WO2000061132A1 - Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft - Google Patents

Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft Download PDF

Info

Publication number
WO2000061132A1
WO2000061132A1 PCT/US2000/010253 US0010253W WO0061132A1 WO 2000061132 A1 WO2000061132 A1 WO 2000061132A1 US 0010253 W US0010253 W US 0010253W WO 0061132 A1 WO0061132 A1 WO 0061132A1
Authority
WO
WIPO (PCT)
Prior art keywords
dendritic cell
agent
cell maturation
immunotoxin
recipient
Prior art date
Application number
PCT/US2000/010253
Other languages
French (fr)
Other versions
WO2000061132A8 (en
Inventor
David M. Neville
Judith M. Thomas
Original Assignee
The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services
The Uab Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, The Uab Research Foundation filed Critical The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services
Priority to CA002382628A priority Critical patent/CA2382628A1/en
Priority to JP2000610465A priority patent/JP2002541195A/en
Priority to AU43542/00A priority patent/AU781547B2/en
Priority to EP00923415A priority patent/EP1171109A1/en
Priority to BR0009772-1A priority patent/BR0009772A/en
Publication of WO2000061132A1 publication Critical patent/WO2000061132A1/en
Publication of WO2000061132A8 publication Critical patent/WO2000061132A8/en
Priority to HK02103064.3A priority patent/HK1042648A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages

Definitions

  • Figures 2A, 2B, and 2C show the results of RT-PCR assays performed on freshly obtained rhesus monkey peripheral blood lymphocytes treated with either whole IgG immunotoxin (FN18-CRM9), F(Ab) 2 immunotoxin (FN18-F(Ab) 2 -CRM9), or control buffered saline (PBS).
  • Figure 2A shows mRNA for IL-2, IL-4, IL-10, IFN- ⁇ in the absence of DSG.
  • Figure 2B shows mRNA for IL-2, IL-4, IL-10, IFN- ⁇ in cells treated with DSG (2.5 ⁇ g/ml).
  • Figure 2C shows the density ratio of the various rnRNAs to the actin control.
  • a "graft" can include an allogenic or xenogenic organ, tissue, or cellular transplant.
  • the graft for example, can be selected from the group consisting of kidney, liver, heart, pancreas, lung, skin, and isolated cell transplants of pancreatic islets, hepatocytes, stem cell precursors, and differentiated stem cell precursors.
  • the donor grafts used in the present methods can comprise cells that are altered, such as by genetically engineering the donor or donor cells.
  • the donor cells could be engineered to reduce antigenicity or to reduce the susceptibility of transplanted cells to immune injury (R. Weiss, Nature 391: 327-28 (1998)).
  • the agent that inhibits dendritic cell maturation is administered to the recipient at least once. Even more preferably, the agent that inhibits dendritic cell maturation can be administered to the recipient at least on the day of transplantation and 1, 2, 3, 4, 5, 6, or 7 additional days within the first week following transplantation or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 additional days within the first two weeks following the day of transplantation.
  • the course of administration can optionally be optimized to produce a sustained increase in the activation of NF-AT transcription pathway mediated cytokines and/or the inhibition of NfkB transcription pathway mediated cytokines.
  • the agent is administered daily, on the day of transplantation and 13 or 14 additional times.
  • immunotoxin administration of up to 4, 5, 6, or 7 days can be used, but such an extended time course of immunotoxin administration would require that the production of antitoxin antibodies, which begins after approximately 5 days of administration, be addressed.
  • the immunotoxin can be administered in subjects beginning anytime after transplantation.
  • a subject with a long term surviving transplant, who has not previously received immunotoxin treatment could still benefit from immunotoxin administration, thereby avoiding the need for long-term treatment with immunosuppressives.
  • a graft recipient who begins to show signs of rejection may benefit from immunotoxin administration to reduce or eliminate the rejection process.
  • recipients showing a rejection responses can be further treated with additional immunotoxin administration.
  • Renal allografts survived without histologic or clinical evidence of acute or chronic rejection in 80%> of IT-treated recipients given MP plus deoxyspergualin x 15d, in 40%) in those given MP plus deoxyspergualin x 5d, and in 0%> of those given only MP or MP plus Neoral.
  • the 3 longest deoxyspergualin survivors are >830 days (2.3 years) with normal renal function and immunologic evidence of specific tolerance.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pathology (AREA)
  • Rheumatology (AREA)
  • Toxicology (AREA)
  • Diabetes (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Transplantation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides a method of inducing immune tolerance to a graft in a recipient, comprising administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population; and administering to the recipient an agent that inhibits dendritic cell maturation. The present invention also provides a method of screening for an agent that acts synergistically with an immunotoxin in inducing immune tolerance and a method of screening for an agent that inhibits dendritic cell maturation. The present invention also provides a method of treating a subject with an autoimmune disease, comprising administering to the subject an immunotoxin, thereby reducing the subject's T-cell population; and administering to the subject an agent that inhibits dendritic cell maturation. Also provided herein is a composition comprising an immunotoxin and an agent that inhibits dendritic cell maturation.

Description

COMPOSITIONS CONTAINING IMMUNOTOXINS AND AGENTS THAT INHIBIT DENDRITIC CELL MATURATION FOR INDUCING IMMUNE TOLERANCE TO A GRAFT
BACKGROUND OF THE INVENTION
Field of The Invention
This invention relates to techniques for inducing immune tolerance using an immunotoxin combined with an agent that inhibits dendritic cell maturation. The invention is in the field of immunobiology .
10
Background Art
Transplant tolerance remains an elusive goal for patients and physicians whose ideal would be to see a successful, xenogeneic transplantation performed without the need for indefinite, non-specific maintenance immunosuppressive drugs and their 15 attendant side effects. As with many transplant procedures, securing viable allogeneic grafts can be difficult. In addition, long term immunosuppression can be problematic.
Over the past 10 years, the majority of transplant recipients have been treated with cyclosporin, azathioprine, prednisone, and a variety of other immunosuppressive
20 agents for maintenance immunosuppression. The average annual cost of maintenance immunosuppressive therapy in the United States is approximately $10,000. In addition to the cost, these agents, because of their non-specific effects, have considerable side effects, including compromising cell and organ function and increasing susceptibility to infection. A major goal in transplant immunobiology is the development of specific
25 immunologic tolerance to transplants with the potential of freeing patients from the side effects of continuous pharmacologic immunosuppression and its attendant complications and costs.
Anti-T cell therapy (anti-lymphocyte globulin) has been used in rodents in 30 conjunction with thymic injection of donor cells (Posselt et al. Science 1990; 249:
1293- 1295 and Remuzzi et al. Lancet 1991; 337: 750-752). Thymic tolerance, which has proved successful in rodent models, involves the exposure of the recipient thymus gland to donor alloantigen prior to an organ allograft from the same donor. However, thymic tolerance has never been reported in large animals, and its relevance to tolerance in humans in unknown.
One approach to achieving immune tolerance has been to expose the recipient to cells from the donor prior to the transplant, with the hope of inducing tolerance to a later transplant. This approach has involved placement of donor cells (e.g., bone marrow) presenting MHC Class I antigens in the recipient's thymus shortly after application of anti-lymphocyte serum (ALS) or radiation. However, this approach has proved difficult to adapt to live primates (e.g., monkeys or humans). ALS and/or radiation render the host susceptible to disease or side-effects and/or are insufficiently effective.
If a reliable, safe approach to specific immunologic tolerance to transplantation, particularly xenogeneic transplantation, could be induced, this would be of tremendous value and appeal to patients and transplant physicians throughout the world, with immediate application to new transplants and with potential application to existing transplants in recipients with stable transplant function. Thus, a highly specific immune tolerance inducement is desired. Furthermore, there is a need for a means for imparting tolerance in primates, without the adverse effects of using ALS or radiation. Moreover, the goal is to achieve tolerance rather than simply delaying the rejection response. An important goal is to inhibit the rejection response to the point that rejection is not a factor in reducing average life span among transplant recipients.
SUMMARY OF THE INVENTION
The present invention provides a method of inducing immune tolerance to a graft in a recipient, comprising administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population; and administering to the recipient an agent that inhibits dendritic cell maturation. The agent that inhibits dendritic cell maturation is administered to the recipient at least once and, preferably, on the day of transplantation. Even more preferably, the agent that inhibits dendritic cell maturation is administered to the recipient an additional four to fourteen times over the course of one to two weeks following transplantation. Optionally, the agent that inhibits dendritic cell maturation can be administered to the recipient prior to transplantation, and/or to the donor prior to harvesting the graft. The agent that inhibits dendritic cell maturation can be an inhibitor of nuclear translocation of NfkB, including, for example, deoxyspergualin, methyl-deoxyspergualin, and other deoxyspergualin derivatives or analogs. Other agents that inhibit dendritic cell maturation can include, for example, a soluble interleukin 17 (IL-17) receptor Fc fusion protein, a glucocorticoid, a blocker of tumor necrosis factor alpha binding, a blocker of granulocyte macrophage colony stimulating factor binding, a blocker of IL-12p70 binding, or a blocker of IL-lβ binding. An agent that inhibits dendritic cell maturation can also include a blocker of an immature dendritic cell epitope or a blocker of a dendritic cell precursor epitope involved in dendritic cell maturation such as an anti-CD40 ligand (i.e., anti-CD154), and, more specifically, the anti-CD40 ligand can be 5C8 or a derivative of analog thereof.
The present invention also provides a method of screening for an agent that acts synergistically with an immunotoxin in inducing immune tolerance, comprising transplanting a donor graft to a recipient; administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population; administering to the recipient the agent to be screened; obtaining a dendritic cell- containing sample; and determining a percentage of the dendritic cells in the sample that express or can be induced to express a marker specific for mature dendritic cells, wherein a low percentage shows a synergistic action. Also provided by the present invention is a method of screening for an agent that inhibits dendritic cell maturation, comprising obtaining a population of immature dendritic cells from a dendritic cell- containing sample of a subject; culturing the population of cells in the presence of the agent to be screened; and determining a percentage of dendritic cells that express or can be induced to express a marker specific for mature dendritic cells, wherein a low percentage shows inhibition of dendritic cell maturation. The present invention also provides a method of treating a subject with an autoimmune disease, comprising administering to the subject an immunotoxin, thereby reducing the subject's T-cell population; and administering to the subject an agent that inhibits dendritic cell maturation.
Also provided herein is a composition comprising an immunotoxin and an agent that inhibits dendritic cell maturation. More specifically, the present invention provides a composition, wherein the immunotoxin is an anti-T cell immunotoxin directed at the CD3 epitope or wherein the agent that inhibits dendritic cell maturation is an inhibitor of nuclear translocation of NfkB.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows that kidney transplant tolerance induction by treatment of the transplant recipient with immunotoxin on days 0 and +1 in combination with DSG on days 0-14 results in a prominent Th2 cytokine polarization. By the second week post transplantation, IL4 polarization (a lower level of IL4 compared to the group treated with immunotoxin alone and lower levels of IL4 compared to IFN-γ levels) is shown in the groups that received the combined treatment. The group that received DSG for two weeks showed a sustained, progressive increase in IL4 polarization at 4 weeks, whereas, in the group that received DSG for only 5 days, the polarization was no longer sustained and the cytokine pattern was reversed. The IL4 polarization occurred independently of the type of anti-CD3 immunotoxin, i.e., the whole IgG or F(Ab)2 form.
Figures 2A, 2B, and 2C show the results of RT-PCR assays performed on freshly obtained rhesus monkey peripheral blood lymphocytes treated with either whole IgG immunotoxin (FN18-CRM9), F(Ab)2 immunotoxin (FN18-F(Ab)2-CRM9), or control buffered saline (PBS). Figure 2A shows mRNA for IL-2, IL-4, IL-10, IFN-γ in the absence of DSG. Figure 2B shows mRNA for IL-2, IL-4, IL-10, IFN-γ in cells treated with DSG (2.5 μg/ml). Figure 2C shows the density ratio of the various rnRNAs to the actin control.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a short course, immune tolerance inducing treatment regimen utilizing an immunotoxin that, when combined with agents that inhibit dendritic cell maturation, prevents transplant rejection while maintaining transplant function. Thus, the present invention provides a method of inducing immune tolerance to a graft in a recipient, comprising administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population; and administering to the recipient an agent that inhibits dendritic cell maturation.
As used throughout, a "graft" can include an allogenic or xenogenic organ, tissue, or cellular transplant. The graft, for example, can be selected from the group consisting of kidney, liver, heart, pancreas, lung, skin, and isolated cell transplants of pancreatic islets, hepatocytes, stem cell precursors, and differentiated stem cell precursors.
As used throughout, the "recipient" or "subject" is preferably a mammal. More preferably, the mammalian recipient is a primate, and, even more preferably, a human. The "recipient" or "subject" being treated can include individual humans, domesticated animals, livestock (e.g., cattle, horses, pigs, etc.), and pets (e.g., cats and dogs).
As used throughout, a "donor" can be a cadaver or a living donor. Furthermore, the donor can be of the same species as the subject being treated or a different species than the subject being treated. Thus, using the method of the invention, transplantation can be performed across primate species (i.e., xenogeneic transplantation or xenograft) and within the same primate line (i.e., allogeneic transplantation or allograft). Even highly sensitive xenografts can maintain function in the presence of this immune tolerance inducing regimen.
The donor grafts used in the present methods can comprise cells that are altered, such as by genetically engineering the donor or donor cells. For example, the donor cells could be engineered to reduce antigenicity or to reduce the susceptibility of transplanted cells to immune injury (R. Weiss, Nature 391: 327-28 (1998)).
As used herein, "an agent that inhibits dendritic cell maturation" refers to any agent that, when in contact with dendritic cell precursors, inhibits all or a portion of the precursors from expressing markers of mature dendritic cells. Thus, an agent that inhibits dendritic cell maturation would cause about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% fewer cells to express markers such as membrane CD83, DR, or CD86 or nuclear Rel-B than in the absence of the agent. The effectiveness of the inhibitor of dendritic cell maturation, therefore, can be assessed by determining the percent decrease in the number of cells expressing markers for mature dendritic cells, as described more fully in the examples provided herein.
The agent that inhibits dendritic cell maturation can be an inhibitor of nuclear translocation of NfkB. More specifically, the inhibitor of nuclear translocation of
NfkB can be deoxyspergualin or a derivative or analog thereof, including, for example, methyl-deoxyspergualin or a deoxyspergualin analog lacking a chiral center (e.g., LF 08-0299) (Andoins et al., 1996, which is incorporated herein by reference). Other derivatives or analogs of deoxyspergualin can be used that include, for example, those identified in U.S. Pat. No. 4,518,532; U.S. Pat. No. 4,518,532; U.S. Pat. No. 4,525,299; U.S. Pat. No. 4,956,504; U.S. Pat. No. 5,162,581; U.S. Pat. No. 5, 476,870; U.S. Pat. No. 5,637,613; W.O. 96/24579; EP 600762; EP 669316; EP 7433000; EP 765866; and EP 755380, which are incorporated herein by reference.
Preferably, the agent that inhibits dendritic cell maturation activates one or more
NF-AT dependent Th2 cytokines (e.g., one or more cytokine selected from the group consisting of IL2, IL4, and IL10). Also, the agent that inhibits dendritic cell maturation preferably inhibits one or more Nf B dependent Thl cytokines (e.g., LNFγ). Such activation or inhibition can be due to an increase or decrease, respectively, in the cytokine gene expression. Thus, alterations in levels of the mRNA encoding the cytokine or protein levels can occur with treatment with the agent. In one embodiment of the invention, the agent activates one or more NF-AT dependent Th2 cytokines and inhibits one or more NfkB dependent Thl cytokines. Thus, for example, the agent can activate IL4 and inhibit LNFγ.
Alternatively, the agent that inhibits dendritic cell maturation can be a soluble
IL-17 receptor Fc fusion protein, a glucocorticoid, a blocker of tumor necrosis factor alpha (TNF- ) binding, a blocker of granulocyte macrophage colony stimulating factor (GM-CSF) binding, a blocker of IL-12p70 binding, or a blocker of IL-lβ binding. An agent that inhibits dendritic cell maturation can also include a blocker of an immature dendritic cell epitope or a blocker of a dendritic cell precursor epitope involved in dendritic cell maturation such as an anti-CD40 ligand (i.e., anti-CD 154), and, more specifically, the anti-CD40 ligand can be 5C8 or a derivative of analog thereof. One skilled in the art would recognize that agents that inhibit dendritic cell maturation can be used in combination for either a synergistic or additive effect.
Preferably, the agent that inhibits dendritic cell maturation is administered to the recipient at least once. Even more preferably, the agent that inhibits dendritic cell maturation can be administered to the recipient at least on the day of transplantation and 1, 2, 3, 4, 5, 6, or 7 additional days within the first week following transplantation or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 additional days within the first two weeks following the day of transplantation. The course of administration can optionally be optimized to produce a sustained increase in the activation of NF-AT transcription pathway mediated cytokines and/or the inhibition of NfkB transcription pathway mediated cytokines. Thus, in one embodiment, the agent is administered daily, on the day of transplantation and 13 or 14 additional times. In addition to administration on and after the day of transplantation, the agent that inhibits dendritic cell maturation can additionally be administered to the recipient prior to transplantation. For example, deoxyspergualin can be administered to the recipient at least once between 24 hours and 0.25 hours, and preferably, 23, 22, 21, 20, 19, 18, 17, 16, 15,14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5 hours, prior to transplantation. Optionally, the present invention can further comprise administering to a transplant donor, prior to harvesting the transplant, an agent that inhibits dendritic cell maturation. For example, deoxyspergualin can be administered to the donor at least once between 24 hours and 0.25 hours, and preferably, 23, 22, 21, 20, 19, 18, 17, 16, 15,14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5 hours, prior to harvesting the graft. Although the time course for administration of inhibitor of dendritic cell maturation prior to transplantation may be limited by the unpredictability of organ availability and procurement, it is preferred to administer the inhibitor to the recipient prior to transplantation and/or to the donor prior to harvesting the graft.
The preferred dose of inhibitors of dendritic cell maturation will also vary depending on the specific inhibitor used; the species, age, weight and general condition of the recipient; the mode of administration; and the like. Thus, it is not possible or necessary to specify an exact dose, as an appropriate time course may be determined without undue experimentation by one of ordinary skill in the art. By way of example, the dose of deoxyspergualin can be between 0.1 and 10 mg/kg/d, including 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 10.0 mg/kg/d or any amount in between.
As used throughout, an "immunotoxin" can be an anti-T cell immunotoxin directed at the CD3 epitope. The immunotoxin can be an immunoconjugate or fusion protein. The immunotoxin can be either monovalent or divalent. The divalent anti-T cell immunotoxin can be UCHT1-CRM9 or a derivative or analog thereof. The divalent anti-T cell immunotoxin can comprise a toxin moiety and a targeting moiety directed to the T cell CD3e epitope, and the toxin moiety can be a diphtheria toxin. The divalent anti-T cell immunotoxin can be an engineered divalent fusion immunotoxin. Various immunotoxins and treatment regimens are described more fully in U.S. Serial No. 09/064,413 and PCT/US 98/04303, which are incorporated herein by reference. The immunotoxin can be, for example, a whole IgG immunotoxin (such as FN18-CRM9) or can be a F(Ab)2 form of the immunotoxin (such as FN18-F(Ab)2 CRM9).
In the present invention, the immunotoxin is administered at least two times, preferably at least on the day of transplantation and on the second day following transplantation. The immunotoxin can be administered beginning at 72 to 0 hours before transplantation and continuing up to several days thereafter. Preferably, the immunotoxin can be administered to the recipient, to the donor, or both 72-48 hours prior to a xenogeneic transplant and 24 to 0 hours prior to an allogeneic transplant. Preferably, the immunotoxin is administered to the recipient for 1, 2, or 3 days following transplantation or any time in between. It is contemplated that immunotoxin administration of up to 4, 5, 6, or 7 days can be used, but such an extended time course of immunotoxin administration would require that the production of antitoxin antibodies, which begins after approximately 5 days of administration, be addressed. The immunotoxin can be administered in subjects beginning anytime after transplantation. Thus, it is contemplated that a subject with a long term surviving transplant, who has not previously received immunotoxin treatment, could still benefit from immunotoxin administration, thereby avoiding the need for long-term treatment with immunosuppressives. It is further contemplated that a graft recipient who begins to show signs of rejection may benefit from immunotoxin administration to reduce or eliminate the rejection process. Thus, even in recipients that have previously been treated with immunotoxins, recipients showing a rejection responses can be further treated with additional immunotoxin administration.
As used throughout, the immunotoxin preferably transiently reduces the subject's or recipient's T cells in the blood and lymph nodes by at least one log unit. Preferably the number of T cells in the blood and lymph nodes will be transiently decreased by 0.7, 0.8, 0.9, 1 , 1.1 , 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, or 3 log units, or any interval amount between 0.7 and 3 log units. By "transiently reduces" is meant that T cells are reduced by 0.7 to 3 log units in the blood and lymph compartments for at least four days before starting to return to normal levels.
The present method can further comprise administering an immunosuppressive agent to the recipient. As used throughout, an immunosuppressive agent, or immunosuppressant, includes, for example, methylprednisolone, Neoral, cyclosporine, mycophenolate moefitil, tacrolimus, azathioprine, rapamycin, a steroid, or any combination thereof. The immunosuppressive agents can be administered beginning 24 to 0 hours prior to transplantation and continuing up to two weeks thereafter.
Preferably, the immunosuppressive agents can be administered for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 days, or any interval time. The regimen of combined immunotoxin and dendritic cell maturation inhibition agent allows short-term immunosuppressive therapy and eliminates the need for long-term treatment with immunosuppressives, thereby avoiding the side effects associated with chronic immunosuppression. Thus, when a course of immunosuppressants is used in conjunction with immunotoxins and an agent that inhibits dendritic cell maturation, the course of immunosuppression can be shorter in duration and/or lower in dosage than is traditionally used to prevent graft rejection.
Adjunct therapies can be used together in the present methods of inducing immune tolerance and treating immune disorders . Thus, the invention includes at least one method of inducing tolerance using immunotoxin (IT): (1) tolerance induction by administering IT in combination with an agent or agents that inhibit dendritic cell maturation; (2) tolerance induction by administering IT, an agent or agents that inhibit dendritic cell maturation, and at least one or a combination of immunosuppressant drug. The adjunct therapy can be administered before, at the same time or after the administration of immunotoxin. Different adjunct therapies can be administered to the recipient at different times or at the same time in relation to the transplant event or the administration of immunotoxin, as further described below. Because the immunosuppressant can be administered before treatment with the immunotoxin and agent that inhibits dendritic cell maturation, the present method of combining immunotoxins and agents that inhibit dendritic cell maturation can be used with a graft recipient who is on an immunosuppressant regimen. This presents a significant opportunity to reduce or eliminate traditional immunosuppressant therapy and its well documented negative side-effects. Also, treatment with agents that inhibit dendritic cell maturation and/or immunosuppressants prior to transplantation could be particularly useful in cadaveric and xenogeneic transplants. In such a setting of pre- transplant treatment, the administration of immunotoxin can be delayed for up to seven or more days post-transplantation.
Examples of schedules of administration of an immunotoxin and an agent that inhibits dendritic cell maturation, for patients receiving organ transplants are as follows:
day -24 to -0 hours: treat recipient with the agent that inhibits dendritic cell maturation (optional) day 0 : perform transplant; immediately following transplant, administer 1st immunotoxin dose; treat the recipient with the agent that inhibits dendritic cell maturation day 1 : treat the recipient with the agent that inhibits dendritic cell maturation day 2 : 2nd immunotoxin dose; treat the recipient with the agent that inhibits dendritic cell maturation day 3 : treat recipient with the agent that inhibits dendritic cell maturation day 6 : treat recipient with the agent that inhibits dendritic cell maturation; or day -24 to -0 hours: treat recipient with the agent that inhibits dendritic cell maturation (optional) day 0 : perform transplant; immediately following transplant, administer 1st immunotoxin dose; treat the recipient with the agent that inhibits dendritic cell maturation day 2 : 2nd immunotoxin dose day 4 : treat recipient with the agent that inhibits dendritic cell maturation day 7 : treat recipient with the agent that inhibits dendritic cell maturation; day 10 : treat recipient with the agent that inhibits dendritic cell maturation; day 13 : treat recipient with the agent that inhibits dendritic cell maturation.
In either example, the donor can be treated with the agent that inhibits dendritic cell maturation 24 to 0 hours prior to harvesting the transplant and/or the recipients can be treated with immunosuppressants prior to transplantation, on the day of transplantation, or following transplantation. One skilled in the art would know to modify the time course according to the individual recipient, depending on the species, age, weight and general condition of the recipient, the particular agents used, the mode of administration, and the like. Thus, it is not possible or necessary to specify an exact time course. However, an appropriate time course may be determined by one of ordinary skill in the art.
The presently preferred doses of the immunotoxin are those sufficient to deplete peripheral blood T-cell levels to 80%o, preferably 90%> (or especially preferably 95%> or higher) of preinjection levels. This should require mg/kg levels for humans similar to those for monkeys (e.g., 0.05 mg/kg to 0.3 mg/kg body weight), which toxicity studies indicate should be well tolerated by humans. Thus, the immunotoxin can be administered to safely reduce the recipients T cell population. The effectiveness of the method of inducing tolerance can be assessed using methods well known in the art. Clinical signs of graft function and graft rejection could be assessed. For example, using an method of inducing tolerance to a pancreatic or islet cell graft, blood glucose levels could be measured, and non-fasting blood glucose levels will preferably be maintained below 160 mg/dl upon completion of the immune tolerance induction regimen would indicate the absence of graft rejection. Other signs of graft rejection include histological characteristics (for example, upon biopsy of the transplant) such as interstitial fibrosis, interstitial hyperplasia, arteriolar narrowing, and ischemic injury to the transplanted organ. Additionally, histological signs of rejection of a kidney transplant include reduplication and thickening of the glomerular basement membrane. The clinical signs of rejection include gradually progressive graft dysfunction. For example, in a subject with a kidney transplant, increasing blood levels of creatinine would indicate rejection. Normal serum levels of creatinine are less than 2.0 mg/dl, and more typically about 0.5 to 1.5 mg/dl. Renal graft dysfunction is evidenced by approximately a two fold increase in baseline levels of creatinine. The presence of antidonor antibody and increasing levels of antidonor antibody could also indicate graft dysfunction. One skilled in the art would recognize other histological indicators and clinical indicators of rejection.
The present invention also provides a method of screening for an agent that acts synergistically with an immunotoxin in inducing immune tolerance, comprising transplanting a donor graft to a recipient; administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population; administering to the recipient the agent to be screened; obtaining a dendritic cell-containing sample; and determining a percentage of the dendritic cells in the sample that express or can be induced to express a marker specific for mature dendritic cells, wherein a low percentage shows a synergistic action. As used throughout, a "dendritic cell-containing sample" can include, for example, a lymph node biopsy, a blood sample, or a tonsil biopsy. Also, as used throughout, a "marker specific for mature dendritic cells" can include, for example, membrane CD83, membrane DR, membrane CD86, cytoplasmic P55, or nuclear Rel-B. These can be routinely detected using methods known in the art. (O'Doherty et al., 1997).
Also provided by the present invention is a method of screening for an agent that inhibits dendritic cell maturation, comprising obtaining a population of immature dendritic cells from a dendritic cell-containing sample of a subject; culturing the population of cells in the presence of the agent to be screened; and determining a percentage of dendritic cells that express or can be induced to express a marker specific for mature dendritic cells, wherein a low percentage shows inhibition of dendritic cell maturation. Preferably, the population of cells is cultured in monocyte-conditioned medium supplemented with TNFα.
The present invention also provides a method of treating a subject with an autoimmune disease, comprising administering to the subject an immunotoxin, thereby reducing the subject's T-cell population; and administering to the subject an agent that inhibits dendritic cell maturation. Autoimmune diseases that can be treated by the present method include, for example, systemic lupus erythematosus, myasthenia gravis, stiff-man syndrome, an autoimmune thyroid disease, Sydenham chorea, rheumatoid arthritis. One skilled in the art would recognize a variety of ways to assess the effectiveness of the treatment, for example, a reduction in clinical symptoms (such as decreased pain, improved muscle function, improved thyroid function) and histological or biochemical evidence of reduced T cells and or mature dendritic cells.
In the present method of treating autoimmune diseases, the agent that inhibits dendritic cell maturation is administered to the subject at least once, or two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen times, preferably over a period of about two weeks, and the immunotoxin is preferably administered at least one, two, or three times, preferably within a period of less than five days. As discussed above in the context of inducing immune tolerance, immunotoxin can be administered for about 4 days if the production of antitoxin antibodies is addressed. Alternatively, the method of treatment can further comprise administering an immunosuppressive agent to the subject. Treatment of immunosuppressants, however, could be of limited duration and a lower dose than traditionally used.
Because the immune functions that govern graft acceptance and rejection and autoimmune diseases are similar across primate species, the combined use of immunotoxins and agents that inhibit the maturation of dendritic cells as described herein is expected to succeed in humans for inducing immune tolerance or for treating autoimmune disorders.
The present invention also provides a composition comprising an immunotoxin and an agent that inhibits dendritic cell maturation. More specifically, the present invention provides a composition, wherein the immunotoxin is an anti-T cell immunotoxin directed at the CD3 epitope or wherein the agent that inhibits dendritic cell maturation is an inhibitor of nuclear translocation of NfkB. Preferably, the agent that inhibits dendritic cell maturation activates one or more NF-AT dependent Th2 cytokines. Also, the agent that inhibits dendritic cell maturation preferably inhibits one or more NfkB dependent Thl cytokines. In one embodiment of the invention, the agent activates one or more NF-AT dependent Th2 cytokines and inhibits one or more NfkB dependent Thl cytokines. Thus, for example, the agent can activate IL4 and inhibit LNFγ.
The composition of the present invention can further comprise a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" is meant a material that is not biologically or otherwise undesirable, i.e., the material can be administered to a subject along with the immunotoxin and agent that inhibits dendritic cell maturation without causing any substantial undesirable biological effects or interacting in a deleterious manner with any of the components of the pharmaceutical composition in which it is contained. The carrier would naturally be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject. Suitable carriers can include, for example, water, pyrogen free saline, pharmaceutically accepted oils, or a mixture of any of these. The carrier can also contain other suitable pharmaceutical additions such as buffers, preservatives, flavoring agents, viscosity or osmo-regulators, stabilizers or suspending agents.
The present invention is more particularly described in the following examples which are intended as illustrative only since numerous modifications and variations therein will be apparent to those skilled in the art.
EXAMPLE 1
Transient arrest of dendritic cell maturation for primate tolerance induction In nonhuman primates, following T cell depletion in lymph nodes and blood after day-of-transplant induction with anti-CD3 immunotoxin, stable tolerance to MHC mismatched renal allografts is favored by inhibition of proinflammatory cytokine responses using brief treatment with the NFKB inhibitor, 15 -deoxyspergualin. The mechanism for this synergistic effect is blockade of dendritic cell precursor maturation during the T cell recovery phase after immunotoxin induction.
Normal 3 kg male rhesus monkeys received a renal allograft from an unrelated, MHC mismatched donor. The monkeys were then treated with two injections of immunotoxin (lOOμg/kg/d) on day 0 and +2, methylprednisolone (MP) tapering from 7 mg/kg/d to zero on days 0 to +3, and deoxyspergualin (2.5 mg/kg/d) on days 0 to +4 (n=5) or 0 to +14 (n=5). Additional recipients had immunotoxin with MP alone (n=3) or MP plus Neoral (100 mg/gk d) on days 0-4 (n=3). Inguinal and axial lymph nodes were biopsied at 5, 15, and 30 days and examined by immunohistochemistry for the presence of markers of mature dendritic cells, specifically, for membrane CD83, DR, and CD86 and nuclear Rel-B.
Renal allografts survived without histologic or clinical evidence of acute or chronic rejection in 80%> of IT-treated recipients given MP plus deoxyspergualin x 15d, in 40%) in those given MP plus deoxyspergualin x 5d, and in 0%> of those given only MP or MP plus Neoral. The 3 longest deoxyspergualin survivors are >830 days (2.3 years) with normal renal function and immunologic evidence of specific tolerance. Compared to lymph nodes from transplants without deoxyspergualin, the day +5 lymph node tissue from deoxyspergualin-treated recipients showed extreme reduction in mature DC, i.e., a mean 58-fold ± 9.9 reduction in membrane expression for DC83, 49- fold ± 9.0 for CD86, and 81-fold± 13.5 for DR. Rel-B nuclear positive cells were reduced 46 fold ±7.1. Partial recovery occurred by day 15, and by day 30, DR+, CD83+, and Rel-B+ cells were within control range. CD86+ cells were still reduced (by 7.5-fold±1.8). The results indicate a dose-dependent NFKB inhibition and DCp maturation arrest by deoxyspergualin.
The data show the NFKB inhibitor, deoxyspergualin, blocks maturation of dendritic cell precursors in vivo in nonhuman primates. We hypothesize that the unusual synergy of immunotoxin and deoxyspergualin in promoting tolerance in this difficult model is due in part to depletion of memory as well as naive T cells by immunotoxin. Conceptually, this would create a transitional circumstance in which repopulating naive T cells are committed to dendritic cell costimulatory requirements for activation, while dendritic cell precursor acquisition of costimulatory function is arrested by deoxyspergualin. The net result, a window of 2-3 weeks, is a fleeting lapse into a "non-dangerous" milieu akin to the neonate's, coincidentally reduced both in T cell scope and dendritic cell precursor maturity, thereby favoring tolerance.
EXAMPLE 2
To directly examine dendritic cell precursor maturation, isolated rhesus peripheral blood dendritic cell precursors in medium containing monocyte conditioned medium (MCM) supplemented with TNFα were examined. Deoxyspergualin (0.6-10 mg/ml) was added to dendritic cell precursor cultures prior to adding MCM/TNFα.
Expression of dendritic cell maturation markers, as in Example 1, was assessed by flow cytometry and by immunocytochemistry staining of cytocentrifuge preparations.
The in vitro data showed a shift from nuclear to cytoplasmic and from membrane to cytoplasmic staining of Rel-B and CD83, respectively, reflecting a dominant immature dendritic cell precursor phenotype in the deoxyspergualin treated cultures. The in vitro results are consistent with the in vivo results of Example 1.
Example 3 Allografts were performed as in Example 1. The graft recipients were treated with either the whole IgG form of the immunotoxin or the F(Ab)2 form of the immunotoxin on days 0 and +1, also as described in Example 1. Some of the recipients also received DSG daily for either five days following transplantation or for two weeks following transplantation. Plasma levels of the Th2 cytokine IL4 and the Thl cytokine interferon gamma (LFN-γ) were assayed 1 week, 2 weeks, or 4 weeks after transplantation. Following treatment with the whole IgG immunotoxin alone, levels of IFN-γ and IL-4 increased progressively from 1 to 4 weeks with levels of IFN-γ higher than levels of IL-4. When treatment with the whole IgG immunotoxin is combined with DSG treatment, the levels of cytokines change. By the second week post transplantation, a clear IL4 polarization is shown in the groups that received the combined treatment. Specifically the groups that received combined treatments showed a substantially lower level of IL4 compared to the group treated with immunotoxin alone and substantially lower levels of IL4 compared to IFN-γ levels. The group that received DSG for two weeks showed a sustained, progressive increase in IL4 polarization at 4 weeks, whereas, in the group that received DSG for only 5 days, the polarization was no longer sustained and the cytokine pattern was reversed. These data indicated that a two-week course of DSG, in combination with immunotoxin, was necessary to achieve the effect of sustained IL4 polarization. The IL4 polarization occurred independently of the type of anti-CD3 immunotoxin, i.e., the whole IgG or F(Ab)2 form. The magnitude of the IL4 response was lower with the F(Ab)2 form, but the polarization was virtually complete inasmuch as there was no measurable interferon-gamma. See Figure 1.
Example 4 RT PCR techniques well known in the art were used with freshly obtained rhesus monkey peripheral blood lymphocytes stimulated with whole IgG immunotoxin (FN18-CRM9), F(Ab)2 immunotoxin (FN18-F(Ab)2-CRM9), or treated with phosphate buffered saline (PBS). The results of the experiment are shown in Figure 2. FN18- CRM9 induced cytokine gene expression for IL2, IL4, IL10, and LNFγ, whereas FN18- F(Ab)2-CRM9 selectively induced only IL4 and IL10 expression. In the presence of low doses of DSG (2.5 μg/ml), FN18-CRM9 induction of IL2, 4, 10 and LNFγ is reduced. In contrast, low doses of DSG in combination with FN18-F(Ab)2-CRM9, IL4 and IL10 are enhanced, which is consistent with the in vivo data in Example 3. Thus, FN18-F(Ab)2-CRM9 in combination with DSG has a unique effect of blocking LNFγ expression (consistent with the inhibitory effect of DSG on NF-κB translocation) while enhancing expression of IL4 and IL10. Thus DSG, unlike cyclosporin or FK506, preserves and increases NF-AT transcription factors activation of IL4 and IL10 but selectively inhibits NF-κB dependent cytokine activation pathways.
Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.
Although the present invention is described with reference to specific details of certain embodiments thereof, it is not intended that such details should be regarded as limitations upon the scope of the invention except as and to the extent that they are included in the accompanying claims.
REFERENCES
1. Andoins et al. (1996), Transplantation 62:1543-1549.
2. Posselt et al.(1990), Science 249:1293-1295.
3. Remuzzi et al. (1991), Lancet 337:750-752.
4. O'Doherty et al. (1997), J. of Immunol. Meth. 207:185-194.

Claims

What is claimed is:
1. A method of inducing immune tolerance to a graft in a recipient, comprising:
(a) administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population; and
(b) administering to the recipient an agent that inhibits dendritic cell maturation.
2. The method of claim 1, wherein the graft is selected from the group consisting of kidney, liver, heart, pancreas, lung, skin, and isolated cell transplants of pancreatic islets, hepatocytes, stem cell precursors, and differentiated stem cell precursors.
3. The method of claim 1, wherein the agent that inhibits dendritic cell maturation is an inhibitor of nuclear translocation of NfkB.
4. The method of claim 3, wherein the inhibitor of nuclear translocation of NfKB is an analog or derivative of deoxyspergualin.
5. The method of claim 1, wherein the agent that inhibits dendritic cell maturation activates one or more NF-AT dependent Th2 cytokines.
6. The method of claim 1, wherein the agent that inhibits dendritic cell maturation inhibits one or more NfkB dependent Thl cytokines.
7. The method of claim 1, wherein the inhibitor of dendritic cell maturation is a soluble IL-17 receptor Fc fusion protein.
8. The method of claim 1, wherein the inhibitor of dendritic cell maturation is a glucocorticoid.
9. The method of claim 1 , wherein the inhibitor of dendritic cell maturation is a blocker of tumor necrosis factor alpha binding.
10. The method of claim 1 , wherein the inhibitor of dendritic cell maturation is a blocker of granulocyte macrophage colony stimulating factor binding.
11. The method of claim 1 , wherein the inhibitor of dendritic cell maturation is a blocker of IL-12p70 binding.
12. The method of claim 1, wherein the inhibitor of dendritic cell maturation is a blocker of interleukin lβ binding.
13. The method of claim 1 , wherein the inhibitor of dendritic cell maturation is anti- CD 154 ligand.
14. The method of claim 1, wherein the agent that inhibits dendritic cell maturation is administered to the recipient at least once.
15. The method of claim 1 , wherein the agent that inhibits dendritic cell maturation is administered to the recipient prior to transplantation.
16. The method of claim 1, wherein the immunotoxin is an anti-T cell immunotoxin directed at the CD3 epitope.
17. The method of claim 1, further comprising administering to a transplant donor, prior to harvesting the transplant, an agent that inhibits dendritic cell maturation.
18. A method of screening for an agent that acts synergistically with an immunotoxin in inducing immune tolerance, comprising:
(a) transplanting a donor graft to a recipient;
(b) administering to the recipient an immunotoxin, thereby reducing the recipient's T-cell population;
(c) administering to the recipient the agent to be screened;
(d) obtaining a dendritic cell-containing sample; and
(e) determining a percentage of the dendritic cells in the sample that express or can be induced to express a marker specific for mature dendritic cells, wherein a low percentage shows a synergistic action.
19. A method of screening for an agent that inhibits dendritic cell maturation, comprising:
(a) obtaining a population of immature dendritic cells from a dendritic cell- containing sample of a subject;
(b) culturing the population of cells in the presence of the agent to be screened; and
(c) determining a percentage of dendritic cells that express or can be induced to express a marker specific for mature dendritic cells, wherein a low percentage shows inhibition of dendritic cell maturation.
20. A composition comprising an immunotoxin and an agent that inhibits dendritic cell maturation.
21. The composition of claim 18, wherein the immunotoxin is an anti-T cell immunotoxin directed at the CD3 epitope.
22. The composition of claim 19, wherein the agent that inhibits dendritic cell maturation is an inhibitor of nuclear translocation of Nf B.
23. The composition of claim 19, wherein the agent that inhibits dendritic cell maturation activates one or more NF-AT dependent Th2 cytokines.
24. The composition of claim 19, wherein the agent that inhibits dendritic cell maturation inhibits one or more NfkB dependent Thl cytokines.
PCT/US2000/010253 1999-04-14 2000-04-14 Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft WO2000061132A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CA002382628A CA2382628A1 (en) 1999-04-14 2000-04-14 Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft
JP2000610465A JP2002541195A (en) 1999-04-14 2000-04-14 A composition comprising an immunotoxin and an agent that inhibits maturation of dendritic cells for inducing immune tolerance to a graft
AU43542/00A AU781547B2 (en) 1999-04-14 2000-04-14 Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft
EP00923415A EP1171109A1 (en) 1999-04-14 2000-04-14 Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft
BR0009772-1A BR0009772A (en) 1999-04-14 2000-04-14 Compositions containing immunotoxins and agents that inhibit the maturation of dendritic cells to induce immune tolerance to a graft
HK02103064.3A HK1042648A1 (en) 1999-04-14 2002-04-24 Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29171299A 1999-04-14 1999-04-14
US09/291,712 1999-04-14

Publications (2)

Publication Number Publication Date
WO2000061132A1 true WO2000061132A1 (en) 2000-10-19
WO2000061132A8 WO2000061132A8 (en) 2001-04-19

Family

ID=23121504

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/010253 WO2000061132A1 (en) 1999-04-14 2000-04-14 Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft

Country Status (8)

Country Link
EP (1) EP1171109A1 (en)
JP (1) JP2002541195A (en)
CN (1) CN1367688A (en)
AU (1) AU781547B2 (en)
BR (1) BR0009772A (en)
CA (1) CA2382628A1 (en)
HK (1) HK1042648A1 (en)
WO (1) WO2000061132A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6632928B1 (en) 1997-03-05 2003-10-14 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxins and methods of inducing immune tolerance
US7288254B2 (en) 1995-10-30 2007-10-30 The United States Of America As Represented By The Secretary, Department Of Health And Human Services, Nih Use of immunotoxins to induce immune tolerance to pancreatic islet transplantation
WO2008043157A1 (en) 2006-10-12 2008-04-17 The University Of Queensland Compositions and methods for modulating immune responses
US7517527B2 (en) 1995-10-30 2009-04-14 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxin with in vivo T cell suppressant activity and methods of use
US7696338B2 (en) 1995-10-30 2010-04-13 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxin fusion proteins and means for expression thereof
US8507431B2 (en) 2003-08-07 2013-08-13 Healor Ltd. Methods for accelerating wound healing by administration of a preadipocyte modulator or an adipocyte modulator
US9624289B2 (en) 2012-03-16 2017-04-18 University Health Network Methods and compositions for modulating Toso activity

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003293411B2 (en) * 2002-12-06 2010-10-28 Northwest Biotherapeutics, Inc. Administration of dendritic cells partially matured in vitro for the treatment of tumors
ZA200600235B (en) * 2003-07-15 2007-04-25 Healor Ltd 14 07 06 Methods and pharmaceutical compositions for healing wounds
WO2005115419A1 (en) * 2004-05-17 2005-12-08 Board Of Trustees Of The University Of Illinois USES OF BISPECIFIC ANTIBODY (Biab) COATED DENDRITIC CELLS PULSED WITH ANTIGENS
AU2014213009B2 (en) * 2013-02-01 2019-01-03 Kira Biotech Pty Limited Anti-CD83 antibodies and use thereof
US11655293B2 (en) 2018-02-22 2023-05-23 Universitat Zurich Ligands to GM-CSF or GM-CSF-receptor for use in leukemia in a patient having undergone allo-HCT
CN115778984A (en) * 2023-02-20 2023-03-14 首都医科大学附属北京朝阳医院 Method and application for inducing transplantation immune tolerance and transplantation tolerance animal model

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736536A (en) * 1994-08-19 1998-04-07 Bristol-Myers Squibb Company Method for treating vascular leak syndrome
US5747474A (en) * 1996-07-29 1998-05-05 Immune Modulation, Inc. Immunosuppression by administration of N6,N6 -disubstituted cAMP's, analogues thereof, and related nucleosides
US5801193A (en) * 1997-04-15 1998-09-01 Immune Modulation, Inc. Compositions and methods for immunosuppressing
WO1998039363A2 (en) * 1997-03-05 1998-09-11 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Immunotoxins and methods of inducing immune tolerance

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU696775B2 (en) * 1995-03-23 1998-09-17 Kirin-Amgen, Inc. IL-17 receptor

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736536A (en) * 1994-08-19 1998-04-07 Bristol-Myers Squibb Company Method for treating vascular leak syndrome
US5747474A (en) * 1996-07-29 1998-05-05 Immune Modulation, Inc. Immunosuppression by administration of N6,N6 -disubstituted cAMP's, analogues thereof, and related nucleosides
WO1998039363A2 (en) * 1997-03-05 1998-09-11 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Immunotoxins and methods of inducing immune tolerance
US5801193A (en) * 1997-04-15 1998-09-01 Immune Modulation, Inc. Compositions and methods for immunosuppressing

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CONTRERAS JUAN L ET AL: "Peritransplant tolerance induction with anti-CD3-immunotoxin: A matter of proinflammatory cytokine control.", TRANSPLANTATION (BALTIMORE), vol. 65, no. 9, 15 May 1998 (1998-05-15), pages 1159 - 1169, XP000925746, ISSN: 0041-1337 *
ECKHOFF D E (REPRINT) ET AL: "Synergy of 15- deoxyspergualin with antiCD3 immunotoxin in tolerance induction in rhesus monkeys", TRANSPLANTATION, (15 MAY 1999) VOL. 67, NO. 9, PP. 60-60. PUBLISHER: LIPPINCOTT WILLIAMS & WILKINS, 227 EAST WASHINGTON SQ, PHILADELPHIA, PA 19106. ISSN: 0041-1337., UNIV ALABAMA, DEPT SURG, TRANSPLANT CTR, BIRMINGHAM, AL 35294, XP000933459 *
HAGGERTY H G (REPRINT) ET AL: "Effect of deoxyspergualin or CTLA4Ig on the immunogenicity and pharmacokinetics of the immunotoxin BR96sFv-PE40 in dogs.", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, (JAN 1997) VOL. 99, NO. 1, PART 2, SUPP. [S], PP. 708-708. PUBLISHER: MOSBY-YEAR BOOK INC, 11830 WESTLINE INDUSTRIAL DR, ST LOUIS, M 63146-3318. ISSN: 0091-6749., BRISTOL MYERS SQUIBB, SYRACUSE, NY;BRISTOL MYERS SQUIBB, NEW BRUNSWICK, NJ, XP000925712 *
SIEGALL C.B. ET AL: "Prevention of immunotoxin -mediated vascular leak syndrome in rats with retention of antitumor activity.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, (1994) 91/20 (9514-9518)., XP000929449 *
THOMAS JUDITH M ET AL: "Peritransplant tolerance induction in macaques: Early events reflecting the unique synergy between immunotoxin and deoxyspergualin.", TRANSPLANTATION (BALTIMORE), vol. 68, no. 11, 15 December 1999 (1999-12-15), pages 1660 - 1673, XP000925747, ISSN: 0041-1337 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7288254B2 (en) 1995-10-30 2007-10-30 The United States Of America As Represented By The Secretary, Department Of Health And Human Services, Nih Use of immunotoxins to induce immune tolerance to pancreatic islet transplantation
US7517527B2 (en) 1995-10-30 2009-04-14 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxin with in vivo T cell suppressant activity and methods of use
US7696338B2 (en) 1995-10-30 2010-04-13 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxin fusion proteins and means for expression thereof
US8217158B2 (en) 1995-10-30 2012-07-10 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Immunotoxin fusion proteins and means for expression thereof
US8987426B2 (en) 1995-10-30 2015-03-24 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Immunotoxin fusion proteins and means for expression thereof
US6632928B1 (en) 1997-03-05 2003-10-14 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxins and methods of inducing immune tolerance
US8507431B2 (en) 2003-08-07 2013-08-13 Healor Ltd. Methods for accelerating wound healing by administration of a preadipocyte modulator or an adipocyte modulator
WO2008043157A1 (en) 2006-10-12 2008-04-17 The University Of Queensland Compositions and methods for modulating immune responses
EP3590503A1 (en) 2006-10-12 2020-01-08 The University of Queensland Compositions and methods for modulating immune responses
US9624289B2 (en) 2012-03-16 2017-04-18 University Health Network Methods and compositions for modulating Toso activity
US9670265B2 (en) 2012-03-16 2017-06-06 University Health Network Methods and compositions for modulating toso activity
US10822388B2 (en) 2012-03-16 2020-11-03 University Health Network Methods and compositions for modulating toso activity

Also Published As

Publication number Publication date
CA2382628A1 (en) 2000-10-19
HK1042648A1 (en) 2002-08-23
BR0009772A (en) 2002-01-08
CN1367688A (en) 2002-09-04
AU781547B2 (en) 2005-05-26
JP2002541195A (en) 2002-12-03
AU4354200A (en) 2000-11-14
WO2000061132A8 (en) 2001-04-19
EP1171109A1 (en) 2002-01-16

Similar Documents

Publication Publication Date Title
First TRANSPLANTATION IN THE NINETIES1
US6718986B2 (en) Mixed chimerism and tolerance
AU781547B2 (en) Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft
EP0621786B1 (en) Induced tolerance to xenografts
JP2002500648A (en) Use of a CD40: CD154 binding blocker to prevent an anti-adaptive immune response, especially graft rejection
Gruber The case for local immunosuppression
BG64976B1 (en) Cd154 blockade therapty in tissue transplantation of pancreatic isles
JP2002504120A (en) Compositions and methods for preventing graft rejection and anti-adaptive T lymphocyte mediated immune response
WO2002040640A2 (en) Methods of using cd8+/tcr- facilitating cells (fc) for the engraftment of purified hematopoietic stem cells (hsc)
WO2006052660A9 (en) Il-7 receptor blockade to suppress immunity
Wekerle et al. Tolerance through bone marrow transplantation with costimulation blockade
US20040228845A1 (en) Methods of using CD8+/TCR- facilitating cells (FC) for the engraftment of purified hematopoietic stem cells (HSC)
Akita et al. Effect of FK506 and anti-CD4 therapy on fetal pig pancreas xenografts and host lymphoid cells in nod/Lt, CBA, and BALB/c mice
Bigaud et al. NVP-AEB071 (AEB), THE NOVEL ORAL INHIBITOR OF PROTEIN KINASE C (PKC) AND EARLY T-CELL ACTIVATION, PROLONGS NON-HUMAN PRIMATES (NHP) KIDNEY ALLOGRAFT SURVIVAL WHEN COMBINED WITH EVEROLIMUS (RAD), ERL080 (ERL) OR FTY720 (FTY) WITHOUT CALCINEURIN INHIBITOR (CNI).
US20060018885A1 (en) Methods for increasing HSC graft efficiency
Xu et al. A delay in bone marrow transplantation after partial conditioning improves engraftment1
WO1995021527A1 (en) Stem cell engraftment
EP0697876B1 (en) Xenograft thymus
den Dulk et al. Combined donor leucocyte administration and immunosuppressive drug treatment for survival of rat heart allografts
US20050214265A1 (en) Method for improving bone marrow engraftment
US20040146503A1 (en) Methods and compositions for inducing stable immune tolerance
EP1048298B1 (en) Mixed chimerism and tolerance
Qian et al. PG695 IS EFFECTIVE AS A MONOTHERAPY AND ACTS SYNERGISTICALLY WITH FK506. PART I: PROLONGING RENAL ALLOGRAFT SURVIVAL IN NON-HUMAN PRIMATES.
Monaco Induction of specific unresponsiveness (tolerance) to experimental and clinical allografts using polyclonal antilymphocyte serum and donor-specific bone marrow
Horner Overcoming skin rejection in composite tissue allotransplantation

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 00808770.9

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: C1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i
ENP Entry into the national phase

Ref document number: 2000 610465

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2382628

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2000923415

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2000923415

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09959006

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWR Wipo information: refused in national office

Ref document number: 2000923415

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2000923415

Country of ref document: EP