WO1997005896A1 - Combined use of interleukin-10 and cyclosporin for immunosuppression therapy - Google Patents

Combined use of interleukin-10 and cyclosporin for immunosuppression therapy Download PDF

Info

Publication number
WO1997005896A1
WO1997005896A1 PCT/US1996/012538 US9612538W WO9705896A1 WO 1997005896 A1 WO1997005896 A1 WO 1997005896A1 US 9612538 W US9612538 W US 9612538W WO 9705896 A1 WO9705896 A1 WO 9705896A1
Authority
WO
WIPO (PCT)
Prior art keywords
interleukin
cells
csa
cyclosporin
mlr
Prior art date
Application number
PCT/US1996/012538
Other languages
English (en)
French (fr)
Other versions
WO1997005896B1 (en
Inventor
Waleed Tadmori
Original Assignee
Schering Corporation
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 Schering Corporation filed Critical Schering Corporation
Priority to HU9903019A priority Critical patent/HUP9903019A3/hu
Priority to EP96928798A priority patent/EP0845994A1/en
Priority to JP9508515A priority patent/JPH11510806A/ja
Priority to MX9801091A priority patent/MX9801091A/es
Priority to AU68422/96A priority patent/AU712606B2/en
Publication of WO1997005896A1 publication Critical patent/WO1997005896A1/en
Publication of WO1997005896B1 publication Critical patent/WO1997005896B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • the invention relates to a method of suppressing graft/tissue rejection, graft-versus-host disease and autoimmune diseases.
  • the invention relates to the combined use of interleukin 10 and cyclosporin for immunosuppression therapy.
  • Interleukin 10 a cytokine produced by T lymphocytes, was first identified by its ability to inhibit interferon gamma (IFN- ⁇ ) and IL-2 synthesis by mouse and human T lymphocytes. Fiorentino etal., 1989, J. Exp. Med. 170:2081-2089; Moore et al., 1990, Science 248: 1230-1252; Vieira et al., 1991 , Proc. Natl. Acad. Sci. USA 88:1172-1177. IL-10 was subsequently shown to be produced by B cells (O'Garra et al., 1990, Intemat. Immunol. 2:821-828) and macrophages (Fiorentino etal., 1991, J. Immunol. 147:3815- 3822).
  • IL-10 exerts a wide range of effects on a variety of cell types. IL-10 inhibits the synthesis of a wide spectrum of cytokines produced by T cells and monocytes. In addition to inhibiting the synthesis of IFN- ⁇ and IL-2, IL-10 has also been shown to inhibit production of the monokines IL-1 ⁇ , IL-1 ⁇ , IL-6 and TNF ⁇ . de Waal et al., 1991 , J. Exp. Med. 174:1209-1217. IL-10 has growth promoting effects on murine thymocytes and T cells (MacNeil et al., 1990, Immunol. 145:4167) and mast cells (Thompson-Snipes etal., 1991, J. Exp. Med. 173:507-512), and it stimulates cytotoxic T-cell development (Chen and Zlotnik, 1991 , J. Immunol. 147:528-533).
  • Mouse and human IL-10 have high sequence similarity with a protein encoded by an open reading frame in the Epstein-Barr Virus.
  • the expression product of this open reading frame named viral IL-10, also has the capacity to inhibit cytokine synthesis. Moore etal., 1990, Science 248:1230-1252; Vieira etal., 1991 , Proc. Natl. Acad. Sci. USA 88:1172-1177.
  • cytokines including IL-2, IFN- ⁇ and TNF- ⁇
  • MLR mixed lymphocyte reaction
  • IFN- ⁇ may pay an important role in MLR graft rejection. Novelli etal., 1991 , J. Immunol. 147:1445-1450; Landolfo etal., 1985, Science 229:176-180. Antibodies to IFN- ⁇ or to TNF (Shalaby etal., 1988, J. Immunol. 141:499-505) have been shown to block MLR-induced proliferation. In these studies it was found that antibodies to IFN- ⁇ suppressed the MLR in human systems as well as allograft reactivity in vitro and in vivo in the mouse.
  • Cyclosporin also known as cyclosporin A; CSA
  • CSA cyclosporin A
  • CSA cyclic peptide produced by the fungus Tolypocladium inflatum Gams and other fungi imperfecti
  • IL-2 production by cyclosporin Shevach, 1985, Annu. Rev. Immunol. 3:397; Fidelus etal., 1982, Transplantation 34:308), or an antibody of CD2 (Tadmori etal., 1985, J. Immunol. 134:4542-4550) depresses T-cell proliferation
  • this invention provides a method for suppressing tissue or organ rejection comprising administering an effective amount of interleukin 10 and cyclosporin to a patient experiencing or at risk of tissue graft rejection.
  • This invention further provides a method for suppressing graft-versus- host disease comprising administering an effective amount of interleukin 10 and cyclosporin to a patient afflicted with or at risk for graft-versus-host disease.
  • this invention provides a method for treating autoimmune diseases comprising administering an effective amount of interleukin 10 and cyclosporin to a patient afflicted with an autoimmune disease.
  • compositions comprising a combination of IL-10 and CSA are also provided by this invention. Brief Description of the Drawing Figure
  • the accompanying drawing Figure demonstrates synergistic suppression of T cell proliferation in MLR by a combination of human IL-10 and cyclosporin.
  • a failure of major organs is a principal cause of disease and death in mammals.
  • normal bodily immune defense mechanisms recognize such organ transplants as foreign and attack them, resulting in graft failure and rejection.
  • a major impediment to transplantation of allogeneic tissues and organs is graft rejection by the transplant recipient.
  • the cell-mediated immune response of the recipient, or host, to the donor tissue plays an important role in the rejection process.
  • This response has two important phases: (i) recognition of the donor cells or tissue as "foreign” in the context of the major histocompatibility complex (MHC); and (ii) destruction of the foreign cells by the host cells.
  • MHC major histocompatibility complex
  • a number of host cells undergo proliferation and acquire cytotoxicity - that is, the ability to kill donor cells displaying the appropriate antigens.
  • cytotoxicity can be described in terms of two measurable functions: proliferation, and cytotoxic activity -- see Dubey etal., chapter 131 in Rose et al., Editors, "Manual of Clinical Laboratory Immunology", 3d edition (American Society of Microbiology, Washington, D.C, 1986).
  • MLR mixed lymphocyte reaction
  • MLR comprises co-culturing a sample of responder cells and a sample of inactivated stimulator cells such that the stimulator cells are allogeneic with respect to the responder cells (i.e., the stimulator cells are obtained from a different person from that from whom the responder cells are taken), and measuring the proliferative response of the responder cells.
  • MLR consists of mixing responder lymphocytes (mimics host cells) in a suitable culture system with stimulator lymphocytes (mimics donor cells), the proliferation and/or transcription machinery of which has been disabled, e.g. by irradiation or treatment with a DNA synthesis inhibitor (e.g., mitomycin C) or the like.
  • the stimulator cells are inactivated so that they can still carry out their stimulatory function but are inhibited from any other functions that could obscure the response measured from the responder cells, i.e., the stimulator cells are treated so that they are incapable of replication, but their antigen processing machinery remains functional.
  • the response measured in the responder cells is cellular proliferation.
  • Proliferation of the responder cells may be determined by the uptake of tritiated thymidine using standard protocols. For example, from 2.5 - 10 x 10 4 stimulator cells are added to 2.4 x 10 4 allogeneic CD4+ (responder) cells in 96-well round-bottom tissue- culture plates and are incubated for 4 days in an appropriate medium.
  • the cells are pulsated with 1 ⁇ Ci of tritiated thymidine for 6 hours, and then they are harvested and measured for tritiated thymidine uptake, e.g., by scintillation counting.
  • tritiated thymidine uptake e.g., by scintillation counting.
  • I L- 10 and CSA can advantageously be used in the suppression of pathology associated with T cell responses, in particular, autoimmune diseases, graft-versus-host disease (GVHD) and tissue graft rejection.
  • the invention can be used to suppress cell-mediated reactions such as allograft rejection and GVHD.
  • IL-10 and CSA may support GVL (graft-versus-leukemia) in allogeneic bone marrow transplants.
  • the invention may be used to prevent the rejection or prolong the survival of allogeneic transplants of skin, heart, kidney, pancreas, bone marrow, small intestine, lung, etc.; to treat autoimmune diseases such as, for example, rheumatoid arthritis, lupus, diabetes mellitus, multiple sclerosis and myasthenia gravis; and to treat other diseases where CSA has been used, such as psoriasis. Due to the activity of IL-10, CSA can by used in lower amounts, thereby avoiding or reducing the serious side effects normally associated with the use of this drug.
  • Transplant recipients may be recipients of kidney, liver, heart, heart- lung, bone marrow, cornea transplant, etc.
  • the transplanted tissue itself is typically human in origin but may also be from another species such as a rhesus monkey, baboon or pig.
  • tissue includes individual cells, such as blood cells, including progenitors and precursors thereof, and pancreatic ceils, as well as solid organs and the like.
  • solid organ means a heart, skin, a liver, a lung, a kidney, a pancreas, an intestine, endocrine glands, a bladder, skeletal muscles, etc.
  • the methods of the invention can be used prophylactically or for treatment of established autoimmune disease, GVHD or graft rejection.
  • Individuals suitable for treatment by the methods of the invention include any individual at risk (predisposed) for developing GVHD or tissue rejection, i.e., a transplant patient, or an individual exhibiting clinical symptoms.
  • Prophylactic use encompasses administration prior to transplantation as well as post- transplantation administration in the absence of any clinical symptoms of GVHD or graft rejection, to prevent or postpone onset of disease/rejection.
  • IL-10 and CSA are to be "concurrently" administered to a patient.
  • Concurrently administering means the IL-10 and CSA are administered to the subject either (a) simultaneously in time (optionally by formulating the two together in a common carrier), or (b) at different times during the course of a common treatment schedule. In the latter case, the two compounds are administered sufficiently close in time to achieve the intended effect.
  • one agent is administered within about the half-life of the first agent, the two agents are considered to be concurrently administered.
  • the active agents may be administered together in a single pharmaceutical composition or separately. Both active agents (i.e., IL-10 and CSA) should be present in the patient at sufficient combined levels to be therapeutically effective.
  • the routes of administration of the IL-10 and CSA may be the same or different.
  • IL-10 and CSA are administered as a pharmaceutical composition comprising an effective amount of IL-10 and CSA in a pharmaceutical carrier.
  • a pharmaceutical carrier can be any compatible, non ⁇ toxic substance suitable for delivering the compositions of the invention to a patient.
  • interleukin 10 or IL-10 is defined as a protein which (a) has an amino acid sequence substantially identical to a known sequence of mature (i.e., lacking a secretory leader sequence) IL-10 as disclosed in International Application Publication No. 91/003249, and (b) has biological activity that is common to native IL-10.
  • glycosylated e.g., produced in eukaryotic cells such as yeast or CHO cells
  • unglycosylated e.g., chemically synthesized or produced in E. coli
  • muteins and other analogs including viral IL-10, which retain the biological activity of IL-10.
  • IL-10 suitable for use in the invention can be obtained from a number of sources. For example, it can be isolated from culture media of activated T-cells capable of secreting the protein. Additionally, the IL-10 or active fragments thereof can be chemically synthesized using standard techniques known in the art. See, e.g., Merrifield, 1986, Science 233:341 -347 and Atherton etal., Solid Phase Peptide Synthesis, A Practical Approach, 1989, IRL Press, Oxford.
  • the protein or polypeptide is obtained by recombinant techniques using isolated nucleic acids encoding the IL-10 polypeptide.
  • General methods of molecular biology are described, e.g., by Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, , 2d Ed., Cold Spring Harbor, New York and Ausubel et al. (eds). Current Protocols in Molecular Biology, Green/Wiley, New York (1987 and periodic supplements).
  • the appropriate sequences can be obtained using standard techniques from either genomic or cDNA libraries.
  • DNA constructs encoding IL-10 may also be prepared synthetically by established standard methods, e.g., in an automatic DNA synthesizer, and then purified, annealed, ligated and cloned in suitable vectors. Atherton etal., 1989. Polymerase chain reaction (PCR)
  • SUBSTITUTE SHEET (RULE 26) techniques can be used. See e.g., PCR Protocols: A Guide to Methods and Applications, 1990, Innis etal. (ed.), Academic Press, New York.
  • the DNA constructs may contain the entire native sequence of IL-10 or a homologue thereof.
  • the term "homologue" is intended to indicate a natural variant of the DNA sequence encoding IL-10 or a variant or fragment produced by modification of the DNA sequence.
  • suitable modifications of the DNA sequence are nucleotide substitutions which do not give rise to another amino acid sequence or nucleotide substitutions which do give rise to a different amino acid sequence and therefore, possibly, a different protein structure.
  • Other examples of possible modifications are insertions of one or several nucleotides into the sequence, addition of one or several nucleotides at either end of the sequence, or deletion of one or several nucleotides at either end or within the sequence.
  • Any homologous DNA sequence encoding a protein which exhibits IL-10 activity e.g., with respect suppression of T cell proliferation
  • modified IL-10 can vary from the naturally- occurring sequence at the primary level, e.g., by amino acid insertions, substitutions, deletions and fusions.
  • amino acid substitutions will be conservative; i.e., basic amino acid residues will be replaced with other basic amino acid residues, etc.
  • Amino acid sequence variants can be prepared with various objectives in mind, including increasing serum half-life, facilitating purification or preparation, improving therapeutic efficacy, and lessening the severity or occurrence of side effects during therapeutic use.
  • the amino acid sequence variants are usually predetermined variants not found in nature, although others may be post-translational variants, e.g., glycosylation variants or proteins which are conjugated to polyethylene glycol (PEG), etc.
  • PEG polyethylene glycol
  • human IL-10 is used forthe treatment of humans, although viral or mouse IL-10, or IL-10 from some other mammalian species, could be used instead. Most preferably, the IL-10 used is recombinant human IL-10. Recombinant production of human IL-10 is described in U.S. Patent No. 5,231 ,012. Preparation of human and mouse IL-10 has been described in Intemational Application Publication No. WO 91/00349. The cloning and expression of viral IL-10 (BCRFI protein) from Epstein Barr virus has been disclosed by Moore et al. (Science 248:1230, 1990), and is described in EP 0 506 836.
  • BCRFI protein viral IL-10
  • CSA may be is administered in a manner as is conventionally practiced. See, e.g., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 7th Ed , 1985, p. 1299.
  • CSA may be provided as an oral solution of 100 mg/ml with 12.5% alcohol, and for intravenous administration as a solution of 50 mg/ml with 33% alcohol and 650 mg of polyoxyethlated castor oil.
  • CSA When administered intravenously, CSA may be given as a dilute solution of 50 mg to 20 - 100 mg of normal saline or 5 % dextrose in water, by slow infusion over a period of several hours.
  • the intravenous dose is typically one third of the oral dose.
  • CSA is orally, either in capsule or tablet form.
  • Such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and a suitable carrier (which may contain one or more accessory ingredients).
  • the formulations can be prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture.
  • a tablet may be prepared by compressing or molding a powder or granules containing the active compound, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules containing the active compound, optionally mixed with a binder, lubricant, inert diluent, and/or surface active dispersing agent(s). Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
  • CSA is disclosed in U.S. Patent No. 4,117,118.
  • CSA which may be used in the practice of the invention is commercially available under the name SANDIMMUNE® from Sandoz Pharmaceuticals Co ⁇ oration.
  • Synergistic suppression of T cell proliferation may also be observed using IL-10 and an analogue of CSA.
  • a "CSA analogue” is meant to include synthetic analogues as well as any agent which exhibits the same activity/mechanism of action as CSA.
  • agents include, for example, FK-506.
  • FK-506 is a macrolide immunosuppressant isolated from Streptomyces tsuk ⁇ baenis no. 9993. EP 0 184 162 (Fujisawa).
  • IL-10 is preferably parenteral by intraperitoneal intravenous, subcutaneous or intramuscular injection of infusion or by an other acceptable systemic method. Administration by intramuscular or subcutaneous injection is most preferred.
  • the IL-10 may be administered by an inplantable or injectable drug delivery system. See, e.g., Urquhart et al, 1984, Ann Rev. Pharacol. Toxicol 24: .99; Lewis, ed., 1981 , Controlled Release of Pesticides and Pharmaceuticals, Plenum Press, New York, New York: U.S. Patent Nos. 3,773,919, and 3,270,960.
  • Oral administration may also be carried out, using well known formulations which protect the IL-10 from gastrointestinal proteases.
  • compositions useful for parenteral administration of such drugs are well known. See, e.g., Remington's Pharmaceutical Science, 11th Ed., 1990, Mack Publishing Co., Easton, PA.
  • the IL-10 is typically formulated in a unit dosage injectable form (solution, suspension, emulsion) in association with a pharmaceutical carrier.
  • a pharmaceutical carrier examples include normal saline, Ringer's solution, dextrose solution, and Hank's solution.
  • Non-aqueous carriers such as fixed oils and ethyl oleate may also be used.
  • a preferred carrier is 5% dextrose/saline.
  • the carrier may contain minor amounts of additives such as substances that enhance isotonicity and chemical stability, e.g., buffers and preservatives.
  • the IL-10 is preferably formulated in purified form substantially free of aggregates and other source proteins at a concentration in the range of about 5 to 20 ⁇ g/ml. Any of the well known carrier proteins such as human serum albumin can also be added if desired.
  • IL-10 can also be delivered by standard gene therapy techniques, including e.g., direct DNA injection into tissues, the use of recombinant viral vectors or phospholipid and implantation of transfected cells. See, e.g., Rosenberg, 1992, J. Clin. Oncol. 10:180.
  • IL-10 and CSA are concurrently administered to a human patient in an amount effective to provide an immunosuppressive effect.
  • effective amount means an amount sufficient to reduce or prevent GVHD , an autoimmune disease or tissue rejection, and refers to the combined effects of the two agents working in concert.
  • One or both agents may, for example, be used at a dose which , if used alone, would be considered suboptimal for the intended purpose.
  • the effective amount for a particular patient may vary depending on such factors as the state, type, and amount of tissue transplanted, the overall health and age of the patient, the route of administration, the severity of observed side-effects, and the like.
  • the effective dose of IL-10 typically will range from about 0.1-25 ⁇ g/kg/day, preferably about 1 -16 ⁇ g/kg/day.
  • the effective dose of CSA typically will range of from about 1-14 mg/kg/day, more preferably from about 1-8
  • administration is to begin simultaneously with transplantation, or 2 to 4 hours before transplantation. Administration may, however, begin within the 24-hour period preceding transplantation or within the 24-hour period following transplantation. It is also contemplated that administration can be started at any time after transplantation to replace or supplement other compounds being administered to a patient to prevent graft rejection. The length of administration may vary and, in some cases, may continue over the remaining lifetime of a patient, to control graft rejection processes.
  • ELISA kits for cytokine production determination were purchased from R&D systems, Minneapolis, MN. Human PBMC and Monocyte Purification
  • peripheral blood monocytes were prepared by incubating PBMC in medium supplemented with 10% fetal bovine serum (FBS) and allowing adherence for 1 hour and 37 °C in 5% CO2 atmosphere in T-75 flasks each containing 10 7 PBMC. After removing the nonadherent cells, the flasks were extensively washed with warm medium, then incubated with cold PBS on ice for 15 minutes. Adherent monocytes were subsequently recovered by repeated pipetting, washed and resuspended in complete medium. Cell purity determined by staining with CD14 monoclonal antibodies and flow microfluorometric (FMF) analysis was 92% CD14+. Viability determined by trypan blue exclusion was >95%.
  • FBS fetal bovine serum
  • the stimulator PBMC were treated with 50 ⁇ l mitomycin C (Sigma Chemical Co., St. Louis, MO) for 20 minutes at 37 °C.
  • the responder PBMC and the stimulator cells were added to a 96-well microtiter plate (Becton Dickinson, Lincoln Park, NJ) at 1 x 10 5 cells per well of each, along with cytokines or antibodies in a total volume of 200 ⁇ l in triplets.
  • the cultures were incubated at 37 °C with 5 % CO2 in air for 6 days.
  • the cultures were then pulsed with 1 ⁇ Ci [ 3 H]TdR (15.6CI/mmol, NEN, Boston, MA) per well for 16 hours.
  • the cells were harvested onto a filter using a 96-well cell harvestor (Skatron, Inc., Sterling, VA) and counted on a beta counter (Pharmacia LKB Nuclear Inc., Gaithersburg, MD). Immunofluorescence and Flow Cvtometry
  • the supematant from the MLR was removed and the non-adherent PBMC harvested.
  • the adherent cells were harvested by incubation with 5 mM EDTA at 4 °C for 20 minutes and gently scraped. Cells were combined, centrifuged at 300 x g for 10 minutes and washed 3 times in ice-cold PBS. The viable cells were enumerated by Trypan Blue exclusion using a Neubaeur counting chamber, resuspended to 1 x 10 7 cell/ml in PBS and 100 ⁇ l of human IgG (1 mg/ml) was added to each well and incubated on ice.
  • PBMC from human blood were collected in heparinated tubes by FICOLL gradient.
  • the blood was diluted 1 :1 with PBS.
  • 30 ml of the diluted blood was laid on top of 20 ml Ficoll in a 50 ml conical tube and spun at 2,000 ⁇ m for 30 minutes.
  • the interface cells were collected and washed 3 times with medium (10% FCS/RPMI).
  • the plastic adherent antigen presenting cells were removed by resuspended PBMC in 1% human serum (Type AB) at 1x10 6 cells/ml, and incubated at 37 °C for 30 min. The cells which floated were collected and placed in media (10% FCS/RPMI).
  • Nylon wool adherent APC were removed as follows: A 10 ml syringe was packed with 1.5 g of nylon wool (Polysciences, Inc., Cat. # 18369) and autoclaved. A 3-way stopcock and a 22-gauge needle were attached to the syringe, and the column was washed with 50 ml of prewarmed medium. A cell suspension of 5x10 7 cells/ml of plastic adherent APC was prepared in prewarmed medium. Just before adding the cell suspension, the wool was rinsed with 5 ml of prewarmed medium, the column was left to run dry, and the stopcock was closed. Cells (5-10x10 7 ) were added. The cells were allowed to penetrate the column, and the stopcock was closed. An additional 0.5 ml of medium was added and the column maintained at 37 °C for 45 minutes. Nonadherent cells were collected into a tube by washing the column with 20 ml of prewarmed medium, eluted at 1 drop per second.
  • IL-10 IL-10
  • Th 1 type IL-2, IFN
  • monocytes TNF- ⁇
  • GVHD graft acceptance and graft versus host disease
  • PBMC peripheral blood mononuclear cells
  • IL-10 (100-200 U/ml) strongly suppressed MLR-induced proliferation.
  • the allogeneic stimulation induced a strong proliferative response and the suppression by IL-10 ranged between 65% and 100% regardless of the stimulation index of the MLR.
  • Table 1 shows that a monoclonal antibody to IL-10 neutralized IL-10-induced suppression of MLR. The suppression observed in these cultures was attributable to IL-10, since the addition of neutralizing monoclonal antibody, but not its isotypic control, reversed the IL-10 suppressive effect .
  • MLR + IL-10 + rat.lgGI 486 ⁇ 14427 a MLR cultures were set up as described above.
  • Human IL-10 (lOOU/ml), 19B1 (a rat monoclonal antibody to IL-10) and its isotypic control rat IgGI were used at 2.5 U g/ml.
  • D Data are 3 HTdR uptake M ⁇ SD of triplicate determination and are representative of 3 experiments.
  • IL-10 suppresses activation of human T-cell clones induced by specific soluble antigens presented on normal monocytes but not when the antigens were presented by EBV-LCL B-cell Iines. de Waal etal.., 1991, J. Exp. Med. 174:915-924. In these studies it was not clear whether the inability of IL-10 to suppress T-cell activation when EBV-LCLs were used was due to the fact they were B cells or to the possibility that these cells represented a different subpopulation, and/or that they were EBV-transformed B cells.
  • IL-10 to suppress the proliferation in MLR when allogeneic PBMC, purified B cells, monocytes or B cell Iines were used as stimulators was studied.
  • Table 2 shows that IL-10 suppresses proliferation in MLR when normal B cells, but not B-cell Iines, are used as stimulators.
  • PBMC+Daudi 37357 ⁇ 4497 45763 ⁇ 3089 14167 ⁇ 2762 a MLR cultures were set up between PBMC (1x10 ⁇ /well) and mitomycine treated B cell line
  • Human IL-10 was used at 200 U/ml and cyclosporin A 40 ng/ml.
  • b Data are (3)HTdR uptake M ⁇ SD of triplicate determination and are representative of 3 experiments.
  • IL-10 strongly suppresses the MLR-induced proliferation when purified normal B cells (985% CD20+) or monocytes (98% CD14+) were used as stimulators and PBMC as responder cells.
  • IL-10 did not suppress the reaction when B cell Iines (Daudi or JY), were uses as stimulators of the MLR.
  • B cell Iines Daudi or JY
  • Higher doses of IL-10 up to 1000 U/ml did not suppress JY-induced MLR.
  • PBMC + PBMC + IL-10 ⁇ 25 ⁇ 50 ⁇ 25 a The MLR cultures were set up as described above using PBMC (1 x 10 6 /ml) as responder and mitomycin C-treated JY cells (5 x 10 5 cell/) or allogeneic PBMC ( 5 x 10 5 ) as stimulators in a 24-well plate for 60 hours. IL-10 was used at 100 U/ml and CSA at 40 ng/ml.
  • SD triplicate determination
  • Table 3 shows that high levels of cytokines were detectable in the supernatants of MLR induced by JY.
  • this data shows that levels of IL-2 and IFN- ⁇ but not that of TNF- ⁇ , are depressed in the supernatants of MLR set up with IL-10.
  • CSA inhibited TNF- ⁇ production. Since supernatants of cultures of JY cells alone (treated or untreated with mitomycin C) did not contain detectable TNF- ⁇ , the source of TNF- ⁇ in these MLRs could be attributed to the activated responder PBMC.
  • CSA Since CSA also has a cytokine-synthesis inhibition ability and currently being used in most immunosuppressive protocols in organ transplantation, the activity of CSA was compared to that of IL-10.
  • hlL-10 was found to be a more potent inhibitor than CSA (IC50: 8pM and 4nM respectively). Moreover, it was found that the addition of combinations of hlL-10 and CSA at low doses caused synergistic suppression of T cell proliferation in MLR.
  • IL-10 and CSA synergistically inhibited T cell activation in MLR. This synergy was observed at lower doses of hlL-10 and CSA. From this data it can be observed that the administration of a combination of IL-10 and CSA is a more effective suppressive therapy than either IL-10 or CSA individually. Effect of IL-10 and CSA on Direct and Indirect Allostimulation
  • Allostimulation of T cell can be exerted via two pathways; direct allostimulation and indirect allostimulation.
  • direct allostimulation means the response of the recipient T cell to alloantigens as intact MHC molecules on the surface of allogeneic stimulator cells in the graft. Direct allostimulation is the cause of acute graft rejection and is the principal contributor to antigraft cytotoxic T cell response mediating early rejection episodes.
  • Indirect allostimulation as used herein means the response of the recipient T cell graft major histocompatibility (MHC) alloantigens that have been processed and presented by the recipients' APC. Indirect allostimulation the cause of chronic allograft rejection and antigraft antibody production, xenograft rejection, and induction of unresponsiveness (tolerance).
  • MHC major histocompatibility
  • Human IL-10 was found to be more effective than CSA in suppressing both the direct allostimulation and the indirect allostimulation of T cells. Since hlL-10 suppresses both the direct and the indirect T cell allostimulation, a combination of IL-10 and CSA can be used to prevent acute graft rejection (mediated by the direct allostimulation), and chronic graft and xenograft rejedction (mediated by the indirect allostimulation).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Transplantation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
PCT/US1996/012538 1995-08-09 1996-08-06 Combined use of interleukin-10 and cyclosporin for immunosuppression therapy WO1997005896A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
HU9903019A HUP9903019A3 (en) 1996-08-06 1996-08-06 Combined use of interleukin-10 and cyclosporin for immunosuppression therapy
EP96928798A EP0845994A1 (en) 1995-08-09 1996-08-06 Combined use of interleukin-10 and cyclosporin for immunosuppression therapy
JP9508515A JPH11510806A (ja) 1995-08-09 1996-08-06 免疫抑制治療のためのインターロイキン10およびシクロスポリンの併用
MX9801091A MX9801091A (es) 1995-08-09 1996-08-06 Uso combinado de interleukina-10 y ciclosporina para terapia de inmunosupresion.
AU68422/96A AU712606B2 (en) 1995-08-09 1996-08-06 Combined use of interleukin-10 and cyclosporin for immunosuppression therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US205795P 1995-08-09 1995-08-09
US60/002,057 1995-08-09
US61995896A 1996-03-20 1996-03-20
US08/619,958 1996-03-20

Publications (2)

Publication Number Publication Date
WO1997005896A1 true WO1997005896A1 (en) 1997-02-20
WO1997005896B1 WO1997005896B1 (en) 1997-03-27

Family

ID=26669861

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/012538 WO1997005896A1 (en) 1995-08-09 1996-08-06 Combined use of interleukin-10 and cyclosporin for immunosuppression therapy

Country Status (5)

Country Link
EP (1) EP0845994A1 (ja)
JP (1) JPH11510806A (ja)
AU (1) AU712606B2 (ja)
CA (1) CA2228379A1 (ja)
WO (1) WO1997005896A1 (ja)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999030730A1 (en) * 1997-12-15 1999-06-24 Universite Laval Methods and compositions for improving the success of cell transplantation in a host
WO2000016796A1 (en) * 1998-09-18 2000-03-30 Applied Research Systems Ars Holding N.V. Chemokine receptor antagonist and cyclosporin in combined therapy
WO2015063613A2 (en) 2013-11-01 2015-05-07 Spherium Biomed S.L. Inclusion bodies for transdermal delivery of therapeutic and cosmetic agents

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3135532B2 (ja) 1998-09-29 2001-02-19 松下電送システム株式会社 ネットワークファクシミリ装置
US6577712B2 (en) 2000-04-07 2003-06-10 Telefonaktiebolaget Lm Ericsson (Publ) Distributed voice mail system
AU2002952492A0 (en) * 2002-11-06 2002-11-21 Cbio Limited Chaperonin 10 immunosuppression

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993017698A1 (en) * 1992-03-04 1993-09-16 Schering Corporation Use of interleukin-10 to suppress graft-vs.-host disease
WO1994008606A1 (en) * 1992-10-01 1994-04-28 Schering Corporation Use of il-10 to prevent or treat insulin-dependent diabetes mellitus
WO1994017773A2 (en) * 1993-02-01 1994-08-18 Université Libre de Bruxelles Use of a pharmaceutical composition comprising an effective amount of interleukin-10, an analog and/or an agonist of interleukin-10

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993017698A1 (en) * 1992-03-04 1993-09-16 Schering Corporation Use of interleukin-10 to suppress graft-vs.-host disease
WO1994008606A1 (en) * 1992-10-01 1994-04-28 Schering Corporation Use of il-10 to prevent or treat insulin-dependent diabetes mellitus
WO1994017773A2 (en) * 1993-02-01 1994-08-18 Université Libre de Bruxelles Use of a pharmaceutical composition comprising an effective amount of interleukin-10, an analog and/or an agonist of interleukin-10

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
TADMORI W ET AL: "Suppression of the allogeneic response by human IL - 10: A critical role for suppression of a synergy between IL-2 and TNF-alpha.", CYTOKINE 6 (5). 1994. 462-471, XP000611817 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999030730A1 (en) * 1997-12-15 1999-06-24 Universite Laval Methods and compositions for improving the success of cell transplantation in a host
WO2000016796A1 (en) * 1998-09-18 2000-03-30 Applied Research Systems Ars Holding N.V. Chemokine receptor antagonist and cyclosporin in combined therapy
EP1000626A1 (en) * 1998-09-18 2000-05-17 Applied Research Systems ARS Holding N.V. Chemokine receptor antagonist and cyclosporin in combined therapy
AU766595B2 (en) * 1998-09-18 2003-10-16 Laboratoires Serono Sa Chemokine receptor antagonist and cyclosporin in combined therapy
US6902728B1 (en) 1998-09-18 2005-06-07 Applied Research Systems Ars Holding N.V. Chemokine receptor antagonist and cyclosporin in combined therapy
WO2015063613A2 (en) 2013-11-01 2015-05-07 Spherium Biomed S.L. Inclusion bodies for transdermal delivery of therapeutic and cosmetic agents

Also Published As

Publication number Publication date
CA2228379A1 (en) 1997-02-20
JPH11510806A (ja) 1999-09-21
EP0845994A1 (en) 1998-06-10
AU6842296A (en) 1997-03-05
AU712606B2 (en) 1999-11-11

Similar Documents

Publication Publication Date Title
CA2087525C (en) Adoptive immunotherapy with interleukin-7
US9301979B2 (en) Method for attenuating immune response using mesenchymal stem cells and cytokines
US6165785A (en) Bone marrow cultures for developing suppressor and stimulator cells for research and therapeutic applications
WO2003087320A2 (en) Antagonists of il-21 and modulation of il-21-mediated t cell responses
Mastino et al. Combination therapy with thymosin α1 potentiates the anti‐tumor activity of interleukin‐2 with cyclophosphamide in the treatment of the lewis lung carcinoma in mice
US20090142317A1 (en) Process for reducing effects of graft versus host disease using ex vivo expanded cd4+cd25+ regulatory t cells
US6074635A (en) T cell activation
WO2006081620A1 (en) Cd4+ cd25+ t-cells activated to a specific antigen
Ramírez et al. Induction of resistance to active experimental allergic encephalomyelitis by myelin basic protein‐specific Th2 cell lines generated in the presence of glucocorticoids and IL‐4
WO1993017698A1 (en) Use of interleukin-10 to suppress graft-vs.-host disease
AU712606B2 (en) Combined use of interleukin-10 and cyclosporin for immunosuppression therapy
US6022536A (en) Combined use of interleukin 10 and cyclosporin for immunosuppression therapy
Keever et al. Natural killer and lymphokine-activated killer cell activities from human marrow precursors. II. The effects of IL-3 and IL-4.
US6319493B1 (en) Treatment of neoplastic disease with interleukin-10
MXPA98001091A (en) Combined use of interleukin-10 and cyclosporine for immunosupres therapy
KR101488224B1 (ko) 면역학적 질병의 치료를 위한 티모신 알파 1의 용도
Wei et al. Lymphokine-activated killer cell regulation of T-cell-mediated immunity to Candida albicans
EP0561927B1 (en) Pharmaceutical compositions for the treatment of b-cell malignancies
Golling et al. In vitro cytokine responses in liver transplant recipients treated with cyclosporine A and tacrolimus
Frankel et al. Immunotherapy of acute myeloid leukemia
US20240108655A1 (en) Methods of producing mixed chimerism after a solid organ transplant
Kloosterman Graft-versus-leukemia: applicable in the treatment of leukemia with bone marrow transplantation?: preclinical studies in rat models relevant for human acute leukemia
Mouse Increased Antitumor Effects Using IL-2 with
Degauque et al. TIM-1 PATHWAY CONSTITUTE A NEW “FIRST SIGNAL PATHWAY” FOR T CELL.
Berger et al. 9th Symposium Molecular Biology of Hematopoiesis (Part 1 of 12)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BB BG BR BY CA CN CZ EE GE HU IL IS JP KG KR KZ LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TR TT UA UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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

Ref document number: 2228379

Country of ref document: CA

Ref country code: CA

Ref document number: 2228379

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1997 508515

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: PA/a/1998/001091

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1996928798

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996928798

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1996928798

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1996928798

Country of ref document: EP