks after mAbs were discontinued) is shown in FIG. 2. Overall, 57% (27/47) of ATX mice treated with this protocol maintained swine grafts and reconstituted their CD4+ T cell compartment. In recent experiments this result was achieved in 90% (9/10) of mice treated with this regimen.
  • As described above, ATX-THY/LIV B10 (H-2b) mice demonstrated specific unresponsiveness to pig antigens while maintaining normal allo-responsiveness to a fully MHC-mismatched B10.BR (H-2k) stimulator. Specific unresponsiveness of B10 mice transplanted with fetal pig THY/LIV grafts to pig antigens in mixed lymphocyte reaction (MLR) was determined as follows. Control ATX-B10 mice which received a swine liver graft without a thymus fragment (ATX-LIV) mounted no responses to any stimulator, demonstrating the importance of the pig thymus graft in the development of functional mouse T cells. Positive control anti-pig MLR was from a mouse immunized with a swine skin graft, since mice do not mount primary anti-pig responses. Sterile splenocyte suspensions from normal B10 (right diagonal bar), normal B10 grafted with GG (SLA-Ic/SLA-IId) pig skin 12 weeks earlier (GG′-B10 solid bar), normal B10BR (crosshatched bar), and thymectomized B10 mice conditioned with the non-myeloablative regimen described above and transplanted with either a fetal pig (SLA-Id/SLA-IId) THY/LIV graft (ATX-THY/LIV stippled bar), or a fetal pig liver graft only (ATX-LIV left diagonal bar) were ACK-lysed, washed and reconstituted in RPMI medium supplemented with 15% CPSR-2 (controlled processed serum replacement, Sigma), 4% nutrient mixture (L-glutamine, nonessential amino acids, sodium pyruvate and penicillin/streptomycin), 1% HEPES buffer and 10−5M 2-me. Swine PBL were prepared by centrifugation over a Ficoll-Hypaque layer. 4×105 responders were incubated with either 4×105 murine stimulators (3 Gy) or 1×105 swine stimulators (3 Gy) in a total volume of 0.2 ml of media at 37 □C for 4 days in 5% CO2. Cultures were pulsed with 1 μCi 3H on the third day, harvested on the fourth day with a Tomtec automated harvester and counted on a Pharmacia LKB liquid scintillation counter. MLR's for all mice tested (N=3) were set up in duplicate and pulsed on Days 3 and 4 and harvested on Days 4 and 5 with similar results.
  • Thus, if murine T cells are permitted to develop in a mouse thymus, they are not tolerized to pig antigens, and they reject pig thymus/liver grafts. (Thus, if the recipient has significant thymic function thymectomy is indicated.) If mouse T cells are continually depleted by mAb, swine thymopoiesis occurs in the swine thymus/liver graft. If a mouse is thymectomized but not chronically treated with anti-T cell mAb's, mouse thymopoiesis occurs in the pig thymus, and these cells are tolerized to pig antigens.
  • Host T Cells which Mature in a Xenogeneic Thymus are Functional
  • B10.BR (full MHC mismatch to B10) and C3H.SW (minor antigen mismatch only) skin grafts (1 mouse) were rejected by mouse T cells which had matured in a pig thymic graft, thus demonstrating their immunocompetence and ability to recognize minor antigens in a host MHC-restricted fashion. The grafts were full thickness tail skin grafts on the upper thorax with a skin bridge separating them. These results show that swine fetal thymic tissue can be used clinically to induce a state of specific xenograft tolerance while ensuring immunocompetence in thymectomized recipients.
  • Alternative Preparative Regimens
  • As is stated above, the depletion of NK cells, whole body irradiation, and thymic irradiation, can in some cases be dispensed with. As is shown by the experiments summarized below, inactivation or depletion of CD4+ cells, e.g., by the administration of an anti-CD4 antibody, is sufficient to allow growth of xenogeneic thymic tissue and maturation of host T cells in the xenogeneic thymic tissue. (The antibodies needed may differ depending on the species combination. E.g., in the case of a human recipient and a pig donor, because human CD8 will likely interact with pig class I molecules, it may also be necessary to administer anti-CD8 antibodies.)
  • As is shown by the data in Table II below, graft growth and host T cell development (as measured by the presence of peripheral T cells 9 and 10 weeks post THY/LIV transplant) was seen in ATX mice treated with anti-CD4 antibodies and whole body irradiation. B10 mice received 3 Gy whole body irradiation and fetal pig THY/LIV graft and 108 fetal liver cells in experiments essentially similar to those described in the previous section except that anti-CD4, anti-THY1.2, and anti NK cell antibodies were not administered.
    TABLE II
    THY/LIV graft growth and T cell maturation
    does not require extensive antibody treatment.
    GRAFT % CD4+/% CD8+
    Animal # TREATMENT L/R T CELLS (WBC)
    639/40 NO ATX/αCD4/8/ 17.7/4.6 
    THY1.2/NK1.1
    643/44 NO ATX/αCD4/8/ 9.4/2.6
    THY1.2/NK1.1
    601/02 ATX +/ +/++ 11.1/0.8 
    αCD4/8/THY1.2/NK1.1
    603/04 ATX +/ +/+ 1.7/1.2
    αCD4/8/THY1.2/NK1.1
    605/06 ATX +/ −/− 0.6/0.3
    αCD4/8/THY1.2/NK1.1
    607/08 ATX +/ ++/++ 0.9/0.6
    αCD4/8/THY1.2/NK1.1
    609/10 ATX +/ ++/+ 1.8/0.5
    αCD4/8/THY1.2/NK1.1
    611/12 ATX +/ ++/++ 3.4/1.6
    αCD4/8/THY1.2/NK1.1
    615/16 ATX + αCD4/8 +/++ 8.9/0.7
    617/18 +/− 2.1/0.3
    619/20 ++/++ 20.2/1.5 
    621/22 +/− 1.1/0.1
    623/24 ++/++ 6.8/0.3
    625/26 ATX + αCD4 ++/− 7.2/4.0
    627/28 −/+ 1.0/5.1
    629/30 ++/++ 10.1/3.7 
    631/32 ++/+ 3.7/3.5
    633/34 −/− 0.4/3.7
    635/36 ++/++ 28.0/4.2 

    ATX = thymectomy;

    ++ = large, bulky graft with vascularization;

    + = moderate sized graft;

    − = poor graft (thin, poor tissue);

    αCD4/8/THY1.2/NK1.1 indicates the administration of the described antibody.
  • As is shown by the data in Table III below, graft growth was seen in ATX mice treated with monoclonal antibodies but given no irradiation. In these experiments, B10 mice were given anti-CD4, CD8, THY1.2, and NK1.1 monoclonal antibodies.
    TABLE III
    THY/LIV graft growth does not require host irradiation.
    GRAFT
    Animal # TREATMENT L/R
    560/61 ATX + mAb's + 3 Gy WBI ++/++
    562/63 +/++
    564/65 −/−
    566/67 −/+
    574/75 −/−
    576/77 −/−
    578/79 −/−
    582/83 +/−
    552/53 ATX + mAb's (no WBI) +/−
    554/55 ++/−
    556/57 ++/+
    568/69 ++/++
    570/71 ++/++
  • As is shown by the data in Table IV below, host T cell development (as measured by the presence of peripheral T cells 7 weeks post THY/LIV transplant) was seen in ATX mice given no irradiation and treated only with anti-CD4 antibodies.
    TABLE IV
    Rapid T cell recovery in THY/LIV graft recipients with
    thymectomy and anti CD4 antibodies alone.
    Mean % (SD)
    peripheral blood
    Number of T cells at 7 weeks
    animals in post-transplant
    group TREATMENT CD4+ CD8+
    3 NO ATX 12.9 (1.7) 2.7 (0.1)
    αCD4/8/THY1.2/NK1.1 + 3
    Gy WBI
    8 ATX  3.4 (2.4) 0.7 (0.5)
    αCD4/8/THY1.2/NK1.1 + 3
    Gy WBI
    7 ATX 19.5 (3.4) 7.1 (1.8)
    αCD4/

    Xenogeneic Thymic Tissue and Stem Cell Transplantation
  • The following procedure introduces donor thymic tissue and donor stem cells into the recipient and thus can be used to restore or induce immune function or to lengthen the time an implanted organ (a xenograft) survives in a xenogeneic host prior to rejection.
  • In the case of an organ graft, the organ can be any organ, e.g., a liver, e.g., a kidney, e.g., a heart. The two main strategies are elimination of natural antibodies by organ perfusion, and transplantation of tolerance-inducing bone marrow.
  • Preparation of the recipient includes any or all of the following steps. Preferably they are carried out in the following sequence.
  • Elimination of NK and T cells. First, a preparation of horse anti-human thymocyte globulin (ATG) is intravenously injected into the recipient. The antibody preparation eliminates mature T cells and natural killer cells. If not eliminated, mature T cells would promote rejection of both the bone marrow transplant and, after sensitization, the xenograft itself of equal importance, the ATG preparation also eliminates natural killer (NK) cells. NK cells probably have no effect on the implanted organ, but would act immediately to reject the newly introduced bone marrow. Anti-human ATG obtained from any mama host can also be used, e.g., ATG produced in pigs, although thus far preparations of pig ATG have been of lower titer than horse-derived ATG. ATG is superior to anti-NK monoclonal antibodies, as the latter are generally not lytic to all host NK cells, while the polyclonal mixture in ATG is capable of lysing all host NK cells. Anti-NK monoclonal antibodies can, however, be used.
  • Thymic tissue transplant. In cases where the procedure is to restore or induce immunocompetence donor thymic tissue (preferably fetal or neonatal thymic tissue) is implanted in the recipient so that donor T cells (and recipient T cells if the are present and functional) can mature. Fetal or neonatal liver or spleen tissue can be implanted with the thymic tissue.
  • The presence of donor antigen in the thymus during the time when host T cells are regenerating post-transplant is critical for tolerizing host T cells. If donor hematopoietic stem cells are not able to become established in the host thymus and induce tolerance before host T cells regenerate repeated doses of anti-recipient T cell antibodies may be necessary throughout the non-myeloablative regimen. Continuous depletion of host T cells may be required for several weeks. Alternatively, e.g., if this approach is not successful, and tolerance (as measured by donor skin graft acceptance, specific cellular hyporesponsiveness in vitro, and humoral tolerance) is not induced in these animals, the approach can be modified to include host thymectomy. In thymectomized recipients, host T cells do not have an opportunity to differentiate in a host thymus, but must differentiate in the donor thymus. Immunocompetence can be measured by the ability to reject a non-donor type allogeneic donor skin graft, and to survive in a pathogen-containing environment.
  • It may also be necessary or desirable to splenectomize the recipient in order to avoid anemia.
  • Creation of hematopoietic space. The recipient is administered low dose radiation in order to make room for newly injected bone marrow cells. A sublethal dose e.g., a dose about equal to 100, or more than 100 and less than about 400, rads, whole body radiation, plus 700 rads of local thymic radiation, has been found effective for this purpose.
  • Natural antibody elimination. Natural antibodies are adsorbed from the recipient's blood by hemoperfusion of a liver of the donor species. Pre-formed natural antibodies-(nAb) are the primary agents of graft rejection. Natural antibodies bind to xenogeneic endothelial cells and are primarily of the IgM class. These antibodies are independent of any known previous exposure to antigens of the xenogeneic donor. B cells that produce these natural antibodies tend to be T cell-independent, and are normally tolerized to self antigen by exposure to these antigens during development. The mechanism by which newly developing B cells are tolerized is unknown. The liver is a more effective adsorber of natural antibodies than the kidney.
  • Implantation of donor stromal tissue. The next step in the non-myeloablative procedure is to implant donor stromal tissue, preferably obtained from fetal liver, thymus, and/or fetal spleen, into the recipient, preferably in the kidney capsule. Stem cell engraftment and hematopoiesis across disparate species barriers is enhanced by providing a hematopoietic stromal environment from the donor species. The stromal matrix supplies species-specific factors that are required for interactions between hematopoietic cells and their stromal environment, such as hematopoietic growth factors, adhesion molecules, and their ligands.
  • Each organ includes an organ specific stromal matrix that can support differentiation of the respective undifferentiated stem cells implanted into the host. Although adult thymus may be used, fetal tissue obtained sufficiently early in gestation is preferred because it is free from mature T lymphocytes which can cause GVHD. Fetal tissues also tend to survive better than adult tissues when transplanted. As an added precaution against GVHD, thymic stromal tissue can be irradiated prior to transplantation, e.g., irradiated at 1000 rads. As an alternative or an adjunct to implantation, fetal liver cells can be administered in fluid suspension.
  • Finally, bone marrow cells (BMC), or another source of hematopoietic stem cells, e.g., a fetal liver suspension, or cord blood stem cells, of the donor are injected into the recipient. Donor stem cells home to appropriate sites of the recipient and grow contiguously with remaining host cells and proliferate, forming a chimeric lymphohematopoietic population. By this process, newly forming B cells (and the antibodies they produce) are exposed to donor antigens, so that the transplant will be recognized as self. Tolerance to the donor is also observed at the T cell level in animals in which hematopoietic stem cell, e.g., BMC, engraftment has been achieved. When an organ graft is placed in such a recipient several months after bone marrow chimerism has been induced, natural antibody against the donor will have disappeared, and the graft should be accepted by both the humoral and the cellular arms of the immune system. This approach has the added advantage of permitting organ transplantation to be performed sufficiently long following transplant of hematopoietic cells, e.g., BMT, e.g., a fetal liver suspension, that normal health and immunocompetence will have been restored at the time of organ transplantation. The use of xenogeneic donors allows the possibility of using bone marrow cells and organs from the same animal, or from genetically matched animals. As liver is the major site of hematopoiesis in the fetus, fetal liver can also serve as an alternative to bone marrow as a source of hematopoietic stem cells.
  • While any of these procedures may aid the survival of an implanted organ, best results are achieved when all steps are used in combination. Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • In the case of xenogeneic grafts, the donor of the implant and the individual that supplies either the tolerance-inducing hematopoietic cells or the liver to be perfused should be the same individual or should be as closely related as possible. For example, it is preferable to derive implant tissue from a colony of donors that is highly or completely inbred.
  • Detailed Protocol
  • In the following protocol for preparing a cynomolgus monkey for receipt of a kidney from a miniature swine donor, zero time is defined as the moment that the arterial and venous cannulas of the recipient are connected to the liver to be perfused.
  • On day −1 a commercial preparation (Upjohn) of horse anti-human anti-thymocyte globulin (ATG) is injected into the recipient. ATG eliminates mature T cells and natural killer cells that would otherwise cause rejection of the bone marrow cells used to induce tolerance. The recipient is anesthetized, an IV catheter is inserted into the recipient, and 6 ml of heparinized whole blood are removed before injection. The ATG preparation is then injected (50 mg/kg) intravenously. Six ml samples of heparinized whole blood are drawn for testing at time points of 30 min., 24 hrs and 48 hrs. Blood samples are analyzed for the effect of antibody treatment on natural killer cell activity (testing on K562 targets) and by FACS analysis for lymphocyte subpopulations, including CD4, CD8, CD3, CD11b, and CD16. Preliminary data from both assays indicate that both groups of cells are eliminated by the administration of ATG. If mature T cells and NK cells are not eliminated, ATG can be re-administered at later times in the procedure, both before and after organ transplantation.
  • Sublethal irradiation is administered to the recipient between days −1 and −8. Irradiation is necessary to eliminate enough of the recipient's endogenous BMC to stimulate hematopoiesis of the newly introduced foreign BMC. Sublethal total body irradiation is sufficient to permit engraftment with minimal toxic effects to the recipient. Whole body radiation (150 Rads) was administered to cynomolgus monkey recipients from a bilateral (TRBC) cobalt teletherapy unit at 10 Rads/min. Local irradiation of the thymus (700 Rads) was also employed in order to facilitate engraftment.
  • Natural antibodies are a primary cause of organ rejection. To remove natural antibodies from the recipient's circulation prior to transplantation, on day 0 an operative adsorption of natural antibodies (nAB) is performed, using a miniature swine liver, as follows. At −90 minutes the swine donor is anesthetized, and the liver prepared for removal by standard operative procedures. At −60 minutes the recipient monkey is anesthetized. A peripheral IV catheter is inserted, and a 6 ml sample of whole blood is drawn. Through mid-line incision, the abdominal aorta and the vena cava are isolated. Silastic cannulas containing side ports for blood sampling are inserted into the blood vessels.
  • At −30 minutes the liver is perfused in situ until it turns pale, and then removed from the swine donor and placed into cold Ringers Lactate. The liver is kept cold until just prior to reperfusion in the monkey. A liver biopsy is taken. At −10 minutes the liver is perfused with warm albumin solution until the liver is warm (37 degrees).
  • At 0 time the arterial and venous cannulas of the recipient are connected to the portal vein and vena cava of the donor liver and perfusion is begun. Liver biopsies are taken at 30 minutes and 60 minutes, respectively. Samples of recipient blood are also drawn for serum at 30 minutes and 60 minutes respectively. At 60 minutes the liver is disconnected from the cannulas and the recipient's large blood vessels are repaired. The liver, having served its function of adsorbing harmful natural antibodies from the recipient monkey, is discarded. Additional blood samples for serum are drawn from the recipient at 2, 24, and 48 hours. When this procedure was performed on two sequential perfusions of swine livers, the second liver showed no evidence of mild ischemic changes during perfusion. At the end of a 30 minute perfusion the second liver looked grossly normal and appeared to be functioning, as evidenced by a darkening of the venous outflow blood compared to the arterial inflow blood in the two adjacent cannulas. Tissue sections from the livers were normal, but immunofluorescent stains showed IgM on endothelial cells. Serum samples showed a decrease in natural antibodies.
  • To promote long-term survival of the implanted organ through T-cell and B-cell mediated tolerance, donor bone marrow cells are administered to the recipient to form chimeric bone marrow. The presence of donor antigens in the bone marrow allows newly developing B cells, and newly sensitized T cells, to recognize antigens of the donor as self, and thereby induces tolerance for the implanted organ from the donor. To stabilize the donor BMC, donor stromal tissue, in the form of tissue slices of fetal liver, thymus, and/or fetal spleen are transplanted under the kidney capsule of the recipient. Stromal tissue is preferably implanted simultaneously with, or prior to, administration of hematopoietic stem cells, e.g., BMC, or a fetal liver cell suspension.
  • To follow chimerism, two color flow cytometry can be used. This assay uses monoclonal antibodies to distinguish between donor class I major histocompatibility antigens and leukocyte common antigens versus recipient class I major histocompatibility antigens.
  • BMC can in turn be injected either simultaneously with, or preceding, organ transplant. Bone marrow is harvested and injected intravenously (7.5×108/kg) as previously described (Pennington et al., 1988, Transplantation 45:21-26). Should natural antibodies be found to recur before tolerance is induced, and should these antibodies cause damage to the graft, the protocol can be modified to permit sufficient time following BMT for humoral tolerance to be established prior to organ grafting.
  • The approaches described above are designed to synergistically prevent the problem of transplant rejection. When a kidney is implanted into a cynomolgus monkey following liver adsorption of natural antibodies, without use of bone marrow transplantation to induce tolerance, renal functions continued for 1-2 days before rejection of the kidney. When four steps of the procedure were performed (adsorption of natural antibodies by liver perfusion, administration of ATG, sublethal irradiation and bone marrow infusion, followed by implant of a porcine kidney into a primate recipient), the kidney survived 7 days before rejection. Despite rejection of the transplanted organ, the recipient remained healthy.
  • When swine fetal liver and thymic stromal tissue were implanted under the kidney capsule of two sublethally irradiated SCID mice, 25-50% of peripheral blood leukocytes were of donor lineage two weeks post-transplantation. A significant degree of chimerism was not detected in a third animal receiving fetal liver without thymus. These procedures did not employ any chemical immunosuppressants.
  • Other Embodiments
  • Other embodiments are within the following claims.
  • For example, implanted grafts may consist of organs such as liver, kidney, heart; body parts such as bone or skeletal matrix; tissue such as skin, intestines, endocrine glands; or progenitor stem cells of various types.
  • The methods of the invention may be employed with other mammalian recipients (e.g., rhesus monkeys, humans) and may use other mammalian donors (e.g., primates, swine, sheep, dogs).
  • The methods of the invention may be employed in combination, as described, or in part.
  • The method of introducing bone marrow cells may be altered, particularly by (1) increasing the time interval between injecting hematopoietic stem cells and implanting the graft; (2) increasing or decreasing the amount of hematopoietic stem cells injected; (3) varying the number of hematopoietic stem cell injections; (4) varying the method of delivery of hematopoietic stem cells; (5) varying the tissue source of hematopoietic stem cells, e.g., a fetal liver cell suspension may be used; or (6) varying the donor source of hematopoietic stem cells. Although hematopoietic stem cells derived from the graft donor are preferable, hematopoietic stem cells may be obtained from other individuals or species, or from genetically-engineered completely or partially inbred donor strains, or from in vitro cell culture.
  • Methods of preparing the recipient for transplant of hematopoietic stem cells may be varied. For instance, the recipient may undergo a splenectomy or a thymectomy. The latter would preferably by administered prior to the non-myeloablative regimen, e.g., at day −14.
  • Hemoperfusion of natural antibodies may: (1) make use of other vascular organs, e.g., liver, kidney, intestines; (2) make use of multiple sequential organs; (3) make use of varying the length of time each organ is perfused; (4) make use of varying the donor of the perfused organ. Irradiation of the recipient may make use of: (1) of varying the adsorbed dose of whole body radiation below the sublethal range; (2) of targeting different body parts (e.g., thymus, spleen); (3) varying the rate of irradiation (e.g., 10 rads/min, 15 rads/min); or (4) varying the time interval between irradiation and transplant of hematopoietic stem cells; any time interval between 1 and 14 days can be used, and certain advantages may flow from use of a time interval of 4-7 days. Antibodies introduced prior to hematopoietic cell transplant may be varied by: (1) using monoclonal antibodies to T cell subsets or NK cells (e.g., anti-NKH1A, as described by U.S. Pat. No. 4,772,552 to Hercend, et al., hereby incorporated by reference); (2) preparing anti-human ATG in other mammalian hosts (e.g., monkey, pig, rabbit, dog); or (3) using anti-monkey ATG prepared in any of the above mentioned hosts.
  • As an alternative or adjunct to hemoperfusion, host antibodies can be depleted by administration of an excess of hematopoietic cells.
  • Stromal tissue introduced prior to hematopoietic cell transplant, e.g., BMT, may be varied by: (1) administering the fetal liver and thymus tissue as a fluid cell suspension; (2) administering fetal liver or thymus stromal tissue but not both; (3) placing a stromal implant into other encapsulated, well-vascularized sites; or (4) using adult thymus or fetal spleen as a source of stromal tissue.
  • As is discussed herein, it is often desirable to expose a graft recipient to irradiation in order to promote the development of mixed chimerism. The inventor has discovered that it is possible to induce mixed chimerism with less radiation toxicity by fractionating the radiation dose, i.e., by delivering the radiation in two or more exposures or sessions. Accordingly, in any method of the invention calling for the irradiation of a recipient, e.g., a primate, e.g., a human, recipient, of a xenograft or allograft, the radiation can either be delivered in a single exposure, or more preferably, can be fractionated into two or more exposures or sessions. The sum of the fractionated dosages is preferably equal, e.g., in rads or Gy, to the radiation dosage which can result in mixed chimerism when given in a single exposure. The fractions are preferably approximately equal in dosage. For example, a single dose of 700 rads can be replaced with, e.g., two fractions of 350 rads, or seven fractions of 100 rads. Hyperfractionation of the radiation dose can also be used in methods of the invention. The fractions can be delivered on the same day, or can be separated by intervals of one, two, three, four, five, or more days. Whole body irradiation, thymic irradiation, or both, can be fractionated.
  • Methods of the invention can include recipient splenectomy.
  • As is discussed herein, hemoperfusion, e.g., hemoperfusion with a donor organ, can be used to deplete the host of natural antibodies. Other methods for depleting or otherwise inactivating natural antibodies can be used with any of the methods described herein. For example, drugs which deplete or inactivate natural antibodies, e.g., deoxyspergualin (DSG) (Bristol), or anti-IgM antibodies, can be administered to the recipient of an allograft or a xenograft. One or more of, DSG (or similar drugs), anti-IgM antibodies, and hemoperfusion, can be used to deplete or otherwise inactivate recipient natural antibodies in methods of the invention. DSG at a concentration of 6 mg/kg/day, i.v., has been found useful in suppressing natural antibody function in pig to cynomolgus kidney transplants.
  • Some of the methods described herein use irradiation to create hematopoietic space, and thereby prepare a recipient for the administration of allogeneic, xenogeneic, syngeneic, or genetically engineered autologous, stem cells. In any of the methods described herein, particularly primate or clinical methods, it is preferable to create hematopoietic space for the administration of such cells by non-lethal means, e.g., by administering sub-lethal doses of irradiation, bone marrow depleting drugs, or antibodies. The use of sublethal levels of bone marrow depletion allows the generation of mixed chimerism in the recipient. Mixed chimerism is generally preferable to total or lethal ablation of the recipient bone marrow followed by complete reconstitution of the recipient with administered stem cells.
  • Xenogeneic thymic tissue is easier to obtain and in general is less likely to harbor human pathogens. Thus, xenogeneic thymic tissue is preferred in methods for restoring or inducing immunocompetence. Allogeneic thymic tissue can however be used in these methods.
  • Some of the methods described herein include the administration of thymic irradiation, e.g., to inactivate host thymic-T cells or to otherwise diminish the host's thymic-T cell mediated responses to donor antigens. It has been discovered that the thymic irradiation called for in allogeneic or xenogeneic methods of the invention can be supplemented with, or replaced by, other treatments which diminish (e.g., by depleting thymic-T cells and/or down modulating one or more of the T cell receptor (TCR), CD4 co-receptor, or CD8 co-receptor) the host's thymus function, e.g., the host's thymic-T cell mediated response. For example, thymic irradiation can be supplemented with, or replaced by, anti-T cell antibodies (e.g., anti-CD4 and/or anti-CD8 monoclonal antibodies) administered a sufficient number of times, in sufficient dosage, for a sufficient period of time, to diminish the host's thymic-T cell mediated response.
  • For best results, anti-T cell antibodies should be administered repeatedly. E.g., anti-T cell antibodies can be administered one, two, three, or more times prior to donor thymus or bone marrow transplantation. Typically, a pre-thymus or bone marrow transplantation dose of antibodies will be given to the patient about 5 days prior to thymus or bone marrow transplantation. Additional, earlier doses 6, 7, or 8 days prior to thymus or bone marrow transplantation can also be given. It may be desirable to administer a first treatment then to repeat pre-thymus or bone marrow administrations every 1-5 days until the patient shows excess antibodies in the serum and about 99% depletion of peripheral T cells and then to perform the bone marrow transplantation. Anti-T cell antibodies can also be administered one, two, three, or more times after thymus or donor bone marrow transplantation. Typically, a post-thymus or bone marrow transplant treatment will be given about 2-14 days after bone marrow transplantation. The post thymus or bone marrow administration can be repeated as many times as needed. If more than one administration is given the administrations can be spaced about 1 week apart. Additional doses can be given if the patient appears to undergo early or unwanted T cell recovery. Preferably, anti-T cell antibodies are administered at least once (and preferably two, three, or more times) prior to donor thymus or bone marrow transplantation and at least once (and preferably two, three, or more times) after donor thymus or bone marrow transplantation.
  • It has also been discovered that much or all of the preparative regimen, if called for, can be delivered or administered to a recipient, e.g., an allograft or xenograft recipient, within a few days, preferably within 72, 48, or 24 hours, of transplantation of tolerizing stem cells and/or the graft. This is particularly useful in the case of humans receiving grafts from cadavers. Accordingly, in any of the methods of the invention calling for the administration of treatments prior to the transplant of stem cells and/or a graft, e.g., treatments to inactivate or deplete host antibodies, treatments to inactivate host T cells or NK cells, or irradiation, the treatment(s) can be administered, within a few days, preferably within 72, 48, or 24 hours, of transplantation of the stem cells and/or the graft. In particular, primate, e.g., human, recipients of allografts can be given any or all of treatments to inactivate or deplete host antibodies, treatments to inactivate host T cells or NK cells, or irradiation, within a few days, preferably within 72, 48, or 24 hours, of transplantation of stem cells and/or the graft. For example, treatment to deplete recipient T cells and/or NK cells, e.g., administration of ATG, can be given on day −2, −1, and 0, and WBI, thymic irradiation, and stem cell, e.g., bone marrow stem cells, administered on day 0. (The graft, e.g., a renal allograft, is transplanted on day 0).
  • As described in PCT/US94/01616, hereby incorporated by reference, it has been discovered that there is a permissible time period (“window”) for hematopoietic stem cell engraftment following the creation of space (e.g., by whole body irradiation) for the donor hematopoietic stem cells in a recipient. It has further been discovered that space created for hematopoietic stem cell engraftment can be monitored over time by monitoring peripheral white blood cell levels in a recipient. The myelosuppressive treatment sufficient to create hematopoietic space generally results in a reduction in white blood cell (WBC) levels (as revealed, e.g., by WBC counts) and the WBC reduction serves as a marker for the presence of hematopoietic space. The marker is a conservative one since WBC counts may recover at a time when space is still present in an animal.
  • Accordingly, in any method which involves hematopoietic stem cell transplantation, and thus also requires the creation of hematopoietic space in a recipient, transplantation can be performed during the permissible window for engraftment following creation of space for the hematopoietic stem cells. Likewise, in any method in which space is created for exogenously administered hematopoietic stem cells, white blood cell levels can be followed to monitor space for the donor hematopoietic stem cells (i.e., to assess the permissible window for engraftment). Examples of procedures involving hematopoietic stem cell transplantation include: 1) conditioning of a recipient for an allo- or xenograft in which hematopoietic stem cell transplantation is performed in conjunction with transplantation of another allo- or xenograft; 2) treatment of various hematopoietic disorders, including leukemias, lymphomas and other hematopoietic malignancies and genetic hematopoietic disorders (e.g., adenosine deaminase deficiency, bare lymphocyte syndrome and other congenital immunodeficiency diseases) in which hematopoietic stem cell transplantation is performed therapeutically; and 3) transplantation of genetically modified hematopoietic stem cells (e.g., genetically modified autologous hematopoietic stem cells) to deliver a gene product to a recipient (e.g., as gene therapy).
  • Accordingly, methods of the invention include a method of determining if a myelosuppressive or hematopoietic-space inducing treatment is sufficient to create hematopoietic space. The method includes administering a myelosuppressive treatment to a recipient, and determining the level of white blood cells in the recipient, e.g., by determining the WBC count of the recipient, a depression in the level of white blood cells being indicative of the presence or induction of hematopoietic space.
  • As is discussed in PCT/US94/01616, hereby incorporated by reference, and elsewhere herein, the engraftment of exogenously supplied hematopoietic stem cells can be promoted by treating the recipient of the cells so as to induce hematopoietic space in the recipient. Hematopoietic space is commonly induced by radiation, but other procedures can replace or reduce the need for WBI. For example, space can be created by treating the recipient with a monoclonal antibody against MHC class I antigens expressed by the recipient (see e.g., Voralia, M. et al. (1987) Transplantation 44:487) or space can be created by treating the recipient with myelosuppressive drugs (see e.g., Lapidot, T. et al. (1990) Proc. Natl. Acad. Sci. USA 87:4595).
  • It has also been found that the direct introduction of donor antigen, e.g., donor hematopoietic stem cells, into the thymus of a recipient, can modify the immune response of the recipient. Thus, embodiments of the invention include methods of promoting the acceptance a graft (e.g., by prolonging the acceptance the graft) by a recipient, by introducing into the recipient, donor antigen. The graft can be an allograft, e.g., a graft from a primate e.g., a human, which is introduced into a primate of the same species. The graft can be a concordant or discordant xenograft. E.g., the graft can be a miniature swine graft introduced into a second species, e.g., a primate, e.g., a human.
  • Induction of Tolerance
  • The invention provides several methods of inducing tolerance to foreign antigens, e.g., to antigens on allogeneic or xenogeneic tissue or organ grafts. These methods can be used individually or in combination. For example, it has been discovered that short-term administration of a help reducing agent, e.g., a short high dose course of cyclosporine A (CsA) significantly prolongs graft acceptance. (Preferably the help reduction regimen of the invention substantially eliminates the initial burst of IL-2 which accompanies the first recognition of an antigen but does not eliminate mature T cells. This is distinct from anti-T cell antibody treatments which eliminate mature T cells.)
  • It has also been discovered that a short course of an immunosuppressant, e.g., cyclosporine, can be used to inactivate T cells which would otherwise promote the rejection of a graft.
  • Experiments which show the effect of cyclosporine-induced tolerance on renal allografts in primates are described in section I below. The help suppression methods of the invention can be combined with other methods for prolonging graft acceptance. Section II below discusses implantation of retrovirally transformed bone marrow cells to induce tolerance to MHC disparity. This method can be combined with help suppression methods of the invention, e.g., a short course of high dose of cyclosporine to induce tolerance to class I and other minor disparities.
  • Section III below discusses implantation of bone marrow cells to induce tolerance to MHC disparity. This method can be combined with a short course of high dose cyclosporine administration to induce tolerance to class I and other minor disparities. A short course of cyclosporine, to eliminate T cells, can also be combined with bone marrow transplant.
  • I. A Short Post-Transplant Course of High Dose of Cyclosporine (Administered in the Absence of Agents which Stimulate Cytokine Release) Prolongs Acceptance of Partially Matched Allografts in Primates.
  • Renal transplants were performed between cynomolgus monkeys with or without a brief course of T cell-help-eliminating immunosuppression in the form of a short course of high dose cyclosporine. This regimen significantly prolonged acceptance of the grafts. Monkeys which received a post-transplant course of high dose cyclosporine (without Prednisone) were tolerant to kidney grafts from MLC (mixed lymphocyte culture assay) nonreactive class I and minor antigen mismatched donors for over 65 days, see below. Monkeys which did not receive post-transplant cyclosporine rejected grafts of the same disparity in less than 20 days. This work is discussed in more detail below.
  • Animals. Cynomolgus monkeys were purchased from Charles River Research Laboratories. The animals were quarantined, tested for a full battery of pathogens, and then housed in environmentally controlled rooms in strict conformance to the N.I.H. Guide for Care and use of Laboratory Animals, in an AAALAC accredited facility.
  • Typing. Animals were typed by a standard complement mediated cytotoxicity assay for class I antigens and by MLC nonreactivity for class II matching.
  • Immunosuppression. An intravenous preparation of CsA (Sandimmune, i.v.) was obtained from Sandoz Pharmaceuticals Corporation, Hanover, N.J. Monkeys received 12 doses of about 10 mg/kg with the first dose given 4 hours prior to graft revascularization. The CsA was diluted in 250 ml of normal saline and infused intravenously over 1 hour. The CsA was administered without other immunosuppressants. The duration of therapy was 12 days. The suitable dosage in pigs is about 15 mg/kg delivered intramuscularly. The dosage in either animal should be such that a blood level of about 500-1,000 ng/ml is maintained.
  • Renal Function, Rejection, and Pathology. Renal function was followed by creatinine and BUN levels in serum. Pathology was by biopsy. Biopsies were performed at day 7, then weekly for 2 months, then monthly.
  • Results. Control recipients (no cyclosporine) rejected transplanted kidneys in less than 15 days. The results with 6 cyclosporine treated animals were as follows: animal 1, died on day 65 (i.e., 65 days after transplant), the transplanted was rejected, (it should be noted that the blood cyclosporine level of this animal was below 500 ng/ml during the first 7 days after transplant); animal 2, died on day 65 from bleeding from a biopsy, the transplanted kidney was normal at the time of death; animal 3 died on day 82, some rejection was apparent on day 55; animal 4 was still normal at day 70 (at which time the experiment was still in progress); animal 5 was still normal at day 40 (at which time the experiment was still in progress); and animal 6 was still normal at day 26 (at which time the experiment was still in progress).
  • II. A Short Course of High Dose Cyclosporine (Administered in the Absence of Treatments which Stimulate the Release of Cytokines, e.g. the Absence of Prednisone) to Induce Tolerance to Class I and Other Minor Disparities Combined with Implantation of Retrovirally Transformed Bone Marrow Cells to Induce Tolerance to Class II Disparity.
  • Retroviral Transformation
  • Retroviral transformation allows the reconstitution of a graft recipient's bone marrow with transgenic bone marrow cells, preferably autologous bone marrow cells, expressing allogeneic or xenogeneic MHC genes. Expression of the transgenic MHC genes confers tolerance to grafts which exhibit the products of these or closely related MHC genes. Thus, these methods provide for the induction of specific transplantation tolerance by somatic transfer of MHC genes. Retroviral introduction of MHC genes can be used alone or combined with the T cell help reducing methods described herein. This approach is discussed in detail below.
  • MHC Genes: MHC genes for a variety of species are well studied. For example the HLA genes in man, see, e.g., Hansen et al., 1989, The Major Histocompatibility Complex, In Fundamental Immunology 2d ed., W. E. Paul, ed., Raven Press Ltd., New York, hereby incorporated by reference, and the SLA genes in swine, see e.g., Sachs et al., 1988, Immunogenetics 28:22-29, hereby incorporated by reference, have been cloned and characterized.
  • A gene encoding a MHC antigen can be used in methods of the invention to confer tolerance to a graft which displays that or a closely related MHC antigen. Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a human.
  • The individual which supplies the MHC genes should be as genetically similar as possible, particularly in terms of the MHC genes, to the individual which supplies the graft. For example, in allogeneic grafts wherein the implant donor is a human and the implant recipient is a human it is preferable to use MHC genes from the donor. In this embodiment, MHC probes, e.g., a probe from a third person or a synthetic consensus probe, can be used to isolate DNA encoding the MHC gene or genes of the implant donor individual. This allows the closest match between the genes used to confer tolerance and the genes which express MHC antigens on the graft.
  • In xenogeneic grafts, the implant donor individual and the individual which supplies the tolerance conferring DNA should be the same individual or should be as closely related as possible. For example, it is preferable to derive implant tissue from a colony of donors which is highly inbred and, more preferably, which is homozygous for the MHC genes. This allows the single cloned MHC sequence to be used for many graft recipients.
  • Transformation of bone marrow cells: MHC genes can be introduced into bone marrow cells by any methods which allows expression of these genes at a level and for a period sufficient to confer tolerance. These methods include e.g., transfection, electroporation, particle gun bombardment, and transduction by viral vectors, e.g., by retroviruses.
  • Recombinant retroviruses are a preferred delivery system. They have been developed extensively over the past few years as vehicles for gene transfer, see e.g., Eglitis et al., 1988, Adv. Exp. Med. Biol. 241:19. The most straightforward retroviral vector construct is one in which the structural genes of the virus are replaced by a single gene which is then transcribed under the control of regulatory elements contained in the viral long terminal repeat (LTR). A variety of single-gene-vector backbones have been used, including the Moloney murine leukemia virus (MoMuLV). Retroviral vectors which permit multiple insertions of different genes such as a gene for a selectable marker and a second gene of interest, under the control of an internal promoter can be derived from this type of backbone, see e.g., Gilboa, 1988, Adv. Exp. Med. Biol. 241:29.
  • The elements of the construction of vectors for the expression of a protein product, e.g., the choice of promoters is known to those skilled in the art. The most efficient expression from retroviral vectors is observed when “strong” promoters are used to control transcription, such as the SV 40 promoter or LTR promoters, reviewed in Chang et al., 1989, Int. J. Cell Cloning 7:264. These promoters are constitutive and do not generally permit tissue-specific expression. However, in the case of class I genes, which are normally expressed in all tissues, ubiquitous expression is acceptable for functional purposes. Housekeeping gene promoters, e.g., the thymidine kinase promoter, are appropriate promoters for the expression of class II genes.
  • The use of efficient packaging cell lines can increase both the efficiency and the spectrum of infectivity of the produced recombinant virions, see Miller, 1990, Human Gene Therapy 1:5. Murine retroviral vectors have been useful for transferring genes efficiently into murine embryonic, see e.g., Wagner et al., 1985, EMBO J. 4:663; Griedley et al., 1987 Trends Genet. 3:162, and hematopoietic stem cells, see e.g., Lemischka et al., 1986, Cell 45:917-927; Dick et al., 1986, Trends in Genetics 2:165-170.
  • A recent improvement in retroviral technology which permits attainment of much higher viral titers than were previously possible involves amplification by consecutive transfer between ecotropic and amphotropic packaging cell lines, the so-called “ping-pong” method, see e.g., Kozak et al., 1990, J. Virol. 64:3500-3508; Bodine et al., 1989, Prog. Clin. Biol. Res. 319: 589-600.
  • Transduction efficiencies can be enhanced by pre-selection of infected marrow prior to introduction into recipients, enriching for those bone marrow cells expressing high levels of the selectable gene, see e.g., Dick et al., 1985, Cell 42:71-79; Keller et al., 1985, Nature 318:149-154. In addition, recent techniques for increasing viral titers permit the use of virus-containing supernatants rather than direct incubation with virus-producing cell lines to attain efficient transduction, see e.g., Bodine et al., 1989, Prog. Clin. Biol. Res. 319:589-600. Because replication of cellular DNA is required for integration of retroviral vectors into the host genome, it may be desirable to increase the frequency at which target stem cells which are actively cycling e.g., by inducing target cells to divide by treatment in vitro with growth factors, see e.g., Lemischka et al., 1986, Cell 45:917-927, a combination of IL-3 and IL-6 apparently being the most efficacious, see e.g., Bodine et al., 1989, Proc. Natl. Acad. Sci. 86:8897-8901, or to expose the recipient to 5-fluorouracil, see e.g., Mori et al., 1984, Jpn. J. Clin. Oncol. 14 Suppl. 1:457-463, prior to marrow harvest, see e.g., Lemischka et al., 1986, Cell 45:917-927; Chang et al., 1989, Int. J. Cell Cloning 7:264-280.
  • N2A or other Moloney-based vectors are preferred retroviral vectors for transducing human bone marrow cells.
  • Preparative Regimen For The Introduction of Transformed Bone Marrow Cells To prepare for bone marrow cells the recipient must undergo an ablation of the immune response which might otherwise resist engraftment.
  • The preparative regimens necessary to permit engraftment of modified autologous hematopoietic stem cells may be much less toxic than those needed for allogeneic bone marrow transplantation—preferably requiring only depletion of mature T cells with monoclonal antibodies, as has been recently demonstrated in a mouse model, see Sharabi et al., 1989, J. Exp. Med. 169:493-502. It is possible that transient expression may be sufficient to induce tolerance, which may then be maintained by the transplant even if expression on hematopoietic cells is lost, as has been observed for heart transplants in a mixed xenogeneic bone marrow transplant model, Ildstad et al., 1985, Transplant. Proc. 17: 535-538.
  • Graft and help reduction: The help reducing methods described above can be administered in conjunction with transplantation of the graft, as is described above.
  • Sustained Expression of a Swine Class II Gene in Murine Bone Marrow Hematopoietic Cells by Retroviral-Mediated Gene Transfer
  • Overview: The efficacy of a gene transfer approach to the induction of transplantation tolerance in miniature swine model was shown by using double-copy retroviral vectors engineered to express a drug-resistance marker (neomycin) and a swine class II DRB cDNA. Infectious particles containing these vectors were produced at a titer of >1×106 G418-resistant colony-forming units/ml using both ecotropic and amphotropic packaging cell lines. Flow cytometric analysis of DRA-transfected murine fibroblasts subsequently transduced with virus-containing supernatants demonstrated that the transferred sequences were sufficient to produce DR surface expression. Cocultivation of murine bone marrow with high-titer producer lines leads to the transduction of 40% of granulocyte/macrophage colony-forming units (CFU-GM) as determined by the frequency of colony formation under G418 selection. After nearly 5 weeks in long-term bone marrow culture, virus-exposed marrow still contained G418-resistant CFU-GM at a frequency of 25%. In addition, virtually all of the transduced and selected colonies contained DRB-specific transcripts. These results show that a significant proportion of very primitive myelopoietic precursor cells can be transduced with the DRB recombinant vector and that vector sequences are expressed in the differentiated progeny of these cells. These experiments are described in detail below.
  • Construction and Screening of SLA-DRB Recombinant Retroviruses As in man, Lee et al., 1982, Nature 299:750-752, Das et al., 1983, Proc. Natl. Acad. Sci. USA 80:3543-3547, the sequence of the swine DRA gene is minimally polymorphic. Therefore, transduction of allogeneic DRB cDNAs into bone marrow cells should be sufficient to allow expression of allogeneic class II DR molecules on cells committed to express this antigen.
  • Details of retroviral constructs are given in FIG. 3. Two types of retroviral constructs, GS4.4 and GS4.5, were prepared. The diagram in FIG. 3 depicts the GS4.5 retroviral construct. The arrows in FIG. 3 indicate the directions of transcription. In GS4.5, the orientation of DRB cDNA transcription is the same as viral transcription. In GS4.4 (not shown), the TK promoter and the DRB cDNA were inserted into the 3′ LTR of N2A in the reverse orientation of transcription with respect to viral transcription and the simian virus 40 3′ RNA processing signal was added. pBSt refers to Bluescript vector sequence (Stratagene). The thymidine kinase (TK) promoter was contained within the 215-base-pair (bp) Pvu II-Pst I fragment from the herpes simplex virus TK gene, McKnight, 1980 Nucleic Acids Res. 8:5949-5964. The simian virus 40 3′ RNA processing signal was contained within the 142-bp Hpa I-Sma I fragment from the pBLCAT3 plasmid, Luckow et al., (1987) Nucleic Acids Res. 15:5490-5497, (see FIG. 3). Sequence analysis of the junctions of the promoter, the class II cDNA, and the vector sequences confirmed that the elements of the constructs were properly ligated.
  • These retroviral constructs were transfected into the amphotropic packaging cell line PA317, and transfectants were selected in G418-containing medium. A total of 24 and 36 clones, transfected, respectively, with the GS4.4 and GS4.5 recombinant plasmids, were tested by PEG precipitation of culture supernatants and slot-blot analysis of viral RNA. Of these, 8 and 12 clones were found, respectively, to be positive for DRB, although the DRB signal was consistently weaker for the GS4.4-derived clones. Analysis of genomic and spliced transcripts from GS4.5 cells by dot-blot analysis of PEG-precipitated particles revealed heterogeneity among viral transcripts in various clones transfected by GS4.5. In one experiment, two clones contained DRB+/Neo+ viral RNA, two contained DRB+/Neo RNA, two contained DRB/Neo+ RNA, and one showed no class II or Neo signal. G418-resistance (G418r) titer determination of supernatants from DRB-positive clones confirmed that the average titer produced by GS4.5-transfected clones (103-104 CFU/ml) was significantly higher than that of the GS4.4-transfected clones (102-103 CFU/ml). Further transduction experiments were, therefore, conducted with the best clone, named GS4.5 C4, which produced an initial G418r titer of 3×104 CFU/ml.
  • Plasmid preparation, cloning procedures, DNA sequencing, RNA preparations, Northern blots, and RNA slot blots were performed by standard methods, Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual 2nd Ed. (Cold Spring Harbor Lab., Cold Spring Harbor). Final washes of blots were carried out in 0.1×SSPE (1×SSPE =0.18 M NaCl/10 mM sodium phosphate, pH 7.4/1 mM EDTA) at 60° C. for 30 min.
  • The packaging cell lines PA317, Miller et al., 1986, Mol. Cell. Biol. 6:2895-2902, GP+E-86, Markowitz et al., 1988, J. Virol 62:1120-1124, psiCRIP, Danos et al., 1988, Proc. Natl. Acad. Sci. USA 85:6460-6464, and their derivatives were maintained at 37° C. in Dulbecco's modified Eagle's medium (DMEM; GIBCO) with 10% (vol/vol) fetal bovine serum (CELLect Silver; Flow Laboratories) supplemented with 0.1 mM nonessential amino acids (Whittaker Bioproducts), antibiotics penicillin (5 units/ml), and streptomycin (5 μg/ml).
  • Improvement of the Viral Titer of the C4 Clone
  • Since recent data indicated that supernatants containing high retroviral titers were the best candidates for transducing bone marrow cells, Bodine et al., 1990, Proc. Natl. Acad. Sci. USA 87:3738-3742, the titer of the C4 producer clone was increased by “ping-pong” amplification, Bestwick et al., 1988, Proc. Natl. Acad. Sci. USA 85:5404-5408. Supernatant from nearly confluent C4 cultures was used to transduce GP+E-86 ecotropic packaging cells and G418 selection was applied. Forty-eight clones were isolated and screened by PEG precipitation for production of viral particles. Supernatants from 18 of these clones were DRB-positive by dot-blot analysis of viral RNA and had G418r titers between 0.5 and 3.5×104 CFU/ml). One positive clone was then amplified by the ping-pong technique with the amphotropic hygromycin-resistant packaging line psiCRIP. Supernatants from 48 hygromycin-resistant clones were examined for presence of DRB-positive viral RNA by PEG precipitation and their G418r titers were determined. All the clones were positive by dot-blot analysis with the DRB probes and produced titers between 1×105 and 1×107 CFU/ml. Amphotropic clone GS4.5 A4, which produced the highest titer, was tested for the presence of helper virus by the S+L-assay. No replication-competent helper virus was detected.
  • Amplification of virus titer was achieved by the ping-pong technique. Since there is evidence that psiCRIP packaging cells are less prone to produce helper virus than PA317 when using certain types of vectors, Miller, 1990, Hum. Gene Therapy 1:5-14, DRB recombinant virions were prepared using the psiCRIP/GP-E-86 producer combination. Titer values>1×107 CFU/ml with no detectable amphotropic helper viruses were obtained, confirming that this strategy produced safe viral particles suitable for in vivo experiments.
  • Northern blot analysis of GS4.5-producing clones C4, A9, and A4, each derived from a different packaging cell line, showed a conserved hybridization pattern. RNA species corresponding to the full-length viral genome, the spliced Neo transcript, and the DRB transcription unit were observed with additional RNA species. High molecular size species observed in these experiments may constitute a read-through transcript starting from the TK promoter and ending in the other long terminal repeat (LTR). In contrast to many of the virion-producer clones-obtained by transfection that presented erratic DRB transcripts, those obtained by transduction showed stable DRB hybridization patterns suggesting that no recombination events occurred during the amplification procedure.
  • Retroviral titers were determined as follows. Replication-defective retroviral particles were produced from packaging cell lines initially transfected with recombinant construct using the standard calcium phosphate precipitation method, Wigler et al., 1978, Cell 14:725-733. Retrovirus production was estimated by the drug-resistance titer (G418-resistant colony-forming units/ml, CFU/ml) as described, Bodine et al., 1990, Proc. Natl. Acad. Sci. USA 87:3738-3742. Except for the psiCRIP line, G418 (GIBCO) selection was carried out in active component at 500 μg/ml for 10-12 days. Hygromycin B selection was applied to psiCRIP-derived packaging clones in medium containing active drug at 50 μg/ml for 10 days. Replication-competent helper virus titer was assayed on PG4 feline cells by the S+L method, Bassen et al., 1971, Nature 229:564-566.
  • PEG precipitation of viral particles was performed as follows. Virions contained in 1 ml of culture supernatant were precipitated with 0.5 ml of 30% (wt/vol) polyethylene glycol (PEG) for 30 min. at 4° C. After centrifugation, the pellets were treated with a mixture of RNase inhibitors (vanadyl ribonuclease complex, BRL), phenol/chloroform-extracted, and ethanol-precipitated. Pellets were then resuspended in 15.7% (vol/vol) formaldehyde and serial dilutions were dotted onto nitrocellulose membrane.
  • Analysis of DRB Transcription in Packaging Cell Clones To test for accurate transcription of the introduced DRB cDNA within the different producer clones, Northern blots containing RNAs isolated from these clones were hybridized with the DRB and Neo probes. FIG. 4 depicts the structure of the provirus genome and the expected sizes of transcripts initiated from either the viral LTR or the TK promoters. Each of the three GS4.5-containing clones, which were derived from PA317 (clone C4), GP+E-86 (clone A9), and psiCRIP (clone A4) cells, showed DRB-positive transcripts. As reported, Hantzopoulos et al., 1989, Proc. Natl. Acad. Sci. USA 86:3519-3523, the unspliced genomic RNA (band a) and the spliced Neo transcript (band b) were observed. In addition a transcript uniquely hybridizable with the DRB probe was detected that corresponds to the size predicted (1700 bases, band c) for the DRB cDNA transcription unit.
  • Surface Expression of the SLA-DR Antigen on Transduced Fibroblasts An in vitro assay was developed to examine surface expression of the SLA-DR antigen on murine fibroblasts. Flow cytometry (FCM) profiles shown in FIG. 5 demonstrate that G418r titers of 3×104 (clone C4) were sufficient to promote expression of the DR antigen on the cell surface of transduced DRA transfectants. In FIG. 5 solid lines indicate DR cell surface expression (anti-DR antibody binding) (22% and 75% of the bulk population of cells 3 days after transduction with GS4.5 C4, (B) and GS4.5 A4 (C), respectively); dashed lines indicate anti-mouse class I antibody binding (positive control); dotted lines indicate anti-pig CD8 antibody binding (negative control). Twenty-two percent of the bulk population of transduced cells were DR-positive and subclones maintained class II expression for more than 5 months. The increase in titer (clone A4) correlated with an increase in the number of cells transduced (75% of the transduced population was DR-positive) and with the brightness of the DR signal.
  • The class II transduction assay was performed as diagrammed in FIG. 6. NIH 3T3 cells were transfected with the SLA-DRAd cDNA inserted in a plasmid expression vector, Okayama et al., 1982, Mol. Cell. Biol. 2:161-170. Approximately 3×104 cells of a stable DRA transfectant (clone 11/12.2F) that expressed a high level of DRA mRNA were then transduced overnight with 1 ml of DRB-containing retroviral supernatant. Cells were subsequently cultivated in fresh DMEM supplemented with 10% fetal bovine serum and antibiotics for 2 additional days and examined for cell surface expression of the DR antigen by FCM analysis.
  • The class II transduction assay described here provides a fast and simple method to test both the expression and functional titer of retroviral constructs. By using cells transfected with DRA, the need for lengthy double selection after transduction by two separated vectors, Yang et al., 1987, Mol. Cell Biol. 1:3923-3928; Korman et al., 1987, Proc. Natl. Acad. Sci. USA 84:2150-2154, is obviated. Cell-surface expression of DR heterodimers was demonstrated by FCM analysis 3 days after transduction, providing direct evidence that the transferred sequences were sufficient to produce significant level of DR β chain. More importantly, this test allows determination of “functional” titers based on the expression of the gene of interest rather than on that of the independently regulated drug-resistance marker.
  • The SLA-DRB probe was an EcoRI cDNA fragment containing the complete coding sequence of the DR β chain, Gustafsson et al., 1990, Proc. Natl. Acad Sci. USA 87:9798-9802. The neomycin phosphotransferase gene (Neo) probe was the Bcl I-Xho I fragment of the N2A retroviral plasmid, Hantzopoulos et al., 1989, Proc. Natl. Acad Sci. USA 86:3519-3523.
  • Expression of Porcine DRB cDNA Transduced into Murine Bone Marrow Progenitor Cells The efficiency with which myeloid clonogenic precursors were transduced was determined by assaying for CFU-GM with and without a selecting amount of G418 after exposure of bone marrow cells to GS4.5-derived virions. Comparison of the number of colonies that formed in the presence and absence of the drug, for two experiments, indicated that ≈40% of the initial population of myeloid progenitor cells were transduced. The frequency of G418r CFU-GM was again determined after a sample of the transduced marrow was expanded under long-term culture conditions for 33 days. Twenty-five percent of the progenitors present after 33 days in culture still gave rise to colonies under G418 selection. Colonies of cells arisen from CFU-GM were examined for the presence of DRB-specific transcripts by converting RNA into cDNA and then performing PCR amplification as described herein and in Shafer et al., 1991 Proc. Natl. Acad. Sci. USA 88:9670. A 360-bp DRB-specific product was detected in five of six G418-selected colonies from freshly transduced marrow, whereas all six colonies similarly derived from transduced progenitors present after 33 days in culture were positive. An additional band of 100 bp observed in some of the samples probably reflects the stochastic nature of nonspecific priming events. DRB-specific transcripts were also detected in the bulk population of drug-resistant colonies and in producer cells but were not detected in controls such as a bulk population of untransduced colonies, fibroblasts used to provide carrier RNA, and a bulk population of transduced colonies processed as above but without reverse transcriptase. These latter data demonstrate that the PCR signal was dependent on the synthesis of cDNA, excluding the possibility that provirus, rather than viral message, was responsible for the amplified fragment.
  • Recent improvements including modifications of the virus design, increase of viral titers, use of growth factors to stimulate precursor cells, and selection of stem cells prior to transduction have been shown to improve long-term expression of transduced genes in the hematopoietic compartment, Bodine et al., 1990, Proc. Natl. Acad. Sci. USA 87:3738-3742; Bodine et al., 1989, Proc. Natl. Acad. Sci. USA 86:8897-8901; Wilson et al., 1990, Proc. Natl. Acad. Sci. USA 87:439-443; Kang et al., 1990, Proc. Natl. Acad. Sci. USA 87:9803-9807; Bender et al., 1989, Mol. Cell. Biol. 9:1426-1434. The experiments herein show the applicability of the retroviral gene-transfer technique in achieving expression of major histocompatibility complex class II genes transferred into hematopoietic cells. To determine the efficiency with which developmentally primitive hematopoietic cells were transduced, the frequency of G418r CFU-GM was assessed after expanding infected marrow cells kept for 33 days in long-term cultures. Expression of the exogenous DRB cDNA was also monitored in cells derived from transduced CFU-GM present either immediately after infection or after an extended culture period. Virtually all of the colonies individually tested were positive for DRB-specific transcript, suggesting that the DRB recombinant vector is suitable for expression in murine hematopoietic cells.
  • Bone marrow cells were obtained from the femora of 6- to 12-week-old female C57BL/10 mice and were prepared as described, Ildstad et al., 1984, Nature 307:168-170. Methylcellulose colony assays for granulocyte/macrophage colony-forming units (CFU-GM), Eaves et al., 1978, Blood 52:1196-1210, were performed as described using 5% (vol/vol) murine interleukin 3 culture supplement (Collaborative Research). Long-term Dexter-type bone marrow cultures were initiated in 60-mm culture dishes with 2×107 nucleated cells, Eaves et al., 1987, CRC Crit. Rev. Oncol. Hematol. 7:125-138.
  • Bone marrow cells were transduced essentially as described, Bodine et al., 1989, Proc. Natl. Acad. Sci. USA 86:8897-8901. Briefly, bone marrow was harvested for 6-12-week-old female C57BL/10 donors that had been treated 2 days with 5-fluorouracil (150 mg/kg). Prestimulation was performed by incubating 1×106 cells per ml for 2 days in long-term Dexter-type bone marrow culture medium to which was added 7.5% interleukin 3 culture supplement and recombinant human interleukin 6 (200 units/ml; gift from J. Jule, National Institutes of Health, Bethesda, Md.). Marrow cells were transduced for 48 hr by adding 5×106 cells per 10-cm plate containing nearly confluent virus-producers, Polybrene (8 mg/ml), and the cytokines described above.
  • Detection of DRB-Specific Transcripts in CFU-Derived Colonies was performed as follows. Cells corresponding to individual CFU colonies and to colonies present on an entire plate (bulk) were first extracted from methylcellulose cultures by dilution in phosphate-buffered saline and centrifugation. These cells were then combined with 1×106 NIH 3T3 cells (to provide carrier RNA), and total RNA was prepared using the guanidine isothiocyanate/CsCl method. First-strand cDNA was prepared from 20 μg of total RNA using the Invitrogen Red Module kit. cDNA was then subjected to 50 cycles of PCR amplification in the presence of the SLA DRB-specific oligonucleotides 04 (5′-CCACAGGCCTGATCCCTAATGG) (Seq. I.D. No. 1) and 17 (5′-AGCATAGCAGGAGCCTTCTCATG) (Seq. I.D. No. 2) using the Cetus GeneAmp kit as recommended (Perkin-Elmer/Cetus). Reaction products were visualized after electrophoresis on a 3% NuSieve agarose gel (FMC) by staining with ethidium bromide.
  • FCM analysis was performed with a FAC-Scan II fluorescence-activated cell sorter (Becton Dickenson) on cells stained with the anti-DR monoclonal antibody 40D, Pierres et al., 1980, Eur. J. Immunol. 10:950-957, an anti-H-2d allo antiserum, or the anti-porcine CD8 monoclonal antibody 76-2-11, Pescovitz et al., 1984, J. Exp. Med. 160:1495-1505, followed by fluorescein isothiocyanate-labeled goat anti-mouse antibodies (Boehringer Mannheim).
  • Expression of Allogeneic Class II cDNA in Swine Bone Barrow Cells Transduced with a Recombinant Retrovirus
  • A MHC gene (DRB) was transferred into clonogenic progenitor cells from swine using a recombinant retroviral vector (GS4.5) and a transduction protocol designed to be applicable in vivo. Both the selectable drug resistance gene and the allogeneic class II cDNA transferred by this vector were expressed in the progeny of these transduced progenitors. Expression of the Neo gene was monitored functionally by colony formation under G418 selection, while the presence of class II transcripts was detected by PCR analysis. With this latter method, the transcriptional expression of both endogenous and virally derived DRB genes in transduced and selected colonies were demonstrated.
  • Primary porcine fibroblasts were cultured with high titer viral supernatants, and then analyzed by northern blotting using probes specific for DRB and Neo. A specific transcript was observed which was uniquely hybridizable with the DRB probe and migrated at the position predicted (1700 bases) for the DRB cDNA transcription unit arising from the TK promoter and terminating at the LTR 3′ RNA processing site.
  • To determine whether GS4.5 containing virions could transduce swine myelopoietic progenitor cells a colony assay adapted for swine CFU-GM was used. Transductions were carried out by incubating bone marrow from a donor of the SLAC haplotype in high titer viral supernatant. Comparisons of the number of colonies which formed in the presence and absence of G418 for a total of 5 independent experiments indicated that 5% to 14% of CFU-GM were transduced.
  • Colonies of cells originating from transduced CFU-GM were examined for the presence of DRB-specific transcripts by converting RNA into cDNA, and then performing PCR amplification. Utilizing a polymorphic Sau3AI restriction site absent from the endogenous DRBc gene, the presence of DRBd-specific transcripts was unambiguously demonstrated. Gel electrophoresis of the PCR product demonstrated that a 183/177 bp doublet indicative of the vector-derived DRBd transcript was amplified in samples derived not only from pools of transduced and selected CFU-GM progeny, but also from at least 4 out of 6 individual colonies tested. A 360 bp PCR fragment, indicative of endogenous DRBc transcripts, was also amplified not only as expected from PBL isolated from an SLAC donor, but also from both of the pooled colony samples and a number of the individual colony samples.
  • Construction of the retrovirus GS4.5, and production of high titer viral supernatants was as described above. Detection of DRB-specific transcripts in CFU-derived colonies by PCR of cDNA were described above and as follows. Bone marrow from an SLAc donor was exposed to GS4.5-containing virions, and G418 selected colonies were tested for the presence of DRBc (endogenous) and DRBd (vector derived) specific transcripts by PCR of cDNA followed by digestion with Sau3AI and agarose gel electrophoresis. Controls were as follows: template synthesized either in the presence or absence of reverse transcriptase; template derived from cells producing GS4.5-containing virions, from PBL isolated from SLAc or SLAd donors, and from untransduced producer cells used as carrier RNA.
  • Transduction of bone marrow was performed as follows. Swine bone marrow was harvested as previously described (Pennington et al., 1988, Transplantation 45:21-26) and transductions were carried out by incubating marrow cells in high titer viral supernatants at an m.o.i. of 3-5 in the presence of 8 μg of polybrene per ml at 37° C. for 5 hr. Myeloid progenitors were assayed by colony formation in methylcellulose cultures using PHA-stimulated swine lymphocyte conditioned medium as a source of growth factors. Selective medium contained 1.2 mg/ml active G418.
  • Transduced bone marrow was administered to a lethally irradiated miniature swine. At 5 weeks peripheral blood lymphocytes were analyzed by Southern, northern, and cell-surface FACS analyses. By all of these test there was evidence of presence of the transduced allogeneic class II gene in these cells and for expression of the product of this gene. In particular, northern analysis showed bands characteristic of the transcribed cDNA, and FACS analysis with a combination of alloantisera and monoclonal antibodies to DR showed presence of the transduced allele of DR beta on the surface of peripheral lymphocytes.
  • Allogeneic Tolerance
  • Development of the B10.MBR-B10.AKM Strain Combination In an attempt to maintain strains which are truly congenic for the MHC, a program of continuous backcrossing of each congenic line to a common background partner strain was instituted more than 15 years ago. Backcross animals were intercrossed and appropriate progeny selected by serologic typing in order to reestablish each congenic line. Thus, C57BL/10 was used as one reference background strain and all other congenic lines on the B10 background were backcrossed once every six to ten generations to this C57BL/10 line.
  • During the backcrossing of each congenic line to its pedigreed reference line, there is of course the chance for an intra-MHC recombination event to occur. Typing of the intercross (F2) generation serologically reveals such recombinant events, and when the recombinant provides a new haplotype of potential interest for genetic studies, it is outcrossed and then intercrossed to produce a homozygous new recombinant H-2 haplotype. One of the most valuable of such recombinants originating in this colony is the B10.MBR line, Sachs et al., 1979, J. Immunol. 123:1965-1969, which was derived from a recombination event during the backcrossing of B10.AKM to C57BL/10. Because this strain was the first to separate Kb from Ik it has been used extensively in studies of R-2 immunogenetics. In addition, in combination with the parental B10.AKM strain, the B10.MBR offers the possibility of examining an isolated K gene as the only MHC difference between these two strains. Thus, as illustrated in FIG. 7, introduction of the Kb gene into B10.AKM bone marrow stem cells, could theoretically lead to expression of all cell surface MHC antigens of the B10.MBR. Expression on bone marrow derived cell populations produces transplantation tolerance to the product of the transduced gene, and this tolerance can be tested by a tissue graft from the B10.MBR strain.
  • Reconstitution of Myeloablated Mice with Transduced Bone Marrow Eighty prospective donor B10.AKM mice were treated with 150 mg/kg 5 FU on day −7. Bone marrow was harvested from these mice on day −5, treated with anti-CD4 and anti-CD8 monoclonal antibodies (mAbs) plus complement to remove mature T cells, and cultured for five days with N2-B19-H2b virus-containing supernatant (H2) from the psi-Crip packaging cell line. As a control, one-half of the marrow was cultivated with supernatant from control packaging cells not containing N2-B19-H2b (A2). On day zero, 45 B10.AKM recipients received 10 Gy total body irradiation (TBI), followed by administration of various concentrations of cultured bone marrow cells (A2 or H2). Kb expression On day 13 several animals receiving the lowest doses of cultured bone marrow were sacrificed and individual spleen colonies were harvested and analyzed by PCR for the presence of N2-B19-H2b DNA. In addition, spleen cell suspensions were prepared and analyzed for cell surface expression of Kb by flow microfluorometry on a fluorescence-activated cell sorter (FACS). FACS analyses indicated that all animals receiving the H2-treated marrow showed some Level of Kb expression above control staining with the non-reactive antibody. The results are shown in FIG. 8 which is a FACS profile of spleen cells from a recipient of transduced bone marrow: A=Anti Kb antibody; B=control antibody. Spleen cells from recipients of non-transduced marrow were also negative. In addition, the PCR analysis showed every colony examined to contain the transduced DNA. Animals were thereafter followed by FACS and PCR on peripheral blood lymphocytes (PBL). On day 28 and again on day 40, PCR analyses were positive. However, FACS analysis for cell-surface expression was variable, with PBL from most H2 animals showing only a slight shift of the entire peak for staining with anti-Kb, as compared to PBL from A2 animals stained with the same antibody, or as compared to PBL from H2 animals stained with the non-reactive HOPC antibody.
  • Allogeneic grafts On day 40 skin from B10.MBR (Kb specific) and B10.BR (control, third party class I disparate) donors was grafted onto all animals. Graft survivals were scored daily by a blinded observer (i.e., readings were made without knowledge of which graft was from which donor strain) until rejection was complete. The survival times are shown in FIG. 9, and indicate marked specific prolongation of survival of the B10.MBR skin grafts on the recipients of Kb-transduced BMC (FIG. 9B), but not on recipients of control marrow (FIG. 9A). One of the animals with a long-standing intact B10.MBR skin graft was sacrificed at day 114 and cell suspensions of its lymphoid tissues were examined by FACS and compared to similar suspensions of cells from an animal which had rejected its B10.MBR skin graft. A striking difference was noted in staining of thymus cells with an anti-Kb mAb. Cell suspensions were prepared and stained either with the anti-Kb mAB 28-8-6 or the control antibody HOPC1. A subpopulation of thymus cells from the tolerant animal showed a marked shift toward increased staining with 28-8-6 compared to HOPC1, while there was essentially no change in the staining pattern of thymocytes from the animal which had lost its graft. FIG. 10 shows FACS analysis on thymocytes from skin graft rejecter (FIGS. 10A, B) and skin graft acceptor (FIGS. 10C, D). Staining with control HOPC1 antibody (FIGS. 10A, C) and with specific anti-Kb antibody (FIGS. 10B, D). A similar comparison of staining patterns on bone marrow cells showed the presence of low level Kb expression on a cell population in the marrow of the tolerant mouse, but not of the mouse which had rejected its skin graft. These results indicate that a pluripotent stem cell or early progenitor cell population expressed Kb in the tolerant mouse but not in the rejecter mouse, and that this BMC stem cell provided a continuous source of Kb antigen in the thymus on cells which are critical for the inactivation of developing thymocytes with Kb-reactive TCR. It is of interest to note that Kb expression was not detected on splenocytes of the tolerant mouse, and that, in general, splenocyte expression did not correlate with skin graft tolerance. Since the spleen contains T cells which mature in the thymus, these results suggest that either thymocytes lose expression of Kb as they mature, or that the Kb-bearing thymocytes of this animal were cells of a non-lymphoid lineage, such as macrophages.
  • Long-term expression As discussed above, the B10.AKM and B10.MBR congenic mouse strains are identical except in the MHC class I region. A recombinant retrovirus containing the class I gene from the B10.MBR stain (H-2Kb) linked to a B19 parvovirus promoter (B19-H2Kb) and a neomycin resistance (neor) gene was introduced into B10.AKM (H-2Kk) marrow cells. As a control, a recombinant retrovirus containing only the neor gene was introduced into B10.AKM marrow cells. The transduced marrow was injected into lethally irradiated AKM recipients pre-treated with an anti-CD8 monoclonal antibody. Twelve weeks post BMT, quantitative PCR was used to show that the B19-H-2Kb proviral sequences were present in 5%-30% of peripheral blood cells in all recipient animals. Reverse transcriptase PCR was used to demonstrate the B19-H-2Kb mRNA in RNA isolated from bone marrow and spleen of a subset of recipient animals.
  • Construction of the Kb Retroviral Vector The retroviral vectors used the Maloney murine leukemia virus based vector N2, Armentano et al., 1987, J. Virol. 61:1647-1650. The coding regions within this virus were deleted during its construction, and replaced with the selectable marker gene, neomycin phosphotransferase (Neo), which is transcribed from the viral LTR promoter, and provides drug resistance to G418. This conventional N2 virus was then further modified by insertion of a parvovirus-derived promoter, B19, Liu et al., 1991, J. Virol. (In Press), downstream from Neo, followed by 1.6 kb of cDNA coding for the class I antigen H-2Kb to form the new recombinant virus N2-B19-H26. FIG. 11 depicts the N2-B19-H26 retroviral vector: P=PstI; X=XhoI; H=HinDIII; E=EcoRI; B=BamHI. This latter cDNA was derived by Waneck et al. during the construction of an H-2b cDNA library for their purposes, Waneck et al., 1987, J. Exp. Med. 165:1358-1370.
  • Viral producer cell lines were developed using the packaging cell lines for amphotropic (psi-Crip), Danos et al., 1988, Proc. Natl. Acad. Sci 85:6460-6464, and ecotropic (psi-2), Sambrook et al., 1989, Molecular cloning: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, viral production. These cell lines have been specially designed to produce structural viral proteins for the recombinant defective virus to be produced. Viral production was achieved by transfecting psi-Crip with N2-B19-H2b. Both amphotropic and ecotropic producer cell lines were then co-cultivated allowing multiple integration events and high expression [i.e. the “ping-pong” technique see Bestwick et al., 1988, Proc. Natl. Acad. Sci 85:5404-5408]. In this technique, co-cultivation overcomes viral antigen receptor blockage by endogenously secreted proteins since amphotropic and ecotropic viruses recognize different receptors. Ecotropic psi-2 viral producer clones were then selected which produced titers of G418 resistance on 3T3 cells of greater than 107 cfu/ml.
  • In order to ensure that Kb was being expressed from the recombinant virus, transduced 3T3 cells were stained with a monoclonal antibody specific to this antigen and analyzed by flow microfluorometry. These experiments clearly demonstrated high level expression of virally derived Kb.
  • Animals and husbandry were as follows. The B10.BMR strain, [Sachs et al., 1979, J. Immunol. 123:1965-1969, was provided to the Jackson Laboratory, Bar Harbor, Me. about 6 years ago, and specific pathogen-free stock animals of this strain are now available from that source. Upon arrival in animals should be transferred to autoclaved microisolator cages containing autoclaved feed and autoclaved acidified drinking water. Sterile animal handling procedures which are effective in maintaining animals free of pathogens so that interpretable survival studies can be performed should be used.
  • Bone marrow transplantation was performed as follows. Techniques for bone marrow transplantation in mice are known to those skilled in the art, see e.g., Sykes et al., 1988, J. Immunol. 140:2903-2911. Briefly, recipient B10.AKM mice aged 12 to 16 weeks are lethally irradiated (1025R, 137Cs source, 110R/min) and reconstituted within 8 hours with 2.5×106 bone marrow cells, obtained from the tibiae and femora of sex-matched donors aged 6-14 weeks. Animals are housed in sterilized microisolator cages, and receive autoclaved food and autoclaved acidified drinking water. For these studies some modifications of this general technique are required, since the syngeneic bone marrow will have been transduced with an allogeneic gene, and since the bone marrow will come from 5FU-treated mice, which should have lower total cell counts but higher stem cell content than normal mice. The protocol is therefore as follows:
  • 1. Donors will be treated with 5-Fluorouracil, 150 mg/kg i.v. on day −7 in order to induce pluripotent stem cells to cycle.
  • 2. Marrow will be harvested from donors on day −5, and T cell depleted with mAbs and complement.
  • 3. Marrow will than be cultured for 5 days in supernatant from an ecotropic packaging cell line (B17H2Kb-18) which produces a high titer of non-infectious retroviral particles containing the Kb gene (see below). IL-3 and IL-6 will be added to the cultures.
  • 4. On day 0, recipient B10.AKM mice will be lethally irradiated (10.25 Gy), and will be reconstituted with 2.5×106 BMC transduced with the Kb gene. Control animals will be similarly treated, except that they will receive marrow exposed to supernatant from a similar ecotropic packaging line not exposed to a Kb-containing vector. The recipient may also be pre-treated with anti-CD8 monoclonal antibody.
  • Cellular and serological assays are performed as Follows.
  • Anti-class I Cell-Mediated Lympholysis (CM) Assay: Spleens are removed from BMT recipients and normal mice, red cells are lysed using ACK buffer, and a single cell suspension is prepared. Cells are filtered through 100-mesh nylon, washed, and resuspended at 4×106/ml in complete medium consisting of RPMI 1640 with 10% fetal calf serum, 0.025 mM 2-mercapteothanol, 0.01M Hepes buffer, 0.09 mM nonessential amino acids, 1 mM sodium pyruvate, 2 mM glutamine, 100 U/ml penicillin and 100 ug/ml streptomycin. 90 μl of responder cells are added to Costar 96-well round-bottomed plates along with irradiated (30 Gy) stimulator splenocytes. Cultures are set up in two rows of 3 replicates each, and after 5 days of incubation in 6% CO2 at 37° C., twofold serial dilutions are prepared from the second row oftriplicates, so that cytolytic capacity can be examined at a total of 5 different responder:target ratios. 51Cr-labelled 2-day concanavalin A-induced lymphoblasts are then added at 104 blasts per well and incubated for 4 hr at 37° C., 6% CO2. Plates are harvested using the Titertek supernatant collection system (Skatron, Inc., Sterling, Va.) and 51Cr release is determined using an automated gamma counter. Cytolytic capacity is measured directly in the original cell culture plated, so that the measurement is based on the number of responders plated, rather than on the number of live cells present at the end of the 5-day incubation period. This methodology has been developed and used successfully in this laboratory for several years for analysis of spleen cell responses from individual animals [Sykes, M., et al., 1988 J. Immunol. 140:2903-2911]. Percent specific lysis is calculated using the formula: % Specific Lysis = Experimental release - Spontaneous release × 100 % Maximum release - Spontaneous release × 100 %
    Limiting dilution analyses: Responder and stimulator (6×105, 30 Gy irradiated) cells are co-cultured for 7 days in complete medium containing 13% TCGF [lectin-inactivated con A supernatant obtained from BALB/c con A-Activated splenocytes) in 96-well plates. Wells containing 105 (24 wells), 3×104 (24 wells), 104 (30 wells), 3000 (30 wells), 1000 (30 wells), 300 (30 wells), and 100 (30 wells) responder cells are prepared. Three thousand 51Cr-labeled con A blasts are added to each well on day 7, and 4 hour 51Cr release is measured. Wells are considered positive if 51Cr release is 3 standard deviations greater than the mean 51Cr release in 24 wells containing stimulator cells only plus similar numbers of target cells. The Poisson distribution is used to determine the frequency of precursor CTL's which recognize each target, and statistical analysis is performed by the Chi square method of Taswell, Taswell, 1981, J. Immunol. 126:1614.
  • Flow microfluorometry: One-color and two-color flow cytometry will be performed, and percentages of cells expressing a particular phenotype will be determined from 2-color data, as previously described in detail Sykes, 1990, J. Immunol. 145:3209-3215. The Lysis II software program (Becton Dickinson) will be used for distinguishing granulocytes from lymphocytes by gating on the basis of forward angle and 90° light scatter. Cell sorting will be performed on a Coulter Epics Elite cell sorter. Cell suspensions for flow cytometry: PBL, BMC, thymocyte, splenocyte, and lymph node suspensions will be prepared as previously described, Sykes, M. et al., 1988, J. Immunol. 140:2903-2911; Sykes, M. 1990, J. Immunol. 145:3209-3215; Sharabi, Y. et al., 1990, J. Exp. Med. 172:195-202. Whole peripheral white blood cell suspensions (including granulocytes) will be prepared by centrifugation of heparinized blood for 2 minutes at 14,000 RPM in an Eppendorf centrifuge, followed by aspiration of the buffy coat layer. These cells will be transferred to a 15 ml. conical tube and washed. Red blood cells (RBC) contaminating the remaining pellet will be lysed by exposure for 5 seconds to 4.5 ml of distilled H2O followed by rescue with 0.5 ml of 10×PBS.
  • Cell staining: One-color and two-color staining will be performed as we have previously described, Sykes, M., 1990, J. Immunol. 145:3209-3215; Sykes et al., 1988, J. Immunol. 141: 2282-2288. Culture supernatant of rat anti-mouse RcτR mAb 2.4G2, Unkeless, J. C., 1979, J. Exp. Med. 150:580-596, will be used for blocking of non-specific staining due to FcτR binding, whenever only direct staining is used. The following mAbs are used: biotinylated murine Kb-specific IgG2a mAb 28-8-6, Ozato et al., 1981, J. Immunol. 126:317-321, and control murine IgG2a, mAb HOPC1 (with no known specific binding to murine antigens) are prepared by purification on a protein A-Sepharose column, and are biotinylated by standard procedures used in our laboratory; rat anti-MAC1 mAb M1/70, Springer et al, 1979, Eur. J. Immunol. 9:301, is used as culture supernatant, and will be stained by mouse anti-rat IgG-specific mAb MAR18.5; FITC-labeled rat-anti-mouse granulocyte antibody Gr1 is purchased from Zymed; FITC-labeled rat-anti-mouse Thy1.2 mAb will be purchased from Becton-Dickinson; FITC-labeled mouse-anti-human CD3 mAb Leu4 (Becton Dickenson) is used as a directly FITC labeled negative control antibody.
  • Thymic tissue immunofluorescence: The tissue is incubated in L15 medium for 24 hours to reduce background staining, and is then cut and embedded in O.C.T. compound for freezing in Isopentane. Frozen sections are prepared (thickness 4 μm) on a cryostat, dried, fixed in acetone, then washed in PBS. The first antibody incubation (with 28-8-6) is performed in the presence of 2% normal mouse serum, in order to saturate Fc receptors. After 45 minutes, the slides are washed 4 times, and FITC-conjugated secondary reagent (monoclonal rat-anti-mouse IgG2a-FITC, purchased from Pandex) is added. After 45 minutes' incubation with the secondary reagent, four washes are performed and the tissue is mounted. Sections are examined under a fluorescence microscope by an observer who is unaware of the group of animals from which the tissue was obtained.
  • Bone Marrow Manipulations and Assays were Performed as Follows:
  • Transduction of murine bone marrow stem cells: The methodology used for transduction of bone marrow cells has been described previously, Karlsson et al., 1988, Proc. Natl. Acad. Sci. 85:6062-6066. Bone marrow is harvested from 6-12 week old female B10.AKM donors treated 2 days previously with 150 mg/kg 5-FU. Following T cell depletion (see above), the marrow is divided and 107 cells per 10 cm plate are cultured for 5 days in the presence of 8 μg of Polybrene per ml, 10% FCS, 0.6% IL-3-containing supernatant, 0.6% IL-6-containing supernatant, and fresh supernatants from B19H2Kb or N2 cells. IL-3- and IL-6-containing supernatant is 48 hour supernatant of COS 7 cells transfected with the murine rIL-3 gene-containing plasmid pCD-IL-3 or with the murine rIL-6 gene-containing plasmid pCD-IL-6, respectively (both plasmids provided by Dr. Frank Lee, DNAX Corp.). IL-3-containing supernatants are tittered by testing proliferation of the IL-3-dependent cell line 32D in the presence of dilutions of these supernatants, and IL-6 is tittered in a similar manner using the IL-6-dependent line T1165 as the indicator cell line. We will also test the effect of murine SCF on bone marrow transduction, as recently described, Zsebo et al., 1990, Cell 63:125-201.
  • The virus-containing supernatants are refreshed on a daily basis by harvesting the non-adherent layer of each plate, pelleting the cells, and resuspending in freshly harvested filtered virus-containing B19H2Kb or N2 supernatant with additives. After 5 days, the non-adherent and adherent BMC are harvested, washed, and resuspended at 2.5×106/ml in Medium 199 with Hepes buffer and Gentamycin plus Heparin 10 U/Ml. One ml. of this suspension is injected i.v. to irradiated mice.
  • Murine CFU-GM assay: To test for the bone marrow progenitor cells known as CFU-GM (colony forming unit-granulocyte/macrophage), bone marrow cells are suspended in plating medium consisting of IMDM medium containing 30% defined fetal bovine serum (FBS) (HyClone, Logan, Utah), 10−4 M β-mercaptoethanol, antibiotics, 5% v/v murine IL-3 culture supplement (Collaborative Research Inc., Bedford, Mass.) and 0.8% methylcellulose (achieved by adding 36% v/v of a commercially prepared solution purchased from the Terry Fox Laboratory, Vancouver). 1.1 ml of this suspension is then dispensed into 35 mm tissue culture plates (in duplicate), and placed in a 37° C. incubator. The resulting CFU-GM derived colonies are enumerated microscopically after 5-7 days. Transduced CFU-GM are selected by including 0.9 μg/ml active G418 in the culture medium. The transduction frequency is then determined by the ratio of CFU-GM which form colonies in the presence and in the absence of the drug.
  • Molecular methods were as Follows:
  • Construction of N2-B19-H2b vector: This vector was constructed staring from the original retroviral vector N2, Eglitis et al., 1985, Science 230:1395-1398, as modified by Shimada to include an additional BamHI site immediately 3′ of the XhoI site. It includes the Kb cDNA previously cloned in the vector pBG367, as described by G. Waneck, Waneck et al., 1987, J. Exp. Med. 165:1358-1370. This gene has been placed under control of the B19 promoter, a highly efficient parvo virus derived promoter, Liu et al., 1991, J. Virol. In Press:] to produce the N2-B19-H2b construct.
  • Southern blot analysis can be performed on DNA extracted from PBL, thymocyte, BMC, splenocyte or lymph node cell suspensions using standard methods, Ausubel et al., 1989, Current protocols in molecular biology. John Wiley & Sons, New York, and probing will be performed with the fragment of Kb cDNA released from pBG367 by EcoRI. The genomic DNA will be cut with enzymes capable of distinguishing the transduced Kb from other class I genes of the B10.AKM strain. From known sequences it would appear that EcoRI may be satisfactory for this purpose, since it should liberate a 1.6 kb band from the transduced Kb cDNA, which is distinct from both the expected endogenous Kk and Dq class I bands of B10.AXM, Arnold et al., 1984, Nucl. Acids Res. 12:9473-9485; Lee et al., J. Exp. Med 168:1719-1739. However, to assure that there is no confusion with bands liberated from other class I and class I-like genes we will test several enzymes first on DNA from B10.AKM and choose appropriate restriction enzyme combinations.
  • PCR analysis of DNA can be performed using primers previously shown to be effective in our preliminary studies (see FIG. 6):
    (Seq. ID No. 3)
    5′ primer: 5′-GGCCCACACTCGCTGAGGTATTTCGTC-3′
    (covers 5′ end of α1 exon)
    (Seq. ID No. 4)
    3′ primer: 5′-GCCAGAGATCACCTGAATAGTGTGA-3′
    (covers 5′ end of α2 exon)
  • DNA is subjected to 25 cycles of PCR amplification using these specific oligonucleotides and the Cetus GeneAmp kit (Perkin Elmer Cetus, Norwalk, Conn.) according to the manufacturer's directions. In addition, [32]PdCTP will be included in the PCR reaction in order to visualize products by autoradiography following electrophoresis.
  • RNA can be isolated from 5×106 to 5×107 cells using the guanidine isothiocyanate and CsCl methods, Chirgwin et al., 1979, Biochem. 18:5294-5308, and will be used for northern analyses, RNase protection analyses, and for PCR analyses of products formed by reverse transcriptase. For situations in which less then 5×106 cells are available, for example following tail bleedings of individual mice, we will utilize the QuickPrep mRNA Purification Kit (Amgen) as miniaturized RNA preparation procedure.
  • Northern analyses can be carried out using standard methods, Ausubel et al., 1989, Current protocols in molecular biology John Wiley & Sons, New York, and the same Kb cDNA-derived probe. Vector-derived Kb mRNA is larger than endogenous class I transcripts (2.5 kb vs. 1.6 kb) due to the inclusion of vector sequences between the 3′ end of the cDNA and the poly-adenylation site in the viral 3′ LTR. It should therefore be easy to distinguish the vector-derive Kb mRNA from endogenous transcripts that might cross-hybridize with a Kb cDNA probe. We will also utilize probes derived from unique non-Kb sequences of the transcript (e.g., from B19 or N2 derived vector sequences).
  • RNAse protection analyses-are more sensitive than standard northern blots, yet still quantitative. Procedures based on published methods, Sambrook et al., 1989, Molecular cloning: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, will be used to derive riboprobes. Briefly, the Kb cDNA will be cloned into a plasmid vector containing the T3 and T7 RNA polymerase promoter sequences (bluescript or Bluescribe plasmids from Stratagene). Using appropriate polymerase and 32P-nucleotides, transcription of the insert will be initiated and the radioactive Kb RNA will be purified. This probe will then be incubated with various RNA preparations followed by treatment with ribonuclease. Presence of RNA will be assessed by electrophoresis on a sequencing gel.
  • PCR following reverse transcriptase treatment of RNA will be used as a highly sensitive procedure for detecting the Kb transcript. Appropriate primers will be designed in order to specifically amplify retroviral derived transcripts (one primer covering the 5′UT region of the construct and second derived from the cDNA sequence). Briefly, RNA will be prepared by the GuSCN/CsCl method and first strand cDNA will be prepared from 5 ug of total RNA using the SuperScript preamplification system (BRL/Life Technologies, Inc., Gaithersburg, Md.). PCR amplifications will be conducted for 50 cycles, Hansen et al., J. Immunol. 118:1403-1408, using the Cetus GeneAmp kit (Perkin Elmer Cetus, Norwalk, Conn.). Reaction products will be visualized following electrophoresis on a 3% NuSieve agarose gel (FMC BioProducts, Rockland, Me.).
  • Allogeneic MHC Gene Transfer Plus Cyclosporine
  • It has been shown previously in partially inbred miniature swine that differences in class II MHC loci are of critical importance in determining the fate of primarily vascularized allografts. Cyclosporine given early in the post-transplant period uniformly leads to tolerance of class II matched class I mismatched kidney allografts. However, cyclosporine alone does not produce tolerance across a full-MHC barrier. Consistent with the importance of class II allogeneic bone marrow transplantation across class II barriers induces tolerance to kidney transplants matched to the class II of the bone marrow donors, but completely disparate to the recipients. In the following experiment specific transplantation tolerance to complete SLA-disparate kidney transplant was induced with autologous bone marrow transplantation in which the recipient's marrow was genetically modified prior to transplant by transduction with a retroviral expression vector carrying an allogeneic SLA class II gene. The retroviral expression vectors contained cDNA for either SLA-DRBa or -DRBc and a drug selection marker (Neo), and high tittered viral supernatants were prepared using amphotropic packaging cell lines. Bone marrow from five animals included in this study was harvested on day 2, and then cultured with virus-containing supernatant for either an allogeneic (n=4) or syngeneic (n-1) MHC gene for a approximately 48 hours. After lethal irradiation (10 Gy in two fractions 24 hours apart) in days −1 and 0, animals were transplanted with 0.4 to 1.3×108 cells/kg on day 0. The effectiveness of the gene transfer was tested using a colony forming unit assay for granulocyte/macrophage progenitors (CFU-GM) in the presence of G418 to select for neomycin resistance. The frequency of G418 resistant CFU-GM varied significantly between animals (6.5% to 25.9%) immediately after transduction and dimnished slowly with time. All animals regained their responsiveness to allogeneic stimuli by the third month post-bone marrow transplantation, as tested by MLR. mAbs specific for DQ and DR molecules of the class II MHC were used for “blocking” MLR studies, to separate the effects of recognition of DQ and DR. In assays using cells from recipients of bone marrow transplantation transduced with the allogeneic DRB gene, the DR portion of the response to the gene-donor type cells was strongly diminished, demonstrating the effectiveness of the transduced gene at inducing DR-specific unresponsiveness. This effect was observed in all experimental animals, although more pronounced in the DRBd to cc combination than in DRBa to gg direction. In MRL using cells from a control animal transduced with a syngeneic gene, blocking of DR or DQ in MLR showed a pattern of reactivity identical to that observed in naive animals of the same haplotype. Five months after BMT, animals were challenged with kidney transplants matched for class II of the gene-donor type, and fully mismatched to the original recipient haplotype. Cyclosporine 10 to 15 mg/kg/day iv was given for 12 days, to tolerize for the class I MHC and minor antigen disparity. Three animals rejected their kidney transplants at days 8, 22 and 40. The accelerated manner of rejection at day 8 suggested sensitization as an undesirable effect of the expression of the allogeneic gene product. In none of these recipients could the presence of anti-donor type antibodies be detected by flow cytometry. One animal became tolerant and exhibited normal creatinine levels at 101 days post-transplant. The animal which received the bone marrow transduced with a syngeneic gene underwent severe rejection with high creatinine levels and vascular changes in pathology. The recipient of the longest surviving kidney transplant also received the most efficiently transduced autologous bone marrow, as judged on the initial frequency of G418r CFU-GM. In this one case, recombinant cytokines (Pixy 321 (Pixy is a human-GM-CSF/IL3 fusion protein) 100 Units/ml; mouse stem cell growth factor 20 Units/ml; although these cytokines were used, cytokines from the same species as the cell being transformed can also be used) were included in the culture medium during transduction with the allogeneic DRB retroviral expression vector. The cytokines may have led to the transduction of multilineage pluripotent hematopoietic stem cells, including the precursors of dendritic cells which ultimately induced DRB-specific hyporesponsiveness. These experiments demonstrate that somatic class II MHC DRB gene transfer into bone marrow cells has profound functional consequences upon the immune responses of the recipient. In vitro, and more importantly in vivo, immune function was significantly modulated, with the induction of donor specific prolongation of fully mismatched kidney transplant survival. The transduction of allogeneic bone marrow stem cells with MHC genes provides a method of inducing tolerance across MHC barriers by a mechanism comparable to lymphohematopoietic chimerism. The lethal irradiation used in the experiments described herein can be replaced with a non-myeloablative conditioning regimen that would permit bone marrow engraftment in a more clinically acceptable fashion.
  • III. The Induction of Tolerance with Bone Marrow Transplantation
  • A Short Course of High Dose of Cyclosporine (Administered in Absence of Treatments. e.g. Treatment with Prednisone which Stimulate Cytokine Release) to Induce Tolerance to Class I and Other Minor Disparities Combined with Implantation of Bone Marrow Cells Induce Tolerance to Class II Disparity.
  • Xenografts: The following procedure was designed to lengthen the time an implanted organ (a xenograft) survives in a xenogeneic host prior to rejection. The organ can be any organ, e.g., a liver, e.g., a kidney, e.g., a heart. The main strategies are elimination of natural antibodies by organ perfusion, transplantation of tolerance-inducing bone marrow, optionally, the implantation of donor stromal tissue, and, optionally, the administration of a short course of a help reducing agent at about the time of introduction of the graft, as described above preparation of the recipient for transplantation includes any or all of these steps. Preferably they are carried out in the following sequence.
  • First, a preparation of horse anti-human thymocyte globulin (ATG) is intravenously injected into the recipient. The antibody preparation eliminates mature T cells and natural killer cells. If not eliminated, mature T cells would promote rejection of both the bone marrow transplant and, after sensitization, the xenograft itself. Of equal importance, the ATG preparation also eliminates natural killer (NK) cells. NK cells probably have no effect on the implanted organ, but would act immediately to reject the newly introduced bone marrow. Anti-human ATG obtained from any mammalian host can also be used, e.g., ATG produced in pigs, although thus far preparations of pig ATG have been of lower titer than horse-derived ATG. ATG is superior to anti-NK monoclonal Antibodies, as the latter are generally not lytic to all host NK cells, while the polyclonal mixture in ATG is capable of lysing all host NK cells. Anti-NK monoclonal antibodies can, however, be used.
  • The presence of donor antigen in the host thymus during the time when host T cells are regenerating post-transplant is critical for tolerizing host T cells. If donor hematopoietic stem cells are not able to become established in the host thymus and induce tolerance before host T cells regenerate repeated doses of anti-recipient T cell antibodies may be necessary throughout the non-myeloablative regimen. Continuous depletion of host T cells may be required for several weeks. Alternatively, e.g. if this approach is not successful, and tolerance (as measured by donor skin graft acceptance, specific cellular hyporesponsiveness in vitro, and humoral tolerance) is not induced in these animals, the approach can be modified to include host thymectomy. In thymectomized recipients, host T cells do not have an opportunity to differentiate in a host thymus, but must differentiate in the donor thymus. If this is not possible, then the animal has to rely on donor T cells developing in the donor thymus for immunocompetence. Immunocompetence can be measured by the ability to reject a non-donor type allogeneic donor skin graft, and to survive in a pathogen-containing environment.
  • It may also be necessary or desirable to splenectomize the recipient in order to avoid anemia.
  • Second, the recipient is administered low dose radiation in order to make room for newly injected bone marrow cells. A sublethal dose of between 100 rads and 400 rads whole body radiation, plus 700 rads of local thymic radiation, has been found effective for this purpose.
  • Third, natural antibodies are adsorbed from the recipient's blood by hemoperfusion of a liver of the donor species. Pre-formed natural antibodies (nAB) are the primary agents of graft rejection. Natural antibodies bind to xenogeneic endothelial cells and are primarily of the IgM class. These antibodies are independent of any known previous exposure to antigens of the xenogeneic donor. B cells that produce these natural antibodies tend to be T cell-independent, and are normally tolerized to self antigen by exposure to these antigens during development. The mechanism by which newly developing B cells are tolerized is unknown. The liver is a more effective adsorber of natural antibodies than the kidney.
  • The fourth step in the non-myeloablative procedure is to implant donor stromal tissue, preferably obtained from fetal liver, thymus, and/or fetal spleen, into the recipient, preferably in the kidney capsule. Stem cell engraftment and hematopoiesis across disparate species barriers is enhanced by providing a hematopoietic stromal environment from the donor species. The stromal matrix supplies species-specific factors that are required for interactions between hematopoietic cells and their stromal environment, such as hematopoietic growth factors, adhesion molecules, and their ligands.
  • As liver is the major site of hematopoiesis in the fetus, fetal liver can also serve as an alternative to bone marrow as a source of hematopoietic stem cells. The thymus is the major site of T cell maturation. Each organ includes an organ specific stromal matrix that can support differentiation of the respective undifferentiated stem cells implanted into the host. Although adult thymus may be used, fetal tissue obtained sufficiently early in gestation is preferred because it is free from mature T lymphocytes which can cause GVHD. Fetal tissues also tend to survive better than adult tissues when transplanted. As an added precaution against GVHD, thymic stromal tissue can be irradiated prior to transplantation, e.g., irradiated at 1000 rads. As an alternative or an adjunct to implantation, fetal liver cells can be administered in fluid suspension.
  • Fifth, bone marrow cells (BMC), or another source of hematopoietic stem cells, e.g., a fetal liver suspension, of the donor are injected into the recipient. Donor BMC home to appropriate sites of the recipient and grow contiguously with remaining host cells and proliferate, forming a chimeric lymphohematopoietic population. By this process, newly forming B cells (and the antibodies they produce) are exposed to donor antigens, so that the transplant will be recognized as self. Tolerance to the donor is also observed at the T cell level in animals in which hematopoietic stem cell, e.g., BMC, engraftment has been achieved. When an organ graft is placed in such a recipient several months after bone marrow chimerism has been induced, natural antibody against the donor will have disappeared, and the graft should be accepted by both the humoral and the cellular arms of the immune system. This approach has the added advantage of permitting organ transplantation to be performed sufficiently long following transplant of hematopoietic cells, e.g., BMT, e.g., a fetal liver suspension, that normal health and immunocompetence will have been restored at the time of organ transplantation. The use of xenogeneic donors allows the possibility of using bone marrow cells and organs from the same animal, or from genetically matched animals.
  • Finally, a short course of a help reducing agent, e.g., a short course of high dose CsA is administered to the recipient. As is described above, the course is begun at about the time of implantation, or a little before, and is continued for a time about equal to the time it takes for a mature T cell to be stimulated and initiate rejection. While any of these procedures may aid the survival of an implanted organ, best results are achieved when all steps are used in combination. Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • While any of these procedures may aid the survival of an implanted organ, best results are achieved when all steps are used in combination. Methods of the invention can be used to confer tolerance to allogeneic grafts, e.g., wherein both the graft donor and the recipient are humans, and to xenogeneic grafts, e.g., wherein the graft donor is a nonhuman animal, e.g., a swine, e.g., a miniature swine, and the graft recipient is a primate, e.g., a human.
  • In the case of xenogeneic grafts, the donor of the implant and the individual that supplies either the tolerance-inducing hematopoietic cells or the liver to be perfused should be the same individual or should be as closely related as possible. For example, it is preferable to derive implant tissue from a colony of donors that is highly inbred.
  • Detailed Protocol
  • In the following protocol for preparing a cynomolgus monkey for receipt of a kidney from a miniature swine donor, zero time is defined as the moment that the arterial and venous cannulas of the recipient are connected to the liver to be perfused.
  • On day −1 a commercial preparation (Upjohn) of horse anti-human anti-thymocyte globulin (ATG) is injected into the recipient. ATG eliminates mature T cells and natural killer cells that would otherwise cause rejection of the bone marrow cells used to induce tolerance. The recipient is anesthetized, an IV catheter is inserted into the recipient, and 6 ml of heparinized whole blood are removed before infection. The ATG preparation is then injected (50 mg/kg) intravenously. Six ml samples of heparinized whole blood are drawn for testing at time points of 30 min., 24 hours and 48 hours. Blood samples are analyzed for the effect of antibody treatment on natural killer cell activity (testing on K562 targets) and by FACS analysis for lymphocyte subpopulations, including CD4, CD8, CD3, CDllb, and CD16. Preliminary data from both assays indicate that both groups of cells are eliminated by the administration of ATG. If mature T cells and NK cells are not eliminated, ATG can be re-administered at later times in the procedure, both before and after organ transplantation.
  • Sublethal irradiation is administered to the recipient between days −1 and −8. Irradiation is necessary to eliminate enough of the recipient's endogenous BMC to stimulate hematopoiesis of the newly introduced foreign BMC. Sublethal total body irradiation is sufficient to permit engraftment with minimal toxic effects to the recipient. Whole body radiation (150 Rads) was administered to cynomolgus monkey recipients from a bilateral (TRBC) cobalt teletherapy unit at 10 Rads/min. Local irradiation of the thymus (700 Rads) was also employed in order to facilitate engraftment.
  • Natural antibodies are a primary cause of organ rejection. To remove natural antibodies from the recipient's circulation prior to transplantation, on day 0 an operative adsorption of natural antibodies (nAB) is performed, using a miniature swine liver, as follows. At −90 minutes the swine donor is anesthetized, And the liver prepared for removal by standard operative procedures. At −60 minutes the recipient monkey is anesthetized. A peripheral IV catheter is inserted, and a 6 ml sample of whole blood is drawn. Through mid-line incision, the abdominal aorta and the vena cava are isolated. Silastic cannulas containing side ports for blood sampling are inserted into the blood vessels.
  • At −30 minutes the liver is perfused in situ until it turns pale, and then removed from the swine donor and placed into cold Ringers Lactate. The liver is kept cold until just prior to reperfusion in the monkey. A liver biopsy is taken. At −10 minutes the liver is perfused with warm albumin solution until the liver is warm (37 degrees).
  • At 0 time the arterial and venous cannulas of the recipient are connected to the portal vein and vena cava of the donor liver and perfusion is begun. Liver biopsies are taken at 30 minutes and 60 minutes, respectively. Samples of recipient blood are also drawn for serum at 30 minutes and 60 minutes respectively. At 60 minutes the liver is disconnected from the cannulas and the recipient's large blood vessels are repaired. The liver, having served its function of adsorbing harmful natural antibodies from the recipient monkey, is discarded. Additional blood samples for serum are drawn from the recipient at 2, 24, and 48 hours. When this procedure was performed on two sequential perfusions of swine livers, the second liver showed no evidence of mild ischemic changes during perfusion. At the end of a 30 minute perfusion the second liver looked grossly normal and appeared to be functioning, as evidenced by darkening of the venous outflow blood compared to the arterial inflow blood in the two adjacent cannulas. Tissue sections from the livers were normal, but immunofluorescent stains showed IgM on endothelial cells. Serum samples showed a decrease in natural antibodies.
  • To promote long-term survival of the implanted organ through T-cell and B-cell mediated tolerance, donor bone marrow cells are administered to the recipient to form chimeric bone marrow. The presence of donor antigens in the bone marrow allows newly developing B cells, and newly sensitized T cells, to recognize antigens of the donor as self, and thereby induces tolerance for the implanted organ from the donor. To stabilize the donor BMC, donor stromal tissue, in the form of tissue slices of fetal liver, thymus, and/or fetal spleen are transplanted under the kidney capsule of the recipient. Stromal tissue is preferably implanted simultaneously with, or prior to, administration of hematopoietic stem cells, e.g., BMC, or a fetal liver cell suspension.
  • To follow chimerism, two color flow cytometry can be used. This assay uses monoclonal antibodies to distinguish between donor class I major histocompatibility antigens and leukocyte common antigens versus recipient class I major histocompatibility antigens. BMC can in turn be injected either simultaneously with, or preceding, organ transplant. Bone marrow is harvested and injected intravenously (7.5×108/kg) as previously described (Pennington et al., 1988, Transplantation 45:21-26). Should natural antibodies be found to recur before tolerance is induced, and should these antibodies cause damage to the graft, the protocol can be modified to permit sufficient time following BMT for humoral tolerance to be established prior to organ grafting.
  • The approaches described above are designed to synergistically prevent the problem of transplant rejection. When a kidney is implanted into a cynomolgus monkey following liver adsorption of natural antibodies, without use of bone marrow transplantation to induce tolerance, renal functions continued for 1-2 days before rejection of the kidney. When four steps of the procedure were performed (adsorption of natural antibodies by liver perfusion, administration of ATG, sublethal irradiation and bone marrow infusion, followed by implant of a porcine kidney into primate recipient), the kidney survived 7 days before rejection. Despite rejection of the transplanted organ, the recipient remained healthy.
  • When swine fetal liver and thymic stromal tissue were implanted under the kidney capsule of two sublethally irradiated SCID mice, 25-50% of peripheral blood leukocytes were of donor lineage two weeks post-transplantation. A significant degree of chimerism was not detected in a third animal receiving fetal liver without thymus.
  • The methods of the invention may be employed in combination, as described, or in part.
  • The method of introducing bone marrow cells may be altered, particularly by (1) increasing the time interval between injecting hematopoietic stem cells and implanting the graft; (2) increasing or decreasing the amount of hematopoietic stem cells injected; (3) varying the number of hematopoietic stem cell injections; (4) varying the method of delivery of hematopoietic stem cells; (5) varying the tissue source of hematopoietic stem cells, e.g., a fetal liver cell suspension may be used; or (6) varying the donor source of hematopoietic stem, cells. Although hematopoietic stem cells derived from the graft donor are preferable, hematopoietic stem cells may be obtained from other individuals or species, or from genetically-engineered inbred donor strains, or from in vitro cell culture.
  • Methods of preparing the recipient for transplant of hematopoietic stem cells may be varied. For instance, recipient may undergo a splenectomy or a thymectomy. The latter would preferably be administered prior to the non-myeloablative regimen, e.g., at day −14.
  • Hemoperfusion of natural antibodies may: (1) make use of other vascular organs, e.g., liver, kidney, intestines; (2) make use of multiple sequential organs; (3) vary the length of time each organ is perfused; (4) vary the donor of the perfused organ. Irradiation of the recipient may make use of: (1) varying the absorbed dose of whole body radiation below the sublethal range; (2) targeting different body parts (e.g., thymus, spleen); (3) varying the rate of irradiation (e.g., 10 rads/min, 15 rads/min); or (4) varying the time interval between irradiation and transplant of hematopoietic stem cells; any time interval between 1 and 14 days can be used, and certain advantages may flow from use of a time interval of 4-7 days. Antibodies introduced prior to hematopoietic cell transplant may be varied by: (1) using monoclonal antibodies to T cell subsets or NK cells (e.g., anti-NKH1A, as described by U.S. Pat. No. 4,772,552 to Hercend, et al., hereby incorporated by reference); (2) preparing anti-human ATG in other mammalian hosts (e.g., monkey, pig, rabbit, dog); or (3) using anti-monkey ATG prepared in any of the above mentioned hosts.
  • The methods of the invention may be employed with other mammalian recipients (e.g., rhesus monkeys) and may use other mammalian donors (e.g., primates, sheep, or dogs). As an alternative or adjunct to hemoperfusion, host antibodies can be depleted by administration of an excess of hematopoietic cells.
  • Stromal tissue introduced prior to hematopoietic cell transplant, e.g., BMT, may be varied by: (1) administering the fetal liver and thymus tissue as a fluid cell suspension; (2) administering fetal liver or thymus stromal tissue but not both; (3) placing a stromal implant into other encapsulated, well-vascularized sites, or (4) using adult thymus or fetal spleen as a source of stromal tissue.
  • Tolerance to fully MHC mismatched renal allografts in chimeric swine
  • Overwhelming importance of major histocompatibility complex (MHC) class II matching for achieving tolerance of kidney transplants (KTx) in miniature swine has been demonstrated previously. When class II antigens are matched, long-term specific tolerance across MHC class I and minor antigens (MA) barrier, can uniformly be induced by a short course of cyclosporine. However, cyclosporine does not produce this effect across a full MHC barrier. Bone marrow transplantation (BMT) across single-haplotype class II MHC+MA barriers creates fully chimeric animals, as confirmed by FCM. These chimeras recover normal cellular immune function 2-3 months after BMT, as tested by MLR and CML. Four such chimeric animals (see Table V, numbers 1-4) received kidney transplants from donors class II matched to BMT donors and fully mismatched to the recipients. A 12-day course of cyclosporine (10 mg/kg/day) was the only immunosuppression following kidney transplantation. All 4 pigs have maintained normal creatinine (Cr) values (<2 mg %) for longer than 300 days, and one recipient is alive over 3 years with good kidney function (Cr<2 mg %) and graft histology showing minimal borderline rejection. These results demonstrate that induction of tolerance to class II antigens by BMT allows a short course of cyclosporine to induce specific tolerance (as tested by skin grafts) to fully allogeneic kidney transplants. Subsequently, we have examined the specificity of this phenomenon by determining if single-haplotype class II+MA mismatched BMT will facilitate cyclosporine induced long-term acceptance of kidney transplants completely mismatched to both the recipient and BMT donor (Table V, numbers 5-10). A 12-day course of cyclosporine allowed long-term survival of such kidney transplants in chimeric recipients. Animal #5 was still alive and clinically well, with normal Cr levels; histology however reveals borderline rejection. Animal #6 was sacrificed 18 months after kidney transplant, with deteriorating kidney function (Cr>11 mg %). Animal #7 was sacrificed at 6 months after kidney transplant due to sepsis, kidney transplants showed moderate tubulointestinal infiltrate without signs of vascular injury. Both long-term survivors (pigs #3 & 5) were recently tested for anti-donor reactivity. CML and MLR revealed specific unresponsiveness to the kidney transplant donor type cells. Pigs #8-10 received kidney transplant from outbred Yorkshire donors. These animals developed irreversible renal failure, starting shortly after cessation of the cyclosporine therapy.
    TABLE V
    # Recipient BMT Donor KTx Donor Outcome (funct./pathol.)
    1 aa (IaaIIaa) aj (IaaIIac) cc (IccIcc) sac 1 y (good/normal)
    2 ac (IacIIac) ag (IacIIad) dd (IddIIdd) died >2.5 y (good/
    chronic rej)
    3 ac (IacIIac) ag (IacIIad) dd (IddIIdd) alive >3 y (good/border
    rej)
    4 ac (IacIIac) ag (IacIIad) dd (IddIIdd) sac 1 y (good/normal)
    5 aa (IaaIIaa) ah (IaaIIad) cc (IccIIcc) alive >2.5 y (good/border
    rej)
    6 aa (IaaIIaa) ah (IaaIIad) cc (IccIIcc) sac >1.5 y (poor/chronic
    rej)
    7 aa (IaaIIaa) aj (IaaIIac) dd (IddIIdd) sac 0.5 y (good/infiltrate)
    8 aa (IaaIIaa) aj (IaaIIac) YORK (I?II?) sac 30 d (poor/acute rej)
    9 ac (IacIIac) ch (IacIIad) YORK (I?II?) sac 70 d (poor/acute rej)
    10 ac (IacIIac) ch (IacIIad) YORK (I?II?) sac 38 d (poor/acute rej)

    sac = sacrificed;

    rej = rejection
  • Thus, a short postoperative course of cyclosporine in MHC class II mismatched BMT recipients allows tolerance to be induced to kidney transplants that are class II matched to the BMT donor. Long-term unresponsiveness to kidney transplants that are fully mismatched to both the recipient and BMT donor can be achieved in some cases, apparently dependent on the degree of disparity at multiple loci (compare with the difference between inbred and outbred donors).
  • A Short Course of Cyclosporine to Suppress T Cell Function in Primate Allogeneic Kidney Transplantation.
  • The following experiment shows that mixed chimerism, obtained during a non-myeloablative protocol to achieve engraftment, is capable of producing multilineage lymphohematopoietic chimerism and long-term tolerance to renal allografts between fully MHC mismatched cynomolgus monkeys. Complete ablation of host lymphohematopoietic elements is neither necessary nor desirable when bone marrow transplantation is utilized as a tolerance-inducing regimen. Instead, it is advantageous to achieve a state of mixed chimerism, in which the presence of certain donor-derived elements induce specific tolerance, while host-type antigen presenting cells maintain normal immunocompetence.
  • It has been demonstrated in murine studies that removal of mature host T cells is important in order to achieve mixed chimerism. In initial studies using fully MHC mismatched cynomolgus monkeys, a variety of monoclonal antibodies were tested to mature T cell subsets (anti-CD4 and anti-CD8) as well as several sources of anti-thymocyte globulin (ATG) as T cell depleting reagents. Although these antibody treatments led to marked depletion of T cells in the peripheral blood, biopsies of lymph nodes demonstrated that residual T cells remained, often coated with antibody. In order to further suppress T cell function, a one-month course of treatment with an i.m. preparation of cyclosporine(CyA) in oil was added to the preparative regimen. This treatment led to therapeutic levels of cyclosporine during drug administration and to tapering levels over a period of 3 weeks after the drug was discontinued. The basic protocol for nonlethal preparative regimen was as follows: Cynomolgus monkeys weighing 6 to 10 kg. (Charles River Primates, Wilmington, Mass.) were treated with 300 Rads of WBI either as a single dose (#M393) on day −6 or as two fractions of 150 Rads each on days −6 and −5 (#M3093 and #M3293). 700 Rads of thymic irradiation was administered on day −1. Horse anti-human thymocyte globulin (ATG) (Upjohn) was administered at 50 mg/kg i.m. on days −2, −1 and 0. Orthotopic kidney transplantation was performed on day 0 through a midline incision using end to side anastamoses of the donor renal artery and renal vein into the recipient aorta and vena cava, respectively, and using a ureteroureteral anastomosis for urinary drainage. Bone marrow was harvested from two donor ribs, prepared as a single cell suspension, and infused i.v. into the recipient at the end of the renal transplant. Treatment with cyclosporine (Sandimmune®, 15 mg/kg/day, suspended in olive oil) i.m. was begun on day 0 and continued for 27 days.
  • Monkey #393 became pancytopenic on day 8, and required three blood transfusions with blood group matched, irradiated whole blood over the next two weeks. However, peripheral blood components recovered gradually thereafter, and were normal by day 30. Renal function has remained normal for over 250 days, and a biopsy on day 215 showed a normal kidney.
  • Sequential flow cytometric (FCM) analyses were performed on this animal utilizing a monoclonal anti-class I antibody previously determined to distinguish donor from host, and analyzing lymphoid, monocytic and neutrophil populations as determined by scatter profiles. Clear evidence for chimerism in all three subpopulations was detected first on day 10, and persisted at similarly high levels until cyclosporine treatment was discontinued on day 27. Thereafter, the levels of chimerism detected in each subpopulation decreased, but chimerism was still detectable by FCM among lymphocytes (1.5%) and monocytes (29%) as late as day 203, the last day tested. In addition, a bone marrow aspirate on day 203 showed 11.2% donor cells by FCM.
  • Mixed lymphocyte reactions performed pre-transplant and on day 159 post-transplant revealed a specific loss of anti-donor reactivity (Table VI).
    TABLE VI
    3rd
    Time Medium Autologous Donor Party # 1 3rd Party #2
    Pre- 888 2434 5946 5571 6986
    Transplant
    (CPM)
    Pre- 1.0 3.3 3.0 3.9
    Transplant
    (Stim. Index)
    Day 159 703 3410 2324 11298 9127
    (CPM)
    Day 159 1.0 0.6 4.2 3.1
    (Stim. Index)
  • This result, combined with normal renal function and normal kidney histology without any additional exogenous immunosuppression since day 27, lead us to conclude that specific transplantation tolerance has been induced in this animal through the establishment of mixed chimerism. Two additional animals were treated by the same protocol, but with an intravenous preparation of cyclosporine which led to an abrupt fall of cyclosporine levels in the blood after discontinuation rather than gradual tapering of levels over a three-week period. One of these animals died of sepsis on day 12 during the period of aplasia, and the other lost evidence for chimerism after discontinuation of cyclosporine and although still alive on day 100, has shown a course consistent with chronic rejection both by clinical and pathological criteria.
  • In order to reduce the toxicity of the preparative regimen, we have subsequently modified the irradiation protocol. In one animal (#3893) the WBI was decreased to 1.5 Gy. This animal failed to develop mixed chimerism and rejected the kidney transplant (Creatinine=12.1 on day 47). In two additional animals (#3093 and #3292) the WBI was maintained at 3.0 Gy, but was fractionated to 1.5 Gy on two successive days (−6 and −5) rather than administered as a single dose. Both of these animals developed mixed multilineage chimerism, first detectable on day 11 and day 20 respectively. They showed much less toxicity from the preparative regimen than did the animals receiving unfractionated irradiation, and both remain chimeric with normal renal function at the time of this writing (day 40 and day 25, respectively).
  • Pig to Monkey Kidney Xenotransplantation by a Mixed Chimerism Approach
  • The following experiment shows the induction of tolerance in monkeys to pig organs by means of a xenogeneic lymphohematopoietic chimerism approach which has previously been shown effective in concordant rodent systems. To date 16 Cynomolgus monkeys have received pig kidney transplants along with xenogeneic bone marrow from the same donor. The preparative regimen for these xenografts included: 1) conditioning with non-myeloablative whole body irradiation (WBI) and thymic irradiation; 2) removal of preformed mAbs by perfusion of monkey blood through a pig liver; 3) splenectomy; 4) T cell depletion with ATG and/or mAbs; and 5) postoperative immunosuppression with cyclosporine and in some animals anti-IgM mAbs. Ten animals have survived more than 4 days, with the longest surviving 13 days, with normal renal function to day 11. In this animal pig cells were detected in the peripheral blood only at day 10 post-transplant, suggesting transient xenogeneic chimerism. Two monkeys received only splenectomy and pig liver perfusion prior to the kidney xenograft. In one of these animals, in which no further immunosuppression was administered post-transplant, the kidney functioned for 3 days, then rapidly lost function, with complete rejection by day 5. Analysis of this monkey's sera by flow cytometry indicated return of high titers of IgM, which correlated with rejection. In the second animal cyclosporine 15 mg/kg/day iv and 15 deoxyspergualin (DSG) 6 mg/kg/day iv were administered post-transplant. The kidney functioned until day 7, then failed and was removed on day 8. Pathologic examination showed a focal inflammatory infiltrate in addition to patchy interstitial hemorrhage. The infiltrate contained approximately 20% T cells, as determined by staining with mAbs to CD3, CD4 and CD8. IgM natural antibodies were effectively removed during liver perfusion in this animal, and strikingly, they did not appear in the serum thereafter, IgG levels started to rise on day 7, correlating with the beginning of renal dysfunction. These results show 1) that natural antibody (IgM) responses can be effectively eliminated by components of the preparative regimen involving pig liver adsorption and post-operative suppression with DSG; and 2) that T cell suppressive components of the preparative regimen (i.e., irradiation, cyclosporine and ATG) are required to prevent cellular and secondary (IgG) responses in these experiments.
  • Other Embodiments
  • Stromal tissue introduced prior to hematopoietic cell transplant, e.g., BMT, may be varied by: (1) administering the fetal liver and thymus tissue as a fluid cell suspension; (2) administering fetal liver or thymus stromal tissue but not both; (3) placing a stromal implant into other encapsulated, well-vascularized sites, or (4) using adult thymus or fetal spleen as a source of stromal tissue.
  • The methods described herein for inducing tolerance to, or promoting the acceptance of, an allogeneic antigen or allogeneic graft can be used where, as between the donor and recipient, there is any degree of mismatch at MHC loci or other loci which influence graft rejection. Preferably, there is a mismatch at least one MHC locus or at least one other locus that mediates recognition and rejection, e.g., a minor antigen locus. With respect to class I and class II MHC loci, the donor and recipient can be: matched at class I and mismatched at class II; mismatched at class I and matched at class II; mismatched at class I and mismatched at class II; matched at class I, matched at class II. In any of these combinations other loci which control recognition and rejection, e.g., minor antigen loci, can be matched or mismatched. As stated above, it is preferable that there is mismatch at least one locus. Mismatched at MHC class I means mismatched for one or more MHC class I loci, e.g., in the case of humans, mismatched at one or more of HLA-A, HLA-B, or HLA-C, or in the case of swine, mismatch at one or more SLA class I loci, e.g., the swine A or B loci. Mismatched at MHC class II means mismatched at one or more MHC class II loci, e.g., in the case of humans, mismatched at one or more of a DP α, a DPβ, a DQ α, a DQ β, a DR α, or a DR β, or in the case of swine, mismatch at one or SLA class II loci, e.g., mismatch at DQ α or β, or DR α or β.
  • The methods described herein for inducing tolerance to an allogeneic antigen or allogeneic graft can be used where, as between the donor and recipient, there is any degree of reactivity in a mixed lymphocyte assay, e.g., wherein there is no, low, intermediate, or high mixed lymphocyte reactivity between the donor and the recipient. In preferred embodiments mixed lymphocyte reactivity is used to define mismatch for class II, and the invention includes methods for performing allogeneic grafts between individuals with any degree of mismatch at class II as defined by a mixed lymphocyte assay. Serological tests can be used to determine mismatch at class I or II loci and the invention includes methods for performing allogeneic grafts between individuals with any degree of mismatch at class I and or II as measured with serological methods. In a preferred embodiment, the invention features methods for performing allogeneic grafts between individuals which, as determined by serological and or mixed lymphocyte reactivity assay, are mismatched at both class I and class II.
  • The methods of the invention are particularly useful for replacing a tissue or organ afflicted with a neoplastic disorder, particularly a disorder which is resistant to normal modes of therapy, e.g., chemotherapy or radiation therapy. Methods of the invention can be used for inducing tolerance to a graft, e.g., an allograft, e.g., an allograft from a donor which is mismatched at one or more class I loci at one or more class II loci or at one or more loci at each of class I and class II. In preferred embodiments: the graft includes tissue from the digestive tract or gut, e.g., tissue from the stomach, or bowel tissue, e.g., small intestine, large intestine, or colon; the graft replaces a portion of the recipient's digestive system e.g., all or part of any of the digestive tract or gut, e.g., the stomach, bowel, e.g., small intestine, large intestine, or colon.
  • Tolerance, as used herein, refers not only to complete immunologic tolerance to an antigen, but to partial immunologic tolerance, i.e., a degree of tolerance to an antigen which is greater than what would be seen if a method of the invention were not employed.
  • As is discussed herein, it is often desirable to expose a graft recipient to irradiation in order to promote the development of mixed chimerism. The inventor has discovered that it is possible to induce mixed chimerism with less radiation toxicity by fractionating the radiation dose, i.e., by delivering the radiation in two or more exposures or sessions. Accordingly, in any method of the invention calling for the irradiation of a recipient, e.g., a primate, e.g., a human, recipient, of a xenograft or allograft, the radiation can either be delivered in a single exposure, or more preferably, can be fractionated into two or more exposures or sessions. The sum of the fractionated dosages is preferably equal, e.g., in rads or Gy, to the radiation dosage which can result in mixed chimerism when given in a single exposure. The fractions are preferably approximately equal in dosage. For example, a single dose of 700 rads can be replaced with, e.g., two fractions of 350 rads, or seven fractions of 100 rads. Hyperfractionation of the radiation dose can also be used in methods of the invention. The fractions can be delivered on the same day, or can be separated by intervals of one, two, three, four, five, or more days. Whole body irradiation, thymic irradiation, or both, can be fractionated.
  • The inventor has also discovered that much or all of the preparative regimen can be delivered or administered to a recipient, e.g., an allograft or xenograft recipient, within a few days, preferably within 72, 48, or 24 hours, of transplantation of tolerizing stem cells and/or the graft. This is particularly useful in the case of humans receiving grafts from cadavers. Accordingly, in any of the methods of the invention calling for the administration of treatments prior to the transplant of stem cells and/or a graft, e.g., treatments to inactivate or deplete host antibodies, treatments to inactivate host T cells or NK cells, or irradiation, the treatment(s) can be administered, within a few days, preferably within 72, 48, or 24 hours, of transplantation of the stem cells and/or the graft. In particular, primate, e.g., human, recipients of allografts can be given any or all of treatments to inactivate or deplete host antibodies, treatments to inactivate host T cells or NK cells, or irradiation, within a few days, preferably within 72, 48, or 24 hours, of transplantation of stem cells and/or the graft. For example, treatment to deplete recipient T cells and/or NK cells, e.g., administration of ATG, can be given on day −2, −1, and 0, and WBI, thymic irradiation, and stem cell, e.g., bone marrow stem cells, administered on day 0. (The graft, e.g., a renal allograft, is transplanted on day 0).
  • Methods of the invention can include recipient splenectomy.
  • As is discussed herein, hemoperfusion, e.g., hemoperfusion with a donor organ, can be used to deplete the host of natural antibodies. Other methods for depleting or otherwise inactivating natural antibodies can be used with any of the methods described herein. For example, drugs which deplete or inactivate natural antibodies, e.g., deoxyspergualin (DSG) (Bristol), or anti-IgM antibodies, can be administered to the recipient of an allograft or a xenograft. One or more of, DSG (or similar drugs), anti-IgM antibodies, and hemoperfusion, can be used to deplete or otherwise inactivate recipient natural antibodies in-methods of the invention. DSG at a concentration of 6 mg/kg/day, i.v., has been found useful in suppressing natural antibody function in pig to cynomolgus kidney transplants.
  • Some of the methods described herein use lethal irradiation to create hematopoietic space, and thereby prepare a recipient for the administration of allogeneic, xenogeneic, syngeneic, or genetically engineered autologous, stem cells. In any of the methods described herein, particularly primate or clinical methods, it is preferable to create hematopoietic space for the administration of such cells by non-lethal means, e.g., by administering sub-lethal doses of irradiation, bone marrow depleting drugs, or antibodies. The use of sublethal levels of bone marrow depletion allows the generation of mixed chimerism in the recipient. Mixed chimerism is generally preferable to total or lethal ablation of the recipient bone marrow followed by complete reconstitution of the recipient with administered stem cells.
  • Alternative methods for the inactivation of thymic T cells are also included in embodiments of the invention. Some of the methods described herein include the administration of thymic irradiation to inactivate host thymic-T cells or to otherwise diminish the host's thymic-T cell mediated responses to donor antigens. It has been discovered that the thymic irradiation called for in allogeneic or xenogeneic methods of the invention can be supplemented with, or replaced by, other treatments which diminish (e.g., by depleting thymic-T cells and/or down modulating one or more of the T cell receptor (TCR), CD4 co-receptor, or CD8 co-receptor) the host's thymic-T cell mediated response. For example, thymic irradiation can be supplemented with, or replaced by, anti-T cell antibodies (e.g., anti-CD4 and/or anti-CD8 monoclonal antibodies) administered a sufficient number of times, in sufficient dosage, for a sufficient period of time, to diminish the host's thymic-T cell mediated response.
  • For best results, anti-T cell antibodies should be administered repeatedly. E.g., anti-T cell antibodies can be administered one, two, three, or more times prior to donor bone marrow transplantation. Typically, a pre-bone marrow transplantation dose of antibodies will be given to the patient about 5 days prior to bone marrow transplantation. Additional, earlier doses 6, 7, or 8 days prior to bone marrow transplantation can also be given. It may be desirable to administer a first treatment then to repeat pre-bone marrow administrations every 1-5 days until the patient shows excess antibodies in the serum and about 99% depletion of peripheral T cells and then to perform the bone marrow transplantation. Anti-T cell antibodies can also be administered one, two, three, or more times after donor bone marrow transplantation. Typically, a post-bone marrow transplant treatment will be given about 2-14 days after bone marrow transplantation. The post bone marrow administration can be repeated as many times as needed. If more than one administration is given the administrations can be spaced about 1 week apart. Additional doses can be given if the patient appears to undergo early or unwanted T cell recovery. Preferably, anti-T cell antibodies are administered at least once (and preferably two, three, or more times) prior to donor bone marrow transplantation and at least once (and preferably two, three, or more times) after donor bone marrow transplantation.
  • The following experiments show that additional T cell-depleting antibodies can replace thymic irradiation in a non-myeloablative conditioning regimen and allow allogeneic bone marrow engraftment and donor-specific tolerance induction.
  • A low toxicity, non-myeloablative conditioning regimen that allows allogeneic bone marrow engraftment and donor-specific tolerance induction in mice has been previously described. A regimen which includes pre-treatment with depleting doses of anti-CD4 and anti-CD8 monoclonal antibodies on day −5, administration of 3 Gy whole body irradiation and 7 Gy of thymic irradiation on day 0 followed by administration of fully MHC-mismatched donor bone marrow cells, allows the induction of permanent mixed chimerism and skin graft tolerance. The thymic irradiation step in this protocol was replaced with additional anti-CD4 and anti-CD8 monoclonal antibody treatment. Multilineage chimerism was compared in B10 (H-2b) mice receiving allogeneic (B10.A, H-2a) bone marrow transplantation on day 0 following 3 Gy whole body irradiation with or without thymic irradiation, and treatment with monoclonal antibodies by a variety of schedules pre and post-bone marrow transplantation. Most (50 of 52) animals that either received thymic irradiation or that received at least two pre-bone marrow transplantation monoclonal antibody treatments demonstrated long-term multilineage peripheral blood mixed allogeneic chimerism (as demonstrated by flow cytometric analysis). In contrast, only 1 of 8 animals receiving only one pre-bone marrow transplantation monoclonal antibody treatment without thymic irradiation developed lasting (more than 20 weeks) mixed chimerism. AU chimeric animals accepted donor skin grafts for more than 100 days and rejected third party BALB/c grafts within 14 days. Therefore, mixed chimerism and donor-specific skin graft acceptance could be induced without the use of thymic irradiation if at least 2 pre-bone marrow transplantation monoclonal antibody treatments were given. (The monoclonal antibody treatments were spaced about 5 days apart with the final treatment 1 day prior to bone marrow transplantation.) However, levels of donor T cell reconstitution were highest in animals receiving thymic irradiation or receiving additional anti-T cell monoclonal antibody treatments following bone marrow transplantation. Eleven of 20 mice receiving two pre-bone marrow transplantation monoclonal antibody treatments (the monoclonal antibody treatments were spaced about 5 days apart with the final treatment 1 day prior to bone marrow transplantation) and no thymic irradiation showed relatively low levels of donor T cell reconstitution (less than 20% donor, more than 80% host) at 6 weeks, and 9 of these showed a marked loss of donor cells in all lineages by 20 weeks. In contrast, 12 of 12 similarly-treated mice receiving 1 or 2 additional post-bone marrow transplantation monoclonal antibody treatments (the monoclonal antibody treatments were spaced about 7 days apart with the first treatment 7 day after bone marrow transplantation) showed high levels of donor T cell reconstitution at 6 weeks (mean 86±12% donor), and high levels of donor reconstitution persisted in all lineages at 20 weeks. Thus, a second dose of pre-bone marrow transplantation T cell-depleting monoclonal antibodies can replace thymic irradiation and allow tolerance induction in our regimen, but additional monoclonal antibodies administered at one and two weeks post-bone marrow transplantation may increase the ability to reliably induce durable mixed chimerism and tolerance. The capacity of repeated anti-T cell monoclonal antibody treatments to replace thymic irradiation in this regimen most likely reflects their ability to deplete host thymocytes that escape depletion by the initial monoclonal antibody treatment. These monoclonal antibodies deplete most host T cells and induce down-modulation of both TCR and CD4 and CD8 co-receptors on the few remaining cells. In these animals, early migration of donor bone marrow-derived cells to the host thymus is associated with complete clonal deletion of mature host-type thymocytes with TCR that recognize donor antigens. Although a small population of host T cells with such TCR persists in the spleens of chimeras, these cells are anergic to stimulation through their TCR. These cells may have escaped depletion by down-modulating CD4 or CD8 after monoclonal antibody treatment. Thus, this relatively non-toxic regimen achieves pluripotent hematopoietic stem cell engraftment and specific tolerance by ablating most of the existing T cell repertoire and allowing new T cell development in the presence of intrathymic donor antigen, and by inducing anergy among the few remaining host T cells in the periphery.
  • Some of the methods herein include the administration of hematopoietic stem cells to a recipient. In many of those methods, hematopoietic stem cells are administered prior to or at the time of the implantation of a graft (an allograft or a xenograft), the primary purpose of the administration of hematopoietic stem cells being the induction of tolerance to the graft. The inventors have found that one or more subsequent administrations (e.g., a second, third, fourth, fifth, or further subsequent administration) of hematopoietic stem cells can be desirable in the creation and/or maintenance of tolerance. Thus, the invention also includes methods in which hematopoietic stem cells are administered to a recipient, e.g., a primate, e.g., a human, which has previously been administered hematopoietic stem cells as part of any of the methods referred to herein.
  • While not wishing to be bound by theory the inventor believes that repeated stem cell administration may promote chimerism and possibly long-term deletional tolerance in graft recipients. Accordingly, any method referred to herein which includes the administration of hematopoietic stem cells can further include multiple administrations of stem cells. In preferred embodiments: a first and a second administration of stem cells are provided prior to the implantation of a graft; a first administration of stem cells is provided prior to the implantation of a graft and a second administration of stem cells is provided at the time of implantation of the graft. In other preferred embodiments: a first administration of stem cells is provided prior to or at the time of implantation of a graft and a second administration of stem cells is provided subsequent to the implantation of a graft. The period between administrations of hematopoietic stem cells can be varied. In preferred embodiments a subsequent administration of hematopoietic stem cell is provided: at least two days, one week, one month, or six months after the previous administration of stem cells; at least two days, one week, one month, or six months after the implantation of the graft.
  • The method can further include the step of administering a second or subsequent dose of hematopoietic stem cells: when the recipient begins to show signs of rejection, e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte response to donor antigen; when the level of chimerism decreases; when the level of chimerism falls below a predetermined value; when the level of chimerism reaches or falls below a level where staining with a monoclonal antibody specific for a donor PBMC antigen is equal to or falls below staining with an isotype control which does not bind to PBMC's, e.g. when the donor specific monoclonal stains less than 1-2% of the cells; or generally, as is needed to maintain tolerance or otherwise prolong the acceptance of a graft. Thus, method of the invention can be modified to include a further step of determining if a subject which has received a one or more administrations of hematopoietic stem cells is in need of a subsequent administration of hematopoietic stem cells, and if so, administering a subsequent dose of hematopoietic stem cells to the recipient.
  • Any of the methods referred to herein can include the administration of agents, e.g., 15-deoxyspergualin, mycophenolate mofetil, brequinar sodium, or similar agents, which inhibit the production, levels, or activity of antibodies in the recipient. One or more of these agents can be administered: prior to the implantation of donor tissue, e.g., one, two, or three days, or one, two, or three weeks before implantation of donor tissue; at the time of implantation of donor tissue; or after implantation of donor tissue, e.g., one, two, or three days, or one, two or three weeks after, implantation of a graft.
  • The administration of the agent can be initiated: when the recipient begins to show signs of rejection, e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte response to donor antigen; when the level of chimerism decreases; when the level of chimerism falls below a predetermined value; when the level of chimerism reaches or falls below a level where staining with a monoclonal antibody specific for a donor PBMC antigen is equal to or falls below staining with an isotype control which does not bind to PBMC's, e.g. when the donor specific monoclonal stains less than 1-2% of the cells; or generally, as is needed to maintain tolerance or otherwise prolong the acceptance of a graft.
  • The period over which the agent is administered (or the period over which clinically effective levels are maintained in the subject) can be long term, e.g., for six months or more or a year or more, or short term, e.g., for less than a year, more preferably six months or less, more preferably one month or less, and more preferably two weeks or less. The period will generally be at least about one week and preferably at least about two weeks in duration. In preferred embodiments the period is two or three weeks long.
  • Preferred embodiments include administration of 15-deoxyspergualin (6 mg/kg/day) for about two weeks beginning on the day of graft implantation.
  • Some of the methods referred to herein include steps in which antibodies, e.g., preformed natural antibodies, are removed from the blood of a recipient. For example, in some methods antibodies are removed by hemoperfusion of an organ from the donor species. The inventor has discovered that an α1-3 galactose linkage epitope-affinity matrix, e.g., in the form of an affinity column, is useful for removing antibodies from the recipient's blood. Accordingly, the use of an α 1-3 galactose linkage epitope-affinity matrix, e.g., matrix bound linear B type VI carbohydrate, can be added to any method referred to herein and can be used in addition to or in place of any antibody perfusion or removal technique, e.g., organ perfusion, in any method referred to herein.
  • Some of the methods referred to herein include the administration of hematopoietic stem cells to a recipient. In many of those methods hematopoietic stem cells are administered prior to or at the time of the administration of a graft (an allograft or a xenograft), the primary purpose of the administration of hematopoietic stem cells being the induction of tolerance to the graft. The inventors have found that administration of one or more cytokines, preferably a cytokine from the species from which the stem cells are derived, can promote tolerance or otherwise prolong acceptance of a graft. Thus, the invention also includes methods in a subject which has previously been administered donor hematopoietic stem cells, is administered one or more cytokine, e.g., a donor-species cytokine.
  • Although not wishing to be bound by theory, the inventor believes that the cytokines, particularly donor species cytokines, promote the engraftment and/or function of donor stem cells or their progeny cells. Accordingly, any method referred to herein which includes the administration of hematopoietic stem cells can further include the-administration of a cytokine, e.g., SCF, IL-3, or GM-CSF. In preferred embodiments the cytokine one which is species specific in its interaction with target cells.
  • Administration of a cytokine can begin prior to, at, or after the implantation of a graft or the implantation of stem cells.
  • The method can further include the step of administering a first or subsequent dose of a cytokine to the recipient: when the recipient begins to show signs of rejection, e.g., as evidenced by a decline in function of the grafted organ, by a change in the host donor specific antibody response, or by a change in the host lymphocyte response to donor antigen; when the level of chimerism decreases; when the level of chimerism falls below a predetermined value; when the level of chimerism reaches or falls below a level where staining with a monoclonal antibody specific for a donor PBMC antigen is equal to or falls below staining with an isotype control which does not bind to PBMC's, e.g. when the donor specific monoclonal stains less than 1-2% of the cells; or generally, as is needed to maintain tolerance or otherwise prolong the acceptance of a graft. Thus, method of the invention can be modified to include a further step of determining if a subject is in need of cytokine therapy and if so, administering a cytokine.
  • The period over which the cytokine(s) is administered (or the period over which clinically effective levels are maintained in the subject) can be long term, e.g., for six months of more or a year or more, or short term, e.g., for a year or less, more preferably six months or less, more preferably one month or less, and more preferably two weeks or less. The period will generally be at least about one week and preferably at least about two weeks in duration.
  • In preferred embodiments the recipient is a primate, e.g., a human, and the donor is from a different species, e.g., the donor is a pig and: pig SCF is administered; pig IL-3 is administered; a combination of pig SCF and pig IL-3 is administered; a pig specific hematopoiesis enhancing factor, e.g., pig GM-SCF, is administered, e.g., after the implantation of stem cells, e.g., about a month after the implantation of stem cells.
  • A particularly preferred embodiment combines a short course, e.g., about a month, of cyclosporine or a similar agent, a short course, e.g., about two weeks, of 15-deoxyspergualin or a similar agent, and a short course, e.g., about two weeks, of donor specific cytokines, e.g., SCF and IL-3. In Cynomolgus monkeys receiving pig grafts and pig stem cells, treatment which included the combination of cyclosporine (15 mg/kg/day for 28 days), 15-deoxyspergualin (6 mg/kg/day for two weeks), and recombinant pig cytokines (SCF and IL-3, each at 10 μg/kg/day, i.v., for two weeks) was found to be useful. Administration began at the time of graft implant. (The monkeys were also given a preparative regimen consisting of 3×100 cGy total body irradiation on day −6, and −5 and hemoperfusion with a pig liver just prior to stem cell administration.)
  • An anti-CD2 antibody, preferably a monoclonal, e.g., BTI-322, or a monoclonal directed at a similar or overlapping epitope, can be used in addition to or in place of any anti-T cell antibodies (e.g., ATG) in any method referred to herein.

Claims (23)

1. A method of restoring or promoting the thymus-dependent ability for T cell progenitors to develop into mature functional T cells in a primate recipient which is capable of producing T cell progenitors but which is thymus-function deficient, the method comprising introducing into said primate recipient xenogeneic fetal or neonatal thymus tissue from a donor animal of a different species, so that recipient T cells can mature in said implanted donor thymus tissue.
2. The method of claim 1, wherein the donor animal is a swine.
3. The method of claim 2, further comprising implanting swine fetal liver tissue in said recipient.
4. The method of claim 2, wherein said primate recipient is a human.
5. The method of claim 2, wherein said donor swine is a miniature swine.
6. The method of claim 2, wherein said swine thymus tissue is capable of supporting in the recipient primate clonal deletion or anergy of thymocytes reactive with donor xenoantigens.
7. The method of claim 2, further comprising introducing swine hematopoietic cells into said recipient and inactivating NK cells of said recipient.
8. The method of claim 2, further comprising the step of inactivating mature CD4+ T cells in the recipient.
9. A method of inducing tolerance in a recipient primate of a first species to a graft obtained from a donor animal of a second discordant species, said method comprising
prior or simultaneous with transplantation of said graft, introducing into said recipient, thymic tissue from the second species; and
implanting said graft in said recipient.
10. The method of claim 9, wherein the donor animal is a swine.
11. The method of claim 10, wherein the same swine is the donor of both the graft and the thymic tissue.
12. The method of claim 9, wherein said primate is a human.
13. The method of claim 9, said method further comprising the step of, prior or simultaneous with transplantation of said graft, inactivating mature CD4+ T cells of the recipient.
14. The method of claim 9, further comprising the step of, prior to thymic tissue transplantation, irradiating the recipient with low dose whole body irradiation.
15. The method of claim 14, wherein said low dose irradiation is at least 100 rads and less than 400 rads.
16. The method of claim 9, further comprising the step, prior to thymic tissue transplantation, adsorbing natural antibodies from the blood of said recipient.
17. The method of claim 9, wherein said thymic tissue is capable of supporting, in the recipient primate, clonal deletion or anergy of thymocytes reactive with donor xenoantigens.
18. The method of claim 9, wherein hematopoietic cells are administered to said recipient and wherein the method includes inactivating NK cells of said recipient.
19. The method of claim 9, wherein said primate recipient is a human and said donor is a miniature swine.
20. A method of providing mature recipient T cells in a primate recipient which is thymus function deficient, comprising introducing into the primate recipient swine xenogeneic donor thymic tissue, so that recipient T cells can mature in said implanted swine donor thymic tissue.
21. The method of claim 20, wherein said primate is a human and said donor is a miniature swine.
22. The method of claim 20, wherein said thymus function is deficient in said recipient due to an immune disorder.
23.-39. (canceled)
US11/151,744 1992-02-19 2005-06-13 Allogeneic and xenogeneic transplantation Abandoned US20060147428A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/151,744 US20060147428A1 (en) 1992-02-19 2005-06-13 Allogeneic and xenogeneic transplantation

Applications Claiming Priority (14)

Application Number Priority Date Filing Date Title
US83859592A 1992-02-19 1992-02-19
US08/114,072 US5624823A (en) 1991-11-22 1993-08-30 DNA encoding procine interleukin-10
US15073993A 1993-11-10 1993-11-10
PCT/US1994/001616 WO1995021527A1 (en) 1994-02-14 1994-02-14 Stem cell engraftment
US21222894A 1994-03-14 1994-03-14
US22037194A 1994-03-29 1994-03-29
PCT/US1994/005527 WO1994026289A1 (en) 1992-02-19 1994-05-16 Allogeneic and xenogeneic transplantation
US08/243,653 US5658564A (en) 1992-02-19 1994-05-16 Xenograft thymus
US08/266,427 US5614187A (en) 1991-11-22 1994-06-27 Specific tolerance in transplantation
US08/451,210 US6296846B1 (en) 1992-01-08 1995-05-26 Induced tolerance to xenografts
US08/458,720 US5876708A (en) 1992-02-19 1995-06-01 Allogeneic and xenogeneic transplantation
US12670498A 1998-07-30 1998-07-30
US09/710,971 US6911220B1 (en) 1992-02-19 2000-11-09 Allogeneic and xenogeneic transplantation
US11/151,744 US20060147428A1 (en) 1992-02-19 2005-06-13 Allogeneic and xenogeneic transplantation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/710,971 Division US6911220B1 (en) 1992-02-19 2000-11-09 Allogeneic and xenogeneic transplantation

Publications (1)

Publication Number Publication Date
US20060147428A1 true US20060147428A1 (en) 2006-07-06

Family

ID=34812502

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/710,971 Expired - Lifetime US6911220B1 (en) 1992-02-19 2000-11-09 Allogeneic and xenogeneic transplantation
US11/151,744 Abandoned US20060147428A1 (en) 1992-02-19 2005-06-13 Allogeneic and xenogeneic transplantation

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/710,971 Expired - Lifetime US6911220B1 (en) 1992-02-19 2000-11-09 Allogeneic and xenogeneic transplantation

Country Status (1)

Country Link
US (2) US6911220B1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070031406A1 (en) * 2003-10-22 2007-02-08 Zand Martin S Anti-thymocyte antiserum and use thereof to trigger b cell apoptosis
WO2009026472A1 (en) * 2007-08-21 2009-02-26 The General Hospital Corporation Methods for inducing tolerance
US9883939B2 (en) 2012-05-08 2018-02-06 The General Hospital Corporation Reducing immunogenicity of xenogeneic transplant tissues
US9999600B2 (en) 2013-04-03 2018-06-19 N-Fold Llc Nanoparticle compositions
WO2019165197A1 (en) 2018-02-23 2019-08-29 Duke University & Medical Center Cultured thymus tissue transplantation promotes donor-specific tolerance to allogeneic solid organ transplants
US11071776B2 (en) 2012-04-23 2021-07-27 N-Fold Llc Nanoparticles for treatment of allergy
US11819520B2 (en) 2018-02-23 2023-11-21 Duke University Cultured thymus tissue transplantation promotes donor-specific tolerance to allogeneic solid organ transplants

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1658096A1 (en) * 2003-08-25 2006-05-24 PanGenetics B.V. Method of inducing immune tolerance
US20060148080A1 (en) * 2004-12-30 2006-07-06 Paul Diamond Methods for supporting and producing human cells and tissues in non-human mammal hosts
US20060147429A1 (en) * 2004-12-30 2006-07-06 Paul Diamond Facilitated cellular reconstitution of organs and tissues
WO2007033221A2 (en) * 2005-09-13 2007-03-22 The General Hospital Corporation Methods and compositions for inhibition of immune responses
EP1977234B1 (en) 2006-01-13 2015-11-04 Indiana University Research & Technology Corporation Type V collagen for use in a method of treatment of idiopathic pulmonary fibrosis
CN100415308C (en) * 2006-09-12 2008-09-03 南方医科大学 A new type of homologous bone and its preparation method
EP2607477B1 (en) 2007-05-03 2020-09-23 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US8574567B2 (en) * 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
BRPI0915582A2 (en) * 2008-06-06 2016-01-26 Baylor Res Inst anto-cd8 antibodies that block the sensitization of cytotoxic effectors and lead to the generation of regulatory cd8 + t cells
JP5688734B2 (en) * 2008-12-12 2015-03-25 国立大学法人 東京大学 Immunological reconstruction promoter or infection preventive agent that preserves graft versus tumor effect
US8858647B2 (en) 2010-05-05 2014-10-14 Barry Markman Method and apparatus for a process creating an internal tissue graft for animal and human reconstructive purposes
US9622845B2 (en) 2010-05-05 2017-04-18 Barry Markman Method and apparatus for creating a reconstructive graft
US11701213B2 (en) 2010-05-05 2023-07-18 Markman Biologics Corporation Method and apparatus for creating a modified tissue graft
US11246697B2 (en) 2010-05-05 2022-02-15 Markman Biologics Corporation Method and apparatus for creating a reconstructive graft
US11877921B2 (en) 2010-05-05 2024-01-23 Markman Biologics Corporation Method and apparatus for creating a modified tissue graft

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5597563A (en) * 1992-09-04 1997-01-28 Beschorner; William E. Method induction of antigen-specific immune tolerance
US5658564A (en) * 1992-02-19 1997-08-19 The General Hospital Corporation Xenograft thymus

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993009815A1 (en) 1991-11-22 1993-05-27 The General Hospital Corporation Specific tolerance in transplantation
AU6268194A (en) 1994-02-14 1995-08-29 General Hospital Corporation, The Stem cell engraftment

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5658564A (en) * 1992-02-19 1997-08-19 The General Hospital Corporation Xenograft thymus
US5597563A (en) * 1992-09-04 1997-01-28 Beschorner; William E. Method induction of antigen-specific immune tolerance

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070031406A1 (en) * 2003-10-22 2007-02-08 Zand Martin S Anti-thymocyte antiserum and use thereof to trigger b cell apoptosis
WO2009026472A1 (en) * 2007-08-21 2009-02-26 The General Hospital Corporation Methods for inducing tolerance
US11071776B2 (en) 2012-04-23 2021-07-27 N-Fold Llc Nanoparticles for treatment of allergy
US9883939B2 (en) 2012-05-08 2018-02-06 The General Hospital Corporation Reducing immunogenicity of xenogeneic transplant tissues
US9999600B2 (en) 2013-04-03 2018-06-19 N-Fold Llc Nanoparticle compositions
WO2019165197A1 (en) 2018-02-23 2019-08-29 Duke University & Medical Center Cultured thymus tissue transplantation promotes donor-specific tolerance to allogeneic solid organ transplants
CN112074277A (en) * 2018-02-23 2020-12-11 杜克大学医学中心 Cultured thymus tissue transplantation promotes donor-specific tolerance to allogenic solid organ transplants
US11819520B2 (en) 2018-02-23 2023-11-21 Duke University Cultured thymus tissue transplantation promotes donor-specific tolerance to allogeneic solid organ transplants

Also Published As

Publication number Publication date
US6911220B1 (en) 2005-06-28

Similar Documents

Publication Publication Date Title
US5876708A (en) Allogeneic and xenogeneic transplantation
US20060147428A1 (en) Allogeneic and xenogeneic transplantation
AU673987C (en) Specific tolerance in transplantation
US6877514B2 (en) Mixed chimerism and tolerance
US6514513B1 (en) Costimulatory blockade and mixed chimerism in transplantation
US5658564A (en) Xenograft thymus
WO1994026289A1 (en) Allogeneic and xenogeneic transplantation
US20020168348A1 (en) Allogeneic and and xenogeneic transplantation
EP0699073B1 (en) xenogeneic transplantation
EP1005866B1 (en) Allogeneic and xenogeneic transplantation
AU771025B2 (en) Specific tolerance in transplantation
EP0697876B1 (en) Xenograft thymus
WO1995013363A1 (en) Hematopoietic stem cells from swine cord blood and uses thereof
EP1048298B1 (en) Mixed chimerism and tolerance
US20020114787A1 (en) Tolerance to natural antibody antigens
EP1310260A1 (en) Mixed chimerism and tolerance

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NIH - DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE GENERAL HOSPITAL CORPORATION;REEL/FRAME:048117/0590

Effective date: 20190124

Owner name: NIH - DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE GENERAL HOSPITAL CORPORATION;REEL/FRAME:048118/0349

Effective date: 20190124