AU768782B2 - Treatment of autoimmune conditions with copolymer 1 and related copolymers and peptides - Google Patents

Treatment of autoimmune conditions with copolymer 1 and related copolymers and peptides Download PDF

Info

Publication number
AU768782B2
AU768782B2 AU52262/99A AU5226299A AU768782B2 AU 768782 B2 AU768782 B2 AU 768782B2 AU 52262/99 A AU52262/99 A AU 52262/99A AU 5226299 A AU5226299 A AU 5226299A AU 768782 B2 AU768782 B2 AU 768782B2
Authority
AU
Australia
Prior art keywords
terpolymer
ala ala
mole fraction
copolymer
use according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU52262/99A
Other versions
AU5226299A (en
Inventor
Rina Aharoni
Ruth Arnon
Masha Fridkis-Hareli
Michael Sela
Jack L. Strominger
Dvora Teitelbaum
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Yeda Research and Development Co Ltd
Harvard College
Original Assignee
Yeda Research and Development Co Ltd
Harvard College
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 Yeda Research and Development Co Ltd, Harvard College filed Critical Yeda Research and Development Co Ltd
Priority claimed from PCT/US1999/016747 external-priority patent/WO2000005250A1/en
Publication of AU5226299A publication Critical patent/AU5226299A/en
Application granted granted Critical
Publication of AU768782B2 publication Critical patent/AU768782B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

Treatment of Autoimmune Conditions with Copolymer 1 and Related Copolymers RELATED
APPLICATIONS
The present application claims the benefit of provisional applications 60/093,859 filed July 23, 1998, 60/101,825, filed September 25, 1998, 60/102,960, filed October 2, 1998, 60/106,350, filed October 30, 1998, and 601108,184, filed 10 November 12, 1998, all of which are incorporated by reference herein.
:GOVERNMENT
FUNDING
This invention was made in part with government support under grant CA47554 awarded by the National Institutes of Health. The government has certain 15 rights in the invention.
INTRODUCTION
The present invention provides compositions and methods for treating autoimmune diseases using therapeutically effective amounts of a polypeptide 20 related to Copolymer 1. Copolymer 1 is a heterogeneous mixture of synthetic random linear copolymers of tyrosine, alanine, glutamic acid and lysine and, in appropriate therapeutic amounts and average molecular sizes, is used to treat multiple sclerosis. When such mixtures of synthetic random linear copolymers consist essentially of the three of the four amino acids found in Copolymer 1, they are referred to as Terpolymers. The present invention relates in part to Terpolymers.
Preferably, the Terpolymers are composed of tyrosine, alanine and lysine, or of glutamic acid, tyrosine and lysine, or of glutamic acid, alanine and lysine.
Surprisingly, the Terpolymers have efficacy for treating a variety of auto-immune diseases and bind to Class II major histocompatibility complex (MHC) molecules as well as to antigen presenting cells.
P4/ BACKGROUND OF THE INVENTION SS Autoimmune diseases occur when an organism's immune system fails to recognize some of the organism's own tissues as "self" and attacks them as "foreign." Normally, self-tolerance is developed early by developmental events within the immune system that prevent the organism's own T cells and B cells from reacting with the organism's own tissues. MHC cell surface proteins help regulate these early immune responses by binding to and presenting processed peptides to T cells.
"'This self-tolerance process breaks down when autoimmune diseases develop.
Now the organism's own tissues and proteins are recognized as "autoantigens" and are attacked by the organism's immune system. For example, multiple sclerosis is believed to be an autoimmune disease occurring when the immune system attacks the myelin sheath, whose function is to insulate and protect nerves. It is a progressive disease characterized by demyelination, followed by neuronal and motor function loss. Rheumatoid arthritis is also believed to 1• be an autoimmune disease which involves chronic inflammation of the synovial joints and infiltration by activated T cells, macrophages and plasma cells, leading to a progressive destruction of the articular cartilage. It is the most severe form of joint disease. The nature of the autoantigen(s) attacked in rheumatoid arthritis is poorly understood, although collagen type II is a candidate.
A tendency to develop multiple sclerosis and rheumatoid arthritis is inherited these diseases occur more frequently in individuals carrying one or more characteristic MHC class II alleles. For example, inherited susceptibility for rheumatoid arthritis is strongly associated with the MHC class II DRB1 *0401, DRB 1 *0404, or DRB 1*0405 or the DRB1*0101 alleles. The histocompatibility locus antigens (HLA) are found on the surface of cells and help determine the individuality of tissues from different persons. Genes for histocompatibility locus antigens are located in the same region of chromosome 6 as the major histocompatibility complex (MHC). The MHC region expresses a number of distinctive classes of molecules in various cells of the body, the genes being, in order of sequence along the chromosome, the Class I, II and III MHC genes. The Class I genes consist of HLA genes, which are further subdivided into A, B and C subregions. The Class II genes are subdivided into the DR, DQ and DP subregions. The MHC-DR molecules are the best known; these occur on the surfaces of antigen presenting cells such as macrophages, dendritic cells of lymphoid tissue and epidermal cells. The Class III MHC products are expressed in various components of the complement system, as well as in some nonimmune related cells.
A number of therapeutic agents have been developed to treat autoimmune diseases, including steroidal and non-steroidal anti-inflammatory drugs, for example, methotrexate; various interferons; and certain inhibitors of prostaglandin synthesis. However, these agents can be toxic when used for more than short periods of time or cause undesirable side effects. Other therapeutic agents bind to and/or inhibit the inflammatory activity of tumor necrosis factor (TNF), for example, anti-TNF specific antibodies or antibody fragments, or a soluble form of the TNF receptor. These agents target a protein on the surface of a T cell and generally prevent interaction with an antigen presenting cell (APC). However, therapeutic compositions containing natural folded proteins are often difficult to produce, formulate, store, and deliver. Moreover, the innate heterogeneity of the immune system can limit the effectiveness of drugs and complicate long-term S: treatment of autoimmune diseases.
Thus in order to effectively treat autoimmune diseases and other immune conditions, new drugs are needed that do not have the side effects of the present therapeutic agents and which adequately address to the innate heterogeneity of ;the immune system:- I RAZ
SEC
S104 3 oo•
REFERENCES
Aharoni, et al., 58 Immunology Letters 79 (1997).
Allison, in IMMUNOSUPPRESSION AND ANTI-INFLAMMATORY DRUGS, ANNALS OF THE NEW YORK ACADEMY OF SCIENCE 696:xi (1993).
Ben-Nun A et al., 243 J NEUROL (Suppl 1) S14-S22 (1996).
Dorling et al., 6 CUR. OPINIONS IMMUNOL. 765 (1994).
Ferrara et al., 324 NEW ENGLAND J. OF MEDICINE 667 (1991).
Fridkis-Hareli, et al., 63 J. NEUROCHEM. 63 (Suppl. 1) S61 (1994).
Fridkis-Hareli, et al., 163 CELL. IMMUNOL. 229. (1995).
Fridkis-Hareli, et al., 160 J. IMMUNOL. 4386 (1998).
Johnson, 1 NEUROLOGY 65-70 (1995).
Kay et al., 22 TRANSPLANTATION PROCEEDINGS 96 (1990).
Kelemen, et al., 102 INT ARCH ALLERGY IMMUNOL. 309 (1993).
15 Mengle-Gaw, The Major Histocompatibility Complex (MHC), in the ENCYCLOPEDIA OF MOLECULAR BIOLOGY 602-06 (Oxford: Blackwell Science Ltd., 1994).
Rothbard, J. et al,. 9 ANNU. REV. IMMUNOL. 527 (1991).
Schlegel, et al., 84 BLOOD 2802 (1994).
Sela M et al., 88 BULL INST PASTEUR 303-14 (1990).
Stazl, 22 TRANSPLANTATION PROCEEDINGS 5 (1990).-- Sykes, 10 THE FASEB JOURNAL 721 (1996).
Teitelbaum et al., 1 EUR. J. IMMUNOL. 242-48 (1971).
Teitelbaum et al., 3 EUR. J. IMMUNOL. 273-79 (1973).
Teitelbaum et al., 64 J. NEUROIMMUNOL. 209-17 (1996).
Thomson, 10 IMMUNOLOGY TODAY 6 (1988).
Van Den Bogaerde, et al., 52 TRANSPLANTATION 15 (1991).
Webb et al. 13 IMMUNOCHEM. 333 (1976).
SEC 4 104 Jt~ SUMMARY OF THE INVENTION Surprisingly, Copolymer 1 and the Terpolymers of the present invention can be used to treat a variety of autoimmune diseases in a heterogeneous patient population. These Terpolymers can inhibit some of the physiological responses of T cells that attack self-antigens as these diseases progress. Moreover, Copolymer 1 and the Terpolymers of the present invention bind with high affinity to antigen presenting cells from different genetic backgrounds and to several class II MHC molecules to block immune cell recognition and attack. In addition, the Terpolymers can stimulate the growth and functioning of Copolymer 1-specific T cells to further treat and prevent various autoimmune diseases.
Accordingly, the present invention provides a composition having a polypeptide comprising three different amino acids selected from the group of amino acids j5 comprising Copolymer 1, that is, glutamic acid, alanine, lysine, and tyrosine, in the approximate relative molar ratios found in Copolymer 1.
The present invention is also directed to pharmaceutical compositions which include a therapeutically effective amount of a Terpolymer consisting essentially 20 of amino acids tyrosine, alanine and lysine, in the molar ratio of from about 0.005 -foabou t-025 tyrosine, from about 0.3 to about 0.6 alanine, and from about 0.1 to about 0.5 lysine, and a pharmaceutically acceptable carrier. This Terpolymer is preferably substantially free of glutamic acid.
The present invention further provides a pharmaceutical composition which includes a therapeutically effective amount of a Terpolymer consisting essentially of glutamic acid, tyrosine and lysine, in the molar ratio of from about 0.005 to about 0.300 glutamic acid, from about 0.005 to about 0.250 tyrosine; and from about 0.3 to about 0.7 lysine, and a pharmaceutically acceptable carrier. The Terpolymer is preferably substantially free of alanine.
The present invention is also directed to pharmaceutical compositions which include a therapeutically effective amount of a Terpolymer consisting essentially of amino acids tyrosine, glutamic acid and alanine in the molar ratio of from about 0.005 to about 0.25 tyrosine, from about 0.005 to about 0.3 glutamic acid, and RA. from about 0.005 to about 0.8 alanine and a pharmaceutically acceptable carrier.
SEC
S104 rr0.
This Terpolymer is preferably substantially free of lysine.
The present invention also provides a pharmaceutical composition which includes a therapeutically effective amount of a Terpolymer consisting essentially of glutamic acid, alanine and lysine, in the molar ratio of from about 0.005 to about 0.3 glutamic acid, from about 0.005 to about 0.6 alanine; and from about 0.2 to ,about 0.7 lysine and a pharmaceutically acceptable carrier. This Terpolymer is 'ipreferably substantially free of tyrosine.
The present invention also provides a pharmaceutical composition to treat autoimmune diseases which includes a therapeutically effective amount of Copolymer 1 or a polypeptide consisting essentially of glutamic acid, alanine, tyrosine and lysine, and a pharmaceutically acceptable carrier.
The present invention further provides methods for treating and preventing autoimmune diseases in a mammal which include administering a therapeutically effective amount of a composition comprising Copolymer 1 or a Terpolymer. In another embodiment, the method for treating autoimmune diseases in a mammal further involves inhibiting proliferation of T cells involved in the immune attack.
In another embodiment, the method for treating autoimmune diseases in a mammalinvolves binding-the Terpolymer to an antigen presenting cell. In yet another embodiment, the method for treating autoimmune disease in a mammal involves binding the Terpolymer to a major histocompatibility complex class II protein which is associated with autoimmune diseases.
Autoimmune diseases contemplated by the present invention include arthritic conditions, demyelinating diseases and inflammatory diseases. For example, autoimmune diseases which can be treated by the present compositions include multiple sclerosis, autoimmune hemolytic anemia, autoimmune oophoritis, autoimmune thyroiditis, autoimmune uveoretinitis, Crone's disease, chronic immune thrombocytopenic purpura, colitis, contact sensitivity disease, diabetes mellitus, Graves disease, Guillain-Barre's syndrome, Hashimoto's disease, idiopathic myxedema, myasthenia gravis, psoriasis, pemphigus vulgaris, rheumatoid arthritis, or systemic lupus erythematosus. The present compositions can be used to treat one or more of these diseases.
_elRA, SEC 6 S104 U; a.
a. a a a *a so BRIEF DESCRIPTION OF THE DRAWINGS As used below "GAL" is a random terpolymer of glutamic acid, alanine and lysine; "TGA" or 'YEA" is a random terpolymer of tyrosine, glutamic acid, and alanine; 'TAL" or "YAK" is a random terpolymer of tyrosine, alanine, and lysine; "GTL" or "YEK" is a random terpolymer of glutamic acid, tyrosine, and lysine; and "YEAK" is Copolymer 1.
Figure 1 illustrates the effect of Copolymer 1 and Terpolymers on proliferation of T cells which are specific for certain myelin basic protein (MBP) peptides. Several of the present Terpolymers inhibit proliferation of T cell lines which are specific for myelin basic protein antigen, MBP 84-102 (SEQ ID NO: The following symbols were used: Copolymer 1 GAL TGA TAL GTL Figure 1A illustrates inhibition of the proliferation of mouse T cell clone MBP-sp-1 specific to MBP 84-102 peptide (0.5 g/well). As a control, proliferation of mouse T cell clone MBP-sp-1 stimulated by MBP 84-102 (SEQ ID NO: 1) without inhibitor was 21,145 cpm. Figure 1B illustrates inhibition of the response of human-Tcell-clone-GP-25-to-MBP 84-102 peptide-(0.-125 gwell).-Proliferation of human T cell clone GP-25 stimulated by MBP 84-102 (SEQ ID NO: 1) peptide without inhibitor was 11,442 cpm.
Figure 2A compares binding of the present polypeptides with-the-binding of Copolymer 1, to Class II major histocompatibility molecules DR1 (top), DR2 (middle) and DR4 (bottom). Binding by Copolymer 1 was compared to binding by the following biotinylated Terpolymers: GAL TGA GTL and TAL The amount of Class II major histocompatibility molecule was held constant at 0.5 Mg/sample and the concentration of polypeptide was varied between 0-8 jM as indicated on the y-axis. Binding was at pH 5.0 for 40 hr at 37 0 C. Binding was detected by capturing the polypeptide-class II complexes with an LB3.1 antibody and detecting the amount of biotinylated polypeptide bound by monitoring the absorbency at 410 nm after reaction with streptavidin-conjugated alkaline phosphatase.
7
SEC
*o 104 A.
Figure 2B provides Lineweaver-Burke plots of the binding data provided in Figure 2A.
Figure 3 illustrates the competitive inhibition of Copolymer 1 binding to Class II major histocompatibility molecules by the present polypeptides. Purified HLA- DR1 (top), HLA-DR2 (middle) and HLA-DR4 (bottom) molecules were incubated Swith a constant amount (1.5 M) of biotinylated Copolymer 1, either alone or in the presence of one of the following unlabeled polypeptides: Copolymer 1 GAL TGA GTL and TAL Inhibition by these polypeptides was compared to inhibition by the myelin basic protein antigen HA 306-318 (SEQ ID NO: Binding was at pH 5.0 for 40 hr at 37"C. The amount of unlabeled polypeptide was varied between 0.1 -1000 m/M, as indicated on the y-axis.
Specific binding is expressed as the percentage of inhibition using equation 1: percent inhibition 100 (signal with competitor background) x (signal without competitor background) i Figure 4A illustrates the competitive inhibition of TAL binding to Class II major histocompatibility molecules by bacterial superantigens SEA, SEB and TSST-1.
urified-HLA-DR1 (top)- HLA-DR2 (middle) and HLA-DR4 (bottom) molecules 0 were incubated with a constant amount of biotinylated TAL, in the presence of increasing amounts of unlabeled bacterial superantigen SEA SEB or TSST-1 Binding was at pH 5.0 for 40 hr at 37 0 C. The amount of superantigen was varied between 0.1 -1000 pM, as indicated on the y-axis.
Specific binding is expressed as the percentage of inhibition using equation 1.
Figure 4B illustrates the competitive inhibition of TGA binding to Class II major histocompatibility molecules by bacterial superantigens SEA, SEB and TSST-1.
Purified HLA-DR1 (top), HLA-DR2 (middle) and HLA-DR4 (bottom) molecules were incubated with a constant amount of biotinylated polypeptide TGA, in the presence of increasing amounts of unlabeled bacterial superantigen SEA SEB or TSST-1 Binding was at pH 5.0 for 40 hr at 37"C. The amount of superantigen was varied between 0.1 -1000 AM, as indicated on the y-axis.
Specific binding is expressed as the percentage of inhibition, calculated according to equation above 1.
SEC 8 1048 Figure 4C illustrates the competitive inhibition of GAL binding to Class II major histocompatibility molecules by bacterial superantigens SEA, SEB and TSST-1.
Purified HLA-DR1 (top), and HLA-DR2 (bottom) molecules were incubated with a constant amount of biotinylated GAL, in the presence of increasing amounts of unlabeled bacterial superantigen SEA SEB or TSST-1 Binding was at pH 5.0 for 40 hr at 37 0 C. The amount of superantigen was varied between 0.1 -1000 as indicated on the y-axis. Specific binding is expressed as the percentage of inhibition using equation 1 above.
Figure 5 shows inhibition of binding of labeled molecules to MHC class II purified proteins by Terpolymers of the present invention. Fig. 5A shows inhibition of binding to the MHC HLA-DR1 protein. Fig. 5B shows inhibition of binding to the MHC HLA-DR4 protein. Unlabeled competitors include Terpolymers of the present invention, influenza virus hemagglutinin (HA) peptide 306-318 (SEQ ID NO:2), and type II collagen .(CII) peptide 261-273 (SEQ ID NO:3). The S: concentration of unlabeled competitor is indicated on the abscissa. In each panel, inhibition by CII 261-273 (SEQ ID NO:3) is shown as open circles inhibition by HA 306-318 (SEQ ID NO: 2) is shown by solid circles inhibition by one of the present terpolymers is indicated as shown by open or solid triangles or :2 squares. The extent of inhibition by GTL is shown using open triangles by o .TAL is -shown-as solid- triangles-(A)- by-TGA as-open-squares-(9) -and by 'Copolymer 1 as solid squares Specific binding observed and shown on the i ordinate was calculated as percentage of inhibition using equation 1 above.
Figure 6 shows-inhibition-of IL-2 production by-DR1-restricted-CII-specific T cell hybridomas in the presence of different polypeptides of the present invention.
Irradiated L57.23 cells (fibroblasts transfected with a gene encoding HLA-DR1) were coincubated in duplicate with collagen peptide CII 261-273 (40 /g/ml) and varying concentrations, shown on the abscissa, of one of the present polypeptides for 2 hr at 37 0 C, then T cells (clone 3.19 or 19.3 as indicated) were added, and the mixtures were further incubated for 24 hr at 37°C. Supematants (30 il) were then removed, and were assayed for activation by IL-2-induced proliferation of IL- 2-dependent cytotoxic T lymphocytes (CTL-L). The extent of inhibition by TAL is shown as solid circles by TGA as solid triangles by GTL as open triangles and by Copolymer 1 as solid squares Percent inhibition of CTL- L proliferation shown on the ordinate was calculated according to equation 1.
*0 Figure 7 shows inhibition of IL-2 production by DR4-restricted CII-specific T cell hybridomas (3838 and D3) in the presence of different polypeptides of the present invention. Fig. 7A shows the effects of coincubating irradiated 3838 or D3 Priess cells with collagen peptide CII 261-273 (SEQ ID NO: 3) at the fixed concentration of 40 /g/ml, and with varying concentrations of each of the present polypeptides, for 2 hr at 37 0 C. Fig. 7B shows the effects of incubating L cells transfected with I a gene encoding HIA-DR4 with collagen peptide CII 261-273 (SEQ ID NO:3) at S'0the fixed concentration of 40 ug/ml, and with varying concentrations of each of the present polypeptides, for 2 hr at 37 0 C. T cells were then added (clones 3838 or D3 as indicated), samples were further incubated for 24 hr at 37°C, and supernatants were assayed as described in Figure 6. Each polypeptide was tested in duplicate. The concentration of the present polypeptides is indicated on the abscissa. The same symbols as used in Figure 6 are used for this figure.
SEC
104 DETAILED DESCRIPTION OF THE INVENTION According to the present invention, polypeptides having at least three different amino acids randomly polymerized in a linear configuration are useful for treating autoimmune diseases. Autoimmune diseases occur when the immune system inappropriately attacks certain tissues or cells. The polypeptides of the present invention can prevent the immune system from attacking, for example, by suppressing the proliferation or function of T or B cells responsible for the attack, or by shielding the tissue from attack by binding to a MHC protein on the surface of the cells that make up the tissue.
Amino acids of the present invention include, but are not limited to the commonly occurring amino acids. Also included are naturally occurring and synthetic derivatives, for example, selenocysteine. Amino acids further include amino acid analogs. An amino, acid "analog" is a chemically related form of the amino acid having a different configuration, for example, an isomer, or a Dconfiguration rather than an L-configuration, or an organic molecule with the approximate size and shape of the amino acid, or an amino acid with modification to the atoms that are involved in the peptide bond, so as to be protease resistant when polymerized in a polypeptide.
The phrases "amino acid" and "amino acid sequence" as defined here and in the claims can include one or more components which are amino acid derivatives and/or amino acid analogs comprising part or the entirety of the residues for any one or more-ef-the-20 naturally-oeurring-amino-acids-indicated by that sequence.
For example, in an amino acid sequence having one or more tyrosine residues, a portion of one or more of those residues can be substituted with homotyrosine.
Further, an amino acid sequence having one or more non-peptide or peptidomimetic bonds between two adjacent residues, is included within this definition.
The one letter and three letter amino acid codes (and the amino acid that each represents) are as follows: A means ala (alanine); C means cys (cysteine);
D
means asp (aspartic acid); E means glu (glutamic acid); F means phe (phenylalanine); G means gly (glycine); H means his (histidine); I means ile RA/ (isoleucine); K means lys (lysine); L means leu (leucine); M means met (methionine); N means asn (asparagine); P means pro (proline); Q means gin (glutamine); R means arg (arginine); S means ser (serine); T means thr (threonine); V means val (valine); W means trp (tryptophan); and Y means tyr (tyrosine).
The term "hydrophobic" amino acid is defined here and in the claims as including S aliphatic amino acids alanine or ala), glycine or gly), isoleucine or ile), leucine or leu), proline or pro), and valine or val), the terms in parentheses being the one letter and three letter standard code abbreviations for each amino acid, and aromatic amino acids tryptophan or trp), phenylalanine (F or phe), and tyrosine or tyr). The amino acids confer hydrophobicity as a function of the length of aliphatic and size of aromatic side chains, when found as residues within a protein.
iq^* The term "charged" amino acid is defined here and in the claims as including amino acids aspartic acid or asp), glutamic acid or glu), histidine or his), arginine or arg) and lysine or lys), which confer a positive (his, lys and arg) or negative (asp and gly) charge at physiological values of pH in aqueous solutions on proteins containing these residues.
Polypeptide Compositions Contemplated by the Invention The polypeptides of the present invention comprise Copolymer 1 and Terpolymers consisting essentially of three of the four amino acids of Copolymer 1, namely tyrosine, glutamic acid, alanine and lysine. However, one of skill in the art can readily substitute structurally-related and/or charge-related amino acids without deviating from the spirit of the invention. Thus, the present invention further contemplates conservative amino acid substitutions for tyrosine, glutamic acid, alanine and lysine in the present polypeptides. Such conservative substitutions are structurally-related amino acid substitutions, including those amino acids which have about the same charge, hydrophobicity and size as tyrosine, glutamic acid, alanine or lysine. For example, lysine is structurally-related to arginine and histidine; glutamic acid is structurally-related to aspartic acid; tyrosine is structurally-related to serine, threonine, phenylalanine and tryptophan; and alanine is structurally-related to valine, leucine and isoleucine. These and other conservative substitutions, such as structurally-related synthetic amino acids, are contemplated by the present invention.
Moreover, the Terpolymers can be composed of or d-amino acids. As is known by one of skill in the art, /-amino acids occur in most natural proteins. However, d-amino acids are commercially available and can be substituted for some or all of the amino acids used to make the Terpolymers. The present invention contemplates Terpolymers formed from mixtures of d- and /-amino acids, as well as Terpolymers consisting essentially of either or d-amino acids.
The average molecular weight and the average molar fraction of the amino acids in the Terpolymers can vary. However, an average molecular weight range of about 2,000 to about 40,000 daltons is contemplated. A preferred average molecular weight range is from about 4,000 to about 12,000 daltons. Preferred average molecular weight ranges and processes of making the Terpolymers are described in U.S. Patent No. 5,800,808, which is hereby incorporated by reference in its entirety.
In one embodiment, the present invention provides Terpolymers containing tyrosine, alanine and lysine. The average molar fraction of the amino acids in these Terpolymers can vary, for example, tyrosine can be present in a mole fraction of about 0.005 to about 0.250; alanine can be present in a mole fraction of about 0.3 to about 0.6; and lysine can be present in a mole fraction of about 0.1 to about 0.5. The average molecular weight is between 2,000 to about 40,000 daltons and preferably between about 3,000 to about 35,000 daltons. In a more preferred embodiment, the -average-molecular weight-is-about 5,000 to about 25,000 daltons.
In another embodiment, the present invention provides Terpolymers containing tyrosine, glutamic acid and lysine. The average molar fraction of the amino acids in these polypeptides can also vary, for example, glutamic acid can be present in a mole fraction of about 0.005 to about 0.300, tyrosine can be present in a mole fraction of about 0.005 to about 0.250; lysine can be present in a mole fraction of about 0.3 to about 0.7. The average molecular weight is between 2,000 to about 40,000 daltons and preferably between about 3,000 to about 35,000 daltons. In a more preferred embodiment, the average molecular weight is about 5,000 to about 25,000 daltons.
In another embodiment, the present invention provides Terpolymers containing glutamic acid, alanine and lysine. The average molar fraction of the amino acids in these polypeptides can also vary, for example, glutamic acid can be present in a mole fraction of about 0.005 to about 0.300, alanine can be present in a mole fraction of about 0.005 to about 0.600; lysine can be present in a mole fraction of about 0.2 to about 0.7. The average molecular weight is between 2,000 to about ,1 40,000 daltons and preferably between about 3,000 to about 35,000 daltons. In more preferred embodiment, the average molecular weight is about 5,000 to about 25,000 daltons.
In another embodiment, the present invention provides Terpolymers containing tyrosine, glutamic acid and alanine. The average molar fraction of the amino acids in these polypeptides can also vary, for example, tyrosine can be present in a mole fraction of about 0.005 to about 0.250; glutamic acid can be present in a mole fraction of about 0.005 to about 0.300; and alanine can be present in a S: mole fraction of about 0.005 to about 0.800. The average molecular weight is between 2,000 to about 40,000 daltons and preferably between about 3,000 to about 35,000 daltons. In a more preferred embodiment, the average molecular weight is about 5,000 to about 25,000 daltons.
"In a more preferred embodiment, the molefraction of amino acids of the Terpolymers is about what is preferred for Copolymer 1. The mole fraction of amino acids in Copolymer 1 is glutamic acid (about 0.14), alanine (about 0.43), i tyrosine (about 0.10) and lysine (about 0.34). The most preferred average molecular weight for Copolymer 1 is between about 5,000 and about 9,000 daltons.
The molar ratios of the monomers of the more preferred terpolymer of glutamic acid, alanine and tyrosine is about 0.21 to about 0.65 to about 0.14.
The molar ratios of the monomers of the more preferred terpolymer of glutamic acid, alanine and lysine is about 0.15 to about 0.48 to about 0.36.
The molar ratios of the monomers of the more preferred terpolymer of glutamic acid, tyrosine, and lysine is about 0.26 to about 0.16 to about 0.58.
The molar ratios of the monomers of the more preferred terpolymer of tyrosine, alanine and lysine is about 0.10 to about 0.54 to about 0.35.
In one embodiment, the Terpolymers of the present invention are capable of binding to an MHC class II protein which, preferably, is associated with an autoimmune disease. Any available method can be used to ascertain whether the Terpolymer binds to one or more MHC class II proteins. For example, the polypeptide can be labeled with a reporter molecule (such as a radionuclide or biotin), mixed with a crude or pure preparation of MHC class II protein and binding is detected if the reporter molecule adheres to the MHC class II protein after removal of the unbound polypeptide.
SIn another embodiment, the Terpolymers of the invention are capable of binding to an MHC class II protein associated with multiple sclerosis. A polypeptide of this embodiment can have similar or greater affinity for the antigen binding groove Iof an MHC class II protein associated with multiple sclerosis than does Copolymer 1. Hence, the contemplated polypeptide can inhibit binding of or displace the binding of myelin autoantigens from the MHC class II protein. One MHC class II protein associated with multiple sclerosis is HLA-DR4 (DRB1*1501).
In another eiboadi-mi-nt,-Copolymer- 1 nd-the-Terpolymers of theivention are capable of binding to an MHC class II protein associated with an arthritic condition, for example, rheumatoid arthritis or osteoarthritis. Copolymer 1 or a Terpolymer of this embodiment can have a greater affinity for the antigen binding groove of an MHC-class II protein associated-with the autoimmune disease than does a type II collagen 261-273 (SEQ ID NO:3) peptide. Hence, the contemplated Copolymer 1 or Terpolymers can inhibit binding of or displace the type II collagen 261-273 peptide from the antigen binding groove of an MHC class II protein. The Class II MHC protein consists of approximately equal-sized a and 1 subunits, both of which are transmembrane proteins. A peptide-binding cleft is formed by parts of the amino termini of both a and 0 subunits. This peptidebinding cleft is the site of presentation of the antigen to T cells. There are at least three types of Class II MHC molecules: HLA-DR, HLA-DQ, and HLA-DP molecules. There are also numerous alleles encoding each type of these HLA molecules. The Class II MHC molecules are expressed predominantly on the -surfaces of B lymphocytes and antigen presenting cells such as macrophages.
The present Terpolymers can be formulated into pharmaceutical compositions containing a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, sweeteners and the like. The pharmaceutically acceptable carriers may be prepared from a wide range of materials including, but not limited 1 to, flavoring agents, sweetening agents and miscellaneous materials such as 'buffers and absorbents that may be needed in order to prepare a particular therapeutic composition. The use of such media and agents with pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. The present compositions may be formulated as an injectable solution or suspension, a spray 1 solution or a suspension.
*9 .o Therapeutic Methods Contemplated by the Invention The present invention further provides methods for treating and preventing autoimmune diseases in a mammal which include administering a therapeutically effective amount of a composition having a polypeptide containing at least three different amino acids selected from the group consisting of the amino acids which comprise Copolymer 1, namely glutamic acid, tyrosine, lysine, and alanine, wherein the selected amino acids are randomly polymerized in a linear configuration. In -one-embodiment-the-polypeptide is Copolymer 1 or a Terpolymer.
Autoimmune diseases contemplated by the present invention include either cellmediated disease T-cell) or antibody-mediated B cell) disorders. Such disorders can be inter alia arthritic conditions, demyelinating diseases and inflammatory diseases. For example, autoimmune diseases which can be treated by the present polypeptides include multiple sclerosis, autoimmune hemolytic anemia, autoimmune oophoritis, autoimmune thyroiditis, autoimmune uveoretinitis, Crohn's disease, chronic immune thrombocytopenic purpura, colitis, contact sensitivity disease, diabetes mellitus, Graves disease, Guillain-Barre's R.A syndrome, Hashimoto's disease, idiopathic myxedema, myasthenia gravis,
SEC
'o 104 uj -16- Y'Arr o 16 psoriasis, pemphigus vulgaris, rheumatoid arthritis, or systemic lupus erythematosus, The present compositions can be used to treat one or more of these diseases.
The term "arthritic condition" as used herein is a condition wherein at least one symptom of rheumatoid arthritis is observed in at least one joint of a mammal, for example in a shoulder, knee, hip, backbone or a digit of the mammal. Examples of arthritic conditions include "Polyarthritis", which is an arthritic condition that affects more than a single joint; "juvenile arthritis", an arthritic condition of humans under the age of 21; and Felty's syndrome, which can include the symptoms of neutropenia, splenomegaly, weight loss, anemia, lymphadenopathy, and pigment spots on the skin.
In one embodiment, any autoimmune disease can be treated by the present polypeptides so long as the contemplated polypeptide binds to an MHC class II protein that has been associated with the autoimmune disease. One aspect of this embodiment provides a method which includes selecting a polypeptide that inhibits binding of an antigenic peptide to an MHC class II protein, for example, **o S° a method wherein step further comprises selecting the heteropolymer that :.o0 inhibits class II-specific T cell responses to an MHC class II protein-peptide compllex, and a method-wherein the aritigenic-peptide is associated with an autoimmune disease; in another embodiment of the invention, a method is provided wherein the MHC class II protein is associated with an autoimmune disease.
In another embodiment, the method for treating an autoimmune disease in a mammal further involves inhibiting the proliferation or function of T cells which are responsive to an autoantigen. RA is a T cell-mediated autoimmune disease which can be treated with the present polypeptides. The pathological process of autoimmune diseases and immune rejection is mediated by T cells. Upon binding to and recognition of an antigen, T cells proliferate, secrete cytokines and recruit additional inflammatory and cytotoxic cells to the site. The present polypeptides prevent T cell proliferation and T cell functions such as cytokine secretion and recruitment of inflammatory and cytotoxic cells to the site. When the autoimmune disease is an arthritic condition the autoantigen can be collagen, and the present AZ, polypeptides can inhibit the proliferation and function of collagen-responsive
T
cells.
In another embodiment, the method for treating an autoimmune disease in a mammal involves binding the polypeptide to an antigen presenting cell such as a macrophage, a dendritic cell of the lymphoid tissue or an epidermal cell. The proliferation and functions of a T cell are activated when an appropriate antigen ,I is presented to it. By binding to antigen presenting cells, the present polypeptides .may block or otherwise interfere with T cell activation.
In yet another embodiment, the method for treating an autoimmune disease in a mammal involves binding the polypeptide to a major histocompatibility complex class II protein which is associated with an autoimmune disease. The Class II MHC proteins are expressed predominantly on the surfaces of B lymphocytes and :o antigen presenting cells such as macrophages. These Class II MHC proteins have a peptide-binding cleft which is the site at which antigenic peptides are presented to T cells. When the present polypeptides bind to a major histocompatibility complex class II protein, those polypeptides can block or otherwise interfere with antigen presentation and/or T cell activation.
In another embodiment, the method for treating an autoimmune disease in a mammal involves biriding the polypeptide to Copolymer 1-reactive B cell antibodies, and/or Copolymer 1-reactive T cells. Copolymer 1-reactive T 2 3
T
cells facilitate the therapeutic effects of Copolymer 1. When binding to Copolymer 1-reactive T cells, the present polypeptides stimulate those T cells to proliferate, secrete antiinflammatory- cytokines and enhance the therapeutic benefits of treatment by the present methods. According to the present invention, the present polypeptides also bind to autoantigen-reactive antibodies which may block the antibody from attacking the target tissue, thereby helping to prevent the autoimmune disease from progressing. For example, when the present polypeptides are bound to MBP-specific antibodies, those antibodies may not bind to MBP and thereby lead to the destruction of MBP in the myelin sheath.
The present polypeptides may be administered by any convenient route. In one embodiment the present polypeptides can be administered by injection to facilitate delivery to the tissues affected by the autoimmune disease. Thus, the present polypeptides may, for example, be injected, ingested, inhaled, or topically applied.
The subject polypeptides may be incorporated into a cream, solution or SEC 18 104 1104 1 8
'CCJ~
suspension for topical administration. The present polypeptides are preferably administered orally, topically or by injection without addition of an adjuvant.
Useful Kits of the Invention Another embodiment of the invention, provides a kit for assaying the binding of an analyte to an MHC protein, which includes a water-soluble MHC protein which has been recombinantly produced in a non-mammalian cell, and a means for detection of the bound analyte on the MHC protein, and instructions for use. The MHC protein used in the kit is an MHC class II protein selected from the group consisting of an MHC class II HLA-DR1 protein, an MHC class II HLA-DR2 protein and an MHC class II HLA-DR4 protein. The kit can further comprise an autoantigenic peptide.
1 In a preferred embodiment, the MHC class II protein is produced in an invertebrate or a microbial cell, such as an insect cell or a yeast cell and is therefore devoid of bound peptide in the antigen cleft. The means for detecting binding of the analyte to the MHC protein can be any radioactive, fluorimetric, chemiluminescent, enzymatic or colorimetric means known to one of ordinary skill in the art. In a preferred embodiment, the MHC protein is a class II HLA-DR1 or HLA-DR4 protein-Furrtherthe-lit-ca-n in-cde-also an autoantigenftpeptide, such as a collagen II peptide, or a peptide derived from myelin basic protein, myelin oligodendrite protein, or a peptide from some other protein implicated in an autoimmune disease.
*29"! Synthesis of the Terpolymers of the Invention The Terpolymers can be made by any procedure available to one of skill in the art.
For example, the Terpolymers can be made under condensation conditions using the desired molar fraction of amino acids in solution or by solid phase synthetic procedures. Condensation conditions include the proper temperature, pH and solvent conditions for condensing the carboxyl group of one amino acid with the amino group of another amino acid to form a peptide bond. Condensing agents, for example, dicyclohexyl-carbodiimide, can be used to facilitate the formation of the peptide bond. Blocking groups can be used to protect functional groups, such as the side chain moieties and some of the amino or carboxyl groups, against undesired side reactions.
For example, the process disclosed in U.S. Patent 3,849,550 can be used where the N-carboxyanhydrides of tyrosine, alanine, y-benzyl glutamate and N-trifluoroacetyl-lysine are polymerized at ambient temperatures in anhydrous dioxane with diethylamine as an initiator. The y-carboxyl group of the glutamic 'Iacid can be deblocked by hydrogen bromide in glacial acetic acid. The trifluoroacetyl groups are removed from lysine by 1 molar piperidine. One of skill in the art readily understands that the process can be adjusted to make polypeptides containing the desired amino acids, for example, only three of the four amino acids in Copolymer 1 by selectively eliminating the reactions that relate to any one of glutamic acid, alanine, tyrosine or lysine. For purposes of the application, the terms "ambient temperature" and "room temperature" mean a temperature ranging from about 20 to about 26 degrees Centigrade The average molecular weight of the Terpolymers can be adjusted during polypeptide synthesis or after the Terpolymers have been made. To adjust the average molecular weight during polypeptide synthesis, the synthetic conditions or the amounts of amino acids are adjusted so that synthesis stops when the polypeptide reaches the approximate length which is desired. After synthesis, polypeptides with the desired average molecular weight can be obtained by any available size selection procedure, for example, chromatography of the polypeptides on a molecular weight sizing column or gel, and collection of the average molecular weight ranges desired. The present polypeptides can also be partially hydrolyzed to remove high molecular weight species, for example, by acid or enzymatic hydrolysis, and then purified to remove the acid or enzymes.
In one embodiment, the present invention provides a process for preparing Terpolymers with a desired average molecular weight which includes reacting a protected polypeptide with hydrobromic acid to form a polypeptide having the desired average molecular weight profile. The reaction is performed for a time and at a temperature which is predetermined by one or more test reactions.
During the test reactions, the time and temperature are varied and the average molecular weight range of a given batch of test polypeptides is determined. The test conditions which provide the optimal average molecular weight range for that batch of polypeptides are used for the batch. Thus, polypeptides having the desired average molecular weight profile, can be produced by a process which includes reacting the protected polypeptide with hydrobromic acid for a time and at a temperature predetermined by test reaction.
In a preferred embodiment, a test sample of protected polypeptide from a given batch is reacted with hydrobromic acid for about 10-50 hours at a temperature of about 20-28*C. The best conditions for that batch are determined by running several test reactions. For example, in one embodiment, the protected polypeptide is reacted with hydrobromic acid for about 17 hours at a temperature of about 26*C.
The examples which follow describe the invention in detail with respect to showing how certain specific representative embodiments thereof can be made, the materials, apparatus and process steps being understood as examples that are intended to be illustrative only. In particular, the invention is not intended to be limited to the methods, materials, conditions, process parameters, apparatus and the like specifically recited herein.
Throughout this application, various publications, patents, and patent applications 2: have been erefi-to. he teachings and -diclosures fo-hese piublications, patents, and patent applications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which the present invention pertains.
It is to be understood and expected that variations in the principles of invention herein disclosed may be made by one skilled in the art and it is intended that such modifications are to be included within the scope of the present invention.
EXAMPLE I PREPARATION OF POLYPEPTIDES Preparation of Protected Polypeptides ,i Protected polypeptides are prepared as described by Teitelbaum et al., using the N-carboxyanhydride (NCA) blocked amino acids tyrosine, alanine, glutamic acid and trifluoroacetyllysine dissolved in dioxane. 1 EUR. J. IMMUN. 242 (1971). The carboxylate group on glutamic acid is blocked with a benzyl (BZ) group.
S. The polymerization process is initiated by the addition of 0.01 0.02% diethylamine. The reaction mixture is stirred at room temperature for 20 hours and then poured into water. The protected polypeptide product is filtered, washed S"with water and dried. The removal of the y-benzyl blocking groups from the glutamate residue is carried out by treating the protected polypeptide with 33% .I hydrobromic acid in glacial acetic acid at room temperature for 6-12 hours with stirring. The product is poured into excess water, filtered, washed and dried, yielding the protected polypeptide.
Preparing Polypeptides with Molecular Weight 7,000 2,000 Da Treatment of y-benzyl protected polypeptides 33% HBr in acetic acid not only insures removal of the benzyl protecting group from the y-carboxylate of the glutamate residue but also cleaves the polypeptides into smaller polypeptides.
The time needed for obtaining a polypeptide of molecular weight 7,000 2,000 daltons depends on the reaction temperature and the size of protected polypeptide. At temperatures of between 20 0 C to 28°C a test reaction is performed on every batch for different time periods, for example, from 10-50 hours. A curve of average molecular weight obtained over time is drawn. The time needed for obtaining molecular weight 7,000 2,000 Da is calculated from the curve and the reaction is performed on a larger scale. On average, at 26*C, the time period for obtaining a mixture of polypeptides with a molecular weight of 7,000 2,000 Da is 17 hours. The product is poured into excess water, filtered, washed and dried, yielding a polypeptide with a desired range of average AWR' molecular weights.
Preparation of low-toxicity lysine-containing polypeptides Protected trifluoroacetyl-polypeptide (20 g) is dispersed in 1 liter of water and 100 g piperidine is added. The mixture is stirred for 24 hours at room temperature and filtered. The solution of crude polypeptide is distributed into dialysis bags and dialyzed at 10-20°C in water until pH 8 is attained. The polypeptide solution is dialyzed in 0.3% acetic acid and then again in water until pH 5.5 to 6.0 is obtained. This solution is then concentrated and lyophilized to dryness.
Synthesis of: Molecular weight 7,600 TGA, poly[L-Tyr°o 6 L-Glu 0 2
L-
Ala 4 *o 1) Raw Materials: L-Ala-NCA 18.74 g L-Tyr-NCA 6.5 g 5-BZ-L-Glu-NCA 13g Diethylamine 0.165 g Dioxane 790 ml HBr/AcOH(33%) 480 ml Pheihnol 4.8 g Piperidine 13.2 ml Deionized water SAcetic acid 2) Procedure: L-Tyr-NCA (6.5 g) is placed in dioxane (211 ml) and heated to 60°C for 10 min, then filtered. 5-BZ-L-Glu-NCA (13 g) is placed in dioxane (226 ml) and stirred at 20-25°C for 10 min, then filtered. L-Ala-NCA (18.74 g) is placed in dioxane (350 ml) and stirred at 20-25°C for 10 min, then filtered.
A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with the solution of L-Tyr-NCA, the solution of 5-BZ-L-Glu-NCA and the solution of L-Ala-NCA.
Diethylamine (0.165 g) in dioxane (2.5 ml) is introduced to the reaction mixture and stirred at 20-25 0 C for 20 hours. The reaction mixture is added to deionized /water filtered and dried at 60 0 C under vacuum. Yield 25.8g.
A solution of phenol (4.8 g) in HBr/AcOH (33%;480 ml) is stirred for 20 hours. A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with a solution of HBr/AcOH, poly[5-BZ-L-Glu,L-Ala,L-LTyr] and stirred at 26±1 "C for 16 hours. The reaction mixture is added to deionized water (2L) and stirred for 1 hour. The precipitate is filtered and washed with deionized water until the pH is 6. The product is dried in a circulating air oven at 40 0 C, for about 40 hours. Yield 24 g.
A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with a piperidine (13.2 ml), deionized water (1200 ml), and poly[L-Glu,L-Ala,L-LTyr], then stirred at 20-25 0 C for 24 hours. The resulting solution is ultrafiltered through polyethersulphone membranes with a cutoff of 5000 daltons until two thirds of the original volume is removed. The original volume is restored by addition of fresh deionized water. The process is repeated 5 times, until the impurity content is less than 1% by HPLC The resulting solution (at full volume) is acidified to pH 4.4 with acetic acid. The solution is ultrafiltered to pH 5.5-6 and the volume is reduced to one third. The resulting solution is lyophilized to dryness.
Synthesis of Molecular Weight 8850 GAL, poly[L-Glu'1 5 sL-Ala479,L-Lys 365 Q 1) Raw Materials L-Ala-NCA 14 g :.*N6-TFA-L-Lys-NCA 22.9 g 5-BZ-L-Glu-NCA 9.8 g Diethylamine 0.12 g Dioxane 850 ml HBr/AcOH 480 ml Phenol 4.8 g Piperidine 13.2 ml Deionized water Acetic acid 2) Procedure N6-TFA-L-Lys-NCA (22.9 g) in dioxane (420 ml) is stirred at 20-25°C for 10 min.
and filtered. 5-BZ-L-Glu-NCA (9.8 g) in dioxane (170 ml) is stirred at 20-25 0 C for min. and filtered. L-Ala-NCA (14 g) in dioxane (260 ml) is stirred at 20-25°C Sfor 10 min. and filtered. A 2L Erlenmeyer equipped with a magnetic stirrer, is
SEC
_V 104 A 4 N24 0-" charged with the solution of N6-TFA-L-Lys-NCA, the solution of and the solution of L-Ala-NCA. Diethylamine (0.12 g) in dioxane (2.5 ml) is added, and the reaction mixture is stirred at 20-25 0 C for 20 hours. The reaction mixture is added to deionized water filtered and dried at 60°C under vacuum.
A solution of phenol (4.8 g) in HBr/AcOH (33%;480 ml) is stirred for 20 hours. A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with a solution of HBr/AcOH, poly[5-BZ-L-Glu,L-Ala,N6-TFA-L-Lys] (24 g) and is stirred at 26±1 °C for 16 hours. The reaction mixture is added to deionized water (2L) and stirred for 1 hour. The precipitate is filtered and washed with deionized water until pH=6.
The product is dried in a circulating air oven at 40°C, about 40 hours.
A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with piperdine (13.2 ml), deionized water (1200 ml) and poly[L-Glu,L-Ala,N6-TFA-L-Lys] (24 g) is stirred at 20-25"C for 24 hours. The solution is ultrafiltered through polyethersulphone membranes with a cut off of 5000 daltons until two thirds of the original volume are removed. The original volume is restored by addition of fresh deionized water. The process is repeated 5 times, until the impurity content is less than 1% (by HPLC). The-resulting solution (at full volume) is acidified to pH z 4.4 with acetic acid. The solution is ultrafiltered until the pH is 5.5-6 and the volume is reduced to one third. The resulting solution is lyophilized to dryness.
Synthesis of Molecular Weight 11,050 TGL, poly[L-TyrPla, L-Glu'e.L- Lys 0 .s 79 1) Raw Materials 10.34 g N6-TFA-L-Lys-NCA 24.16 g L-Tyr-NCA 5.2 g Diethylamine 0.095 g Dioxane 810 ml HBr/AcOH 460 ml Phenol 4.6 g Piperidine 150 ml Deionized water Acetic acid 2) Procedure L-Tyr-NCA (5.2 g) in dioxane (180 ml) is heated to 60 0 C for 10 min, and filtered.
N6-TFA-L-Lys-NCA (24.16 g) in dioxane (450 ml) is stirred at 20-25 0 C for 10 min.
and filtered. 5-BZ-L-Glu-NCA (10.35 g) in dioxane (180 ml) is stirred at 20-25°C 'for 10 min. and filtered.
A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with the solution of N6-TFA-L-Lys-NCA, the solution of L-Tyr-NCA and the solution of NCA. Diethylamine (0.095 g) in dioxane (2.5 ml) is introduced, and the reaction mixture is stirred at 20-25°C for 20 hours. The reaction mixture is added to deionized water filtered and dried at 60°C under vacuum.
A solution of phenol (4.6 g) in HBr/AcOH (33%;460 ml) is stirred for 20 hours. A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with the solution of HBr/AcOH and the poly[5-BZ-L-Glu,L-Tyr,N6-TFA-L-Lys]. This mixture is stirred at 26*1 °C for 16 hours and then the mixture is added to deionized water (1.5 L) aned-stirred-foT-Yrour-The-precipitate is filtered and washed with deionized water until the pH is :2t SA 2L Erlenmeyer equipped with a magnetic stirrer, is charged with a piperidine (150 ml), deionized water (1400 ml), and poly[L-Glu,L-Tyr,N6-TFA-L-Lys] (50 g) and is stirred at 20-25°C for 24 hours. The resulting solution of poly[L-Glu, L-Tyr, L-Lys] is ultrafiltered through polyethersulphone membranes with a cut off of 5000 daltons until two thirds of the original volume were removed. The original volume is restored by addition of fresh deionized water. The process is repeated 5 times, until the impurity content is less than 1% (by HPLC). The resulting solution (at full volume) is acidified to pH 4.2 with acetic acid. The solution is ultrafiltered to a pH of 5.5-6 and the volume is reduced to one third. The resulting solution is lyophilized to dryness.
Synthesis of Molecular Weight 20,000 TAL, poly[L-TyrO 1 02 L-Ala 054 2
L-
Lys 0 5 3 1) Raw Materials L-Ala-NCA 14 g N6-TFA-L-Lys-NCA 22.9 g L-Tyr-NCA 4.9 g Diethylamine 0.1 g Dioxane 850 ml HBr/AcOH 500 ml Phenol 5 g Piperidine 162 ml Deionized water Acetic acid 2) Procedure L-Tyr-NCA (4.9 g) in dioxane (170 ml) is heated to 60°C for 10 min, and filtered.
N6-TFA-L-Lys-NCA (22.9 g) in dioxane (417 ml) is stirred at 20-25"C for 10 min.
and filtered. L-Ala-NCA (14 g) in-dioxane (260 ml) is stirred at 20-25°C for min. and filtered.
A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with the solution of N6-TFA-L-Lys-NCA, the solution of Tyr-NCA in dioxane and the solution of L-Ala- NCA. Diethylamine (0.1 g) in dioxane (2 ml) is introduced and the mixture is stirred at 20-25"C for 20 hours. The reaction mixture is then added to deionized water filtered and dried at 60*C under vacuum.
A solution of phenol (5 g) in HBr/AcOH (33%;500 ml) is stirred for 20 hours. A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with the solution of HBr/AcOH and the poly[L-Ala,L-Tyr,N6-TFA-L-Lys] and is stirred at 26*1 °C for 16 hours. The reaction mixture is then added to deionized water (2L) and stirred for hour. The precipitate is filtered and washed with deionized water until the pH is A 2L Erlenmeyer equipped with a magnetic stirrer, is charged with a piperidine (162 ml), water (1500 ml) and the poly[L-Ala,L-Tyr,N6-TFA-L-Lys] (30 and is stirred at 20-25°C for 24 hours. The resulting solution of poly[L-Ala,L-Tyr,L-Lys] is ultrafiltered through polyethersulphone membranes with a cut off of 5000 daltons until two thirds of the original volume were removed. The original volume is restored by addition of fresh deionized water. The process is repeated 5 times, until the impurity content is less than 1% (by HPLC). The resulting solution (at full volume) is acidified to pH 4.4 with acetic acid. The solution is ultrafiltered to pH 5.5-6 and the volume is reduced to one third. The resulting solution is lyophilized to dryness.
EXAMPLE 2 LOW MOLECULAR WEIGHT POLYPEPTIDES IN VIVO TESTS .o i. Three batches of copolymer-1 having an average molecular weight of 7.3 and 8.4 SKDa (less than 2.5% copolymer-1 species over 40KDa) and 22KDa (more than copolymer-1 species over 40KDa) were subjected to the toxicity test described below. Five mice were used-in-eacthexperimental group.
Method Copolymer-1 is dissolved in distilled water to yield a solution of 2 mg/ml of the active ingredient. Each mouse is injected with 0.5 ml of the test solution into the lateral tail vein. Mice were observed for mortality and relevant clinical signs over a 48 hour period. Observations were recorded 10 minutes, 24 hours and 48 hours post-injection. If, at the end of 48 hours, all the animals were alive and no adverse signs had been observed, then the batch is designated "non-toxic". If, however, one or more of the mice had died or had shown adverse signs, then the batch is designated "toxic".
Results Three of five mice died after 48 hours when treated with the 22 kDa average 3C molecular weight Copolymer 1 polypeptides. Accordingly, this high average molecular weight batch is designated "toxic". The Copolymer 1 batches having average molecular weights of 7.3 and 8.4 KDa were both designated "non-toxic." IN VITRO RAT BASOPHILIC LEUKEMIA CELL DEGRANULATION TESTS Histamine (or serotonin) release from basophile is an in vitro model for immediate hypersensitivity. The Rat Basophilic Leukemia cell line, RBL-2H, is uniform and easy to maintain in culture but is a highly sensitive and reproducible system for testing for degranulation. E.L. Basumian, et al., 11 EUR. J. IMMUNOL. 317 (1981).
The physiological stimulus for histamine release involves binding of the antigen to membrane-bound IgE molecules, which triggers an intricate biochemical cascade. Degranulation is induced by non-lgE-mediated stimuli, including various peptides and synthetic polymers, e.g. polylysine. R.P. Siraganian, TRENDS IN PHARMACOLOGICAL SCIENCES 432 (Oct. 1983). The RBL degranulation test is, 15 therefore, used to screen out those batches of COP-1 which evoke substantial degranulation and thus might elicit undesirable local and/or systemic side effects.
Method :*2P Rat Basophilic Leukemia cells (RBL-2Ho), are loaded with [H]-serotonin, followed by incubation with 100 Ag of the COP-1 to be tested. Batches of COP-1 which induce non-specific degranulation, release [H3]-serotonin into the medium. The radioactivity in the medium is counted by a scintillation counter and the total radiolabeled serotonini incorporated irto-th-ecells is determined- in the pelleted cells. Percent degranulation is calculated as the percentage of serotonin released out of the total incorporated.
Results Four batches of Copolymer 1, with average molecular weight between 8,250- 14,500 were analyzed for both percentage of the species with molecular weight over 40 kDa and for degranulation of RBL's. Results are summarized in Table 1.
Table 1 Average M.W. of species with M.W. Serotonin Release over 40 KDa 6,250 2.5 12.4 7,300 2.5 21.0 13,000 5 66.9 14,500 5 67.8 I,1 9 9 *9 9 a 9* 15 9 9* 9 9 a 25 As can be seen, when the percentage of high average molecular weight species is low (less than the percent release of serotonin is also low, and vice versa. These data indicate that lower average molecular weight Copolymer 1 polypeptides are preferable to higher average molecular weight Copolymer 1 polypeptides.
EXAMPLE 3 SUPPRESSION OF EAE BY THE POLYPEPTIDES Injection of Copolymer 1 in incomplete Freund's adjuvant before disease induction can suppress experimental allergic encephalomyelitis (EAE). This suppression appears to be mediated by Copolymer 1-specific suppressor T cells of the TN 2 type which cross react with myelin basic protein. Lando et al., 123 J. IMMUNOL.
2156 (1979); Aharoni et al., 17 EUR. J. IMMUNOL. 23 (1993); Aharoni et al., 94 PROC. NATL. ACAD. SCI. USA 10821 (1997). Other researchers have observed that the therapeutic effect of Copolymer 1 in multiple sclerosis patients is also associated with the induction of TH2 cells. Lahat et al., 244 J. Neurol. 129 (1997).
In this example, EAE is suppressed to by different polypeptides of the present invention.
0 0: 3t5
SEC
104 r
SEC
-v io0d
METHODS
Copolymer 1 Copolymer 1 batches 02095 and 55495, with average molecular weights of 6000 Da and 5800 Da, respectively, were obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel).
Terpolymers Four terpolymers were obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel). The properties of these terpolymers are provided below: 1) The GAL terpolymer (SD-1689) is a mixture of polypeptides containing the following mole fraction of amino acids glutamic acid (0.153), alanine (0.479) and lysine (0.365). The range of GAL average molecular weights is about 4650 daltons to about 20,050 daltons. The average molecular weight of this GAL preparation is 8850 daltons.
2) The TGA terpolymer (SD-1690) is a mixture of polypeptides containing the following mole fraction of amino acids tyrosine (0.136), glutamic acid (0.210) and alanine (0.648). The range of TGA average molecular weights is about 1000 daltons to about 40,000 daltons. The average molecular weight of this TGA preparation is 7600 daltons.
3) The TAL terpolymer (SD-1691) is a mixture of polypeptides containing the following mole fraction of amin-db-acids tyrosih-i(0.10 2 ala-inre (0.542) and lysine (0.353). The range of TAL average molecular weights is about 5700 daltons to about 34,400 daltons. The average molecular weight of this TAL preparation is about 20,000 daltons.
4) The GTL terpolymer (SD-1697) is a mixture of polypeptides containing the following mole fraction of amino acids glutamic acid (0.259), tyrosine (0.162) and lysine (0.579). The range of GTL average molecular weights is about 4,000 daltons to about 23,500 daltons. The average molecular weight of this GTL preparation is about 11050 daltons.
A control polypeptide is used, consisting of a mixture of polypeptides containing a. 1:1:1 mixture of amino acids alanine, glutamic acid and tyrosine, with an average molecular weight 26,700 Da. This polypeptide is obtained from Sigma 31 Chemical Company (St. Louis, MO).
Induction of EAE Two to three month old female (SJL/JxBALB/c)FI mice are injected in all four ;.footpads with mouse spinal cord homogenate (3.5mg/mouse) emulsified in a 1:1 ratio in complete Freund's adjuvant (CFA) supplemented with 4 mg/ml H37Ra.
Pertussis toxin (0.25ml, 250ng, Sigma) is injected intravenously, immediately after and 48 hr later. Mice are examined daily from day 10 post induction for clinical signs of EAE which were scored on a 0-5 scale as described in Lando et al., 123 J. IMMUNOL. 2156 (1979).
S EAE suppression by injection with incomplete adjuvant 9.
.15 The tested polypeptides (10mg/mouse) are injected in incomplete Freund's adjuvant (ICFA) subcutaneously in one nuchal area in 2-3 spots. EAE is induced 3 weeks later as described above.
EAE suppression by oral administration o In a second test, female Lewis rats are fed 5 mg/kg guinea pig BP or Copolymer i' 1 dissolved in phosphate buffered saline (PBS) at 2-3 day intervals before EAE induction. EAE is induced two days after the last feeding by injection of 25 pg guinea pig MBP emulsified in 1:1 CFA containing 4 mg/ml mycobacterium tuberculosis (H37Ra) (Difco Lab, Detroit, Mich.). A total volume of 0.1 ml is injected into each of two hindfoot pads. Control rats are mock fed with phosphate buffered saline.
The efficacy of orally administered Copolymer 1 for preventing EAE in rats is compared to that of rats fed guinea pig MBP by the method of Higgins et al., 140 J. IMMUNOL. 440 (1988).
Animals were examined daily from day 10 post induction for signs of disease.
EAE is scored as follows: 0 no disease; 1 limp tail; 2 hind limb paralysis; 3 S= paralysis of all four limbs; 4 moribund condition; and 5 death.
RESULTS
EAE suppression by injection with incomplete adjuvant Table 2 illustrates the effects of administering the present polypeptides by injection in ICFA prior to induction of EAE. Of the four polypeptides tested, Copolymer 1 and TAL caused the greatest EAFE suppression. GTL also exhibited good suppressive activity. GAL and TGA, were somewhat less effective.
10 Table 2 Suppression of EAE in mice by Injection of the Present Polypeptides 1 0*e g* 0. t a 06 Polyeptde Icidnce MS*Suppression Polpepid J Iniec MM~ jIncidence[ MMS* None11/11 (Negative Control) IOFA 11/11 3.4 0.0 15.0 (Negative Control) Copolymer 1 (Positive ~04 208.
Control) SD-1689 GAL 7/8 3.5 12.5 12.5 SD)-1690 TGA 6/8 2.5 25.0 37.5 SDA1691 -TAL 3/8 1.1 62.5 72.5 SD-1697 GTL 5/8 1.5 37.5 62.5 D-Copolymer 1 11/11 4.1 0..0.0 MMS =an Maximal Score D-CopalYmfer I is a copolymer of d-lysifle, d-tyrosifle, d-glutamic acid and d-alanine in the molar ratios of Copolymer 1.
T able 3 illustrates the efficacy of orally administered Copolymer I in preventing the clinical manifestations of EAE in Lewis rats compared to rats receiving only phosphate buffered saline (PBS) or guinea pig basic protein (GPBP).
Table 3 Suppression of EAE in Rats by Oral Administration of the Present Polypeptides ,d Antiaen Incidence MMS SD Mean Onset (days) Ir Control) 27/28 1.8 0.5 11.9 lk 4
C
.:o ms .e 0 s se em em S *s C 25 3. e eemey 0 C me me m se me mmcm C C 0 e re, 4.
c e
PB~
SG
GI
3BP 10/17 0.9 0.5 11.4 p=0.0 0 26 Copolymer 1 13/28 0.78 0.45 12.6 p= 0 0 0 0 05 Each numerical value represents the cumulative results of 3-5 independent experiments.
The p values represent the statistical significance of the difference from the control (PBS) group.
Mean maximal score is calculated for the entire group.
These data indicate that the present polypeptides are therapeutically effective for preventing the onset and severity of EAE when administered either orally or by injection.
EXAMPLE 4 BINDING TO ANTIGEN PRESENTING
CELLS
Several of the present polypeptides bind efficiently to living antigen presenting cells.
METHODS
Copolymer 1 Copolymer 1 batches 02095 and 55495, with average molecular weights of 6000 Da and 5800 Da, respectively, were obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel).
Terpolymers GAL, TGA, TAL, GTL and control polypeptides are as described above under SExample 3.
Biotinylation of antigens Biotinylation of Copolymer 1 and Terpolymers is performed at 0°C with biotin-Nhydroxysuccinimide (Sigma) according to Fridkis-Hareli et al. 91 PROC. NATL.
ACAD. SCI. USA 4872 (1994).
Binding of biotinylated antigens to antigen presenting cells The biotinylated polypeptides were examined for binding to living antigen presenting cells of mouse and human origin, using fluorescently labeled streptavidin and FACS analysis. Adherent spleen cells from (SJLJxBALB/c)FI mice, or EBV transformed human B cells of DR7,w I I haplotype (I x 10/100 pl), were incubated at 37°C for 20 hr. with 50 /g biotinylated Copolymer 1 or o" Terpolymers dissolved in 100 /l-PBS containing 0.1% BSA. The cells were then .1 incubated at 4*C for 30 min. with phycoerythrin-conjugated streptavidin (Jackson Immuno Research, West Grove, PA) at a concentration of 0.5 Ag/100 .I cell suspension. After each incubation the cells were washed three times with PBS containing 0.1% BSA. Thereafter, cells were analyzed by flow cytometry using FACScan (Becton-Dickinson, Mountain View, CA). For each analysis, 5000 cells were examined. Dead cells were excluded on the basis of forward and side-angle light scatter.
SEpstein-Barr virus (EBV) transformed B-cell lines EBV-transformed B-cell lines were initiated according to Teitelbaum et al., 89 PROC. NATL.ACAD. SCI. USA 137 (1992). Approximately 20 x 106 peripheral blood mononuclear cells were cultured with B95.8 cell line supernatant, for 1 hr at 37 The cells were then washed and cultured in RPMI medium with 10% FCS and cyclosporin A (10 g/lml) to deplete T cells.
RESULTS
Table 4 illustrates the binding of terpolymers and Copolymer 1 polypeptides to living antigen presenting cells, including mouse spleen macrophages and human _Ii EBV transformed B-cell lines. Data illustrating both the percent of binding and the intensity of cell staining are provided (Table 4 1+11). TAL bound the most efficiently better even than Copolymer 1. GAL and GTL also bound very well the same or even somewhat better than Copolymer 1. TGA bound well but somewhat less than Copolymer 1.
I, TAL bound most efficiently both to spleen macrophages of (SJL/J x BALB/c) F, mice and to EBV-transformed B cells from a normal DR7.w11 donor, as expressed by the intensity of the binding (Table 4).
Table 4 Binding of Terpolymers to antigen presenting cells I. Antigen presenting cells from mouse spleen macrophages Polypeptide Binding
MFI*
Copolymer 1 85 493 (positive control) SD-1689 GAL 88 600 SD-1690 TGA 74 39 SD-1691 TAL 100 1929 SD-1697 GTL 90 .973 *MFI Mean Fluorescence Intensity II. Antigen presenting cells from human EBV transformed B-cell line Polypeptide I Binding
MFI*
Copolymer 1 (positive control) SD-1689
GAL
SD-1690
TGA
SD-1691
TAL
SD-1697
GTL
*MFI Mean Fluorescence Intensity 96 95 72 100 97 727 661 49 1438 1057 EXAMPLE THE PRESENT POLYPEPTIDES
BIND
TO PURIFIED HUMAN LEUKOCYTE ANTIGENS (HLA) This example illustrates that polypeptides of the present invention bind to human B cells and to purified human lymphocyte antigens with high affinity, including the HLA-DR1, HLA-DR2 and HLA-DR4 molecules.
METHODS
o. o SEC o f SEC 0 4 Y r Cell lines and antibodies Homozygous EBV-transformed human B lymphocyte lines used for immunoaffinity purification of HLA-DR1, -DR2 and -DR4 molecules were LG-2 (DRBI), MGAR (DRB1-1501) and Preiss (DRB:0401/DRB4:0101), respectively.
Cells were grown in RPMI 1640 supplemented with 10% FCS, 2mM glutamine, U/ml penicillin G and 50-pg/ml streptomycin in roller bottles and stored as pellets at -80*C. The anti-DR hybridoma LB3.1 (IgG2b) is grown in serum-free medium (Macrophage-SFM, Gibco BRL). See Gorga et al., 103 CELL. IMMUNOL.
160 (1986).
Protein purification Immunoaffinity purification of HLA-DR1, -DR2 and -DR4 molecules is performed, with minor modifications, as previously reported by Gorga et al., 262 J. BIOL.
CHEM. 16087 (1987). Briefly, detergent soluble membrane preparations from LG- 2, MGAR and Priess cells were passed at a flow rate of approximately 11 ml/hr through a series of columns in the following sequence: Sepharose CL-6B (30 ml), normal mouse serum-Aff-gel 10 (10 ml), protein A-Sepharose CL-4B (5 ml) and LB3.1-protein A-Sepharose CL-4B (5 ml). DR2a (DRB5*0101) and Drw53 (DRB4*1001), the products of DR genes linked to the DRB1 alleles were not removed from the MGAR and Priess lysates before passage through the LB3.1 immunoaffinity column, and contaminate the DR2 and DR4 preparations in the amount of 5-10%. All the subsequent steps were as previously described by 37 Gorga et al., 262 J. BIOL. CHEM. 16087 (1987). The eluate is dialyzed against 0.1% deoxycholate, 10 mM Tris-HCI, pH 8.0 and concentrated on a Centriprep membrane (Amicon). Protein concentrations were determined by bicinchonitric acid assay (Pierce Chemical Co.).
Polypeptides and Control Antigens Copolymer 1 Copolymer 1 batches 55495 and 52596, with average molecular weight of 5800 daltons and 8,150 daltons, respectively, were obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel). Copolymer 1 batch 52596 had a molar ratio of 1 tyrosine: 1.5 glutamic acid: 4.3 alanine: 3.1 Slysine.
*o Myelin Basic Protein and Hemagglutinin Control Peptides MBP peptides were synthesized on an Applied Biosystems Peptide Synthesizer using solid phase techniques. Barany et al., THE PEPTIDES 1 (1979). Peptides purified by reversed-phase HPLC. The peptides used Swere HA 306-318, having the sequence PKYVKQNTLKLAT (MW 1718) (SEQ ID NO: and MBP 84 102, having the sequence DENPWHFFKNIVTPRTPP (MW 2529) (SEQ ID NO: 1).
Terpolymers GAL, TGA, TAL, GTL and control polypeptides are as described above under Example 3.
Polypeptide labeling Biotinylation of the various polypeptides is performed as in Example 4. Unreacted biotin is removed by dialysis (Spectra/Por@ membrane MWCO 500, Spectrum Medical Industries).
Assays for polypeptide binding to class II MHC proteins Solutions: The solutions used in this assay were the following. Binding buffer is 20 mM 2-[N-morpholino]ethanesulfonic acid (MES), 1% n-octyl 3- D-glycopyranoside, 140 mM NaCI, 0.05% NaN 3 pH 5.0, unless otherwise specified. PBS is 150 mM sodium chloride, 7.5 mM sodium phosphate, dibasic, 2.5 mM sodium phosphate, monobasic, pH 7.2. TBS is 137 mM sodium chloride, 25 mM Tris pH 8.0, 2.7 mM potassium chloride. TTBS is TBS plus 0.05% Microtiter Assay Plate Preparation: Ninety-six well microtiter immunoassay plates (PRO-BIND
TM
Falcon) were coated with 1 ug/well affinity-purified LB3.1 monoclonal antibodies in PBS (100 pl total) for 18 hrs at 4"C. The wells were then blocked with TBS containing 3% BSA for 1 hr at 37 0 C and washed three times with TTBS. Before sample addition, 50 /l of TBS/1% BSA is added to each well.
Binding reactions: -Detergent=solubilized -HLA-DR1, -DR2 and -DR4 molecules g/sample) were incubated with biotinylated control peptides at various concentrations for 40 hours at 37*C in 50 Ul of the binding buffer and transferred to prepared microtiter assay plates and incubated for 1 hr at 37°C for capture of polypeptide-class II complexes.
Inhibition reactions: Biotinylated polypeptides at a final concentration of 1.5 IM in 50 /l of binding buffer were coincubated with unlabeled polypeptides as well as the peptides HA 306-318 (SEQ ID NO: 2) or MBP 84-102 (SEQ ID NO: used as inhibitors, and HLA-DR molecules for hr at 37"C.
Detection of class Illpolypeptide complexes: Bound polypeptide-biotin is detected using streptavidin-conjugated alkaline phosphatase as follows.
Plates were washed three times with TTBS and incubated with 100 Jl of streptavidin-conjugated alkaline phosphatase (1:3000, BioRad) for 1 hr at 37°C, followed by addition of p-nitrophenyl phosphate in triethanolamine a,'b buffer (BioRad). The absorbency at 410 nm is monitored by a microplate reader (Dynatech MR4000).
RESULTS
Terpolymers Binding to Class II MHC Proteins Detergent-soluble HLA-DR1, HLA-DR2 and HLA-DR4 proteins were purified from homozygous EBV-transformed B cell lines LG-2 (DRB1*0101),
MGAR
(DRB1*1501) and Priess (DRB1*0401), respectively, as described previously by Fridkis-Hareli et al., 160 J. IMMUNOL. 4386 (1998). Three different preparations of Copolymer 1 had bound to these molecules with high affinity. Id. To determine the affinity of the terpolymers for HLA-DR proteins, binding assays were carried out with biotinylated Terpolymers, and compared to Copolymer 1. The polypeptides were incubated at a range of concentrations with purified HLA-DR1, HLA-DR2 and HLA-DR4 molecules at pH 5.0 followed by capture with class IIspecific mAb and detection with alkaline phosphatase-streptavidin.
0 0 0 00 sees Binding byTAt-and-Copolymer 1 tcrdetergent-soluble HLA-DR1 and HLA-DR4 molecules is better than that of GAL, TGA or GTL. However, GTL and Copolymer 1 bound better than the other polypeptides to HLA-DR2, based on the saturation binding curves (Fig. 2A), and on K, values, calculated from the double-reciprocal plots of the binding data (Fig. 2B, Table Competitive binding assays were carried out with biotinylated Copolymer 1 and the following unlabeled inhibitors: Copolymer 1, TAL, TGA, GAL, TGL, MBP 84- 102 and HA 306-318 polypeptides (Fig. The MBP 84-102 (SEQ ID NO: 1) peptide is a poor inhibitor of the binding of Copolymer 1 to HLA-DR2. The binding of biotinylated Copolymer 1 to detergent-soluble HLA-DR1 and -DR4 molecules is efficiently inhibited by unlabeled TGA, TAL, HA 306-318 (SEQ ID NO: 2) and Copolymer 1. However, binding of biotinylated Copolymer 1 to detergent-soluble HLA-DR1 and -DR4 molecules is inhibited more than less by TGL, as indicated by 50% inhibitory dosages (IC5o) (Fig. 3, Table Similarly, GAL is also a poor inhibitor of Copolymer 1 binding to HLA-DR1 and DR4 molecules. In general, the binding pattern to HLA-DR2 is similar to that
SEC
104 three amino acids, in particular TAL and TGA bind to class 11 MHC molecules with an affinity range similar to that of antigenic peptides and of Copolymer 1. Hence, TAL and TGA are effective competitors for the class 11 MHC molecules to which Copolymer 1 binds. Based on their binding capacity, the polypeptides could be arranged in the following order: binding to HLA-DRI: Copolymer I TAL GTL TGA GAL; binding to HLA-DR2: Copolymer 1 GTL TAL GAL TGA; binding to HLA-DR4: TAL Copolymer 1 TGA GTL GAL.
to ev*.: .9 see
S.
S
S.
*SSS
*5 S Table Affinity of the present polypeptides for purified human HLA-DR1, -DR2 and -DR4 molecules Polypeptide' DR1 2 DR2 DR4
K
1 3
IC
5 0 4 Kd IC 5 0 K ICso Copolymer 1 7.4 8.8 8.2 10.1 1.5 10.8 TAL 2.0 3.3 1.7 2.7 2.0 GAL 0.5 s 1.7 s 0.3 TGA 1.0 1.3 0.8 9.5 0.8 TGL 0.4 43.0 5.0 25.4 0.8 43.0 Effect of superantigens on the binding of polypeptides to HLA-DR molecules Bacterial superantigens SEA, SEB and TSST-1 have been shown to inhibit Copolymer 1 binding to purified HLA-DR molecules only at very high concentrations. Fridkis-Hareli et al., 160 J. IMMUNOL. 4386 (1998). To examine the effect of these superantigens on the binding of Terpolymers to purified HLA- Copolymer 1 polypeptides with average MW of 5,800; TAL, MW 20,000; GAL, MW 8,850; TGA, MW 7,600; and TGL, MW 11,050, were incubated at a range of concentrations with purified HLA-DR1, -DR2 and -DR4 molecules at pH followed by capture with class II-specific mAb and peptide detection with alkaline phosphatase-streptavidin.
2 Detergent-soluble HLA-DR1, -DR2 and -DR4 molecules were purified as described in Materials and Methods.
3 Kd, the dissociation constant at equilibrium, is calculated from the slope of the double reciprocal plot (Fig. 2B), and is expressed as x 10 M.
4 ICo, inhibitory concentration giving 50% inhibition, is calculated based on the competitive binding assays (Fig. and is expressed as x 106 M.
ICso values for GAL could not be determined exactly, but were less than 1000 MM (see Fig. 3).
DR1, HLA-DR2 and HLA-DR4 molecules, competitive binding assays were carried out with unlabeled SEA, SEB and TSST-1.
Binding of TAL to HLA-DR1, HLA-DR2 and HLA-DR4 is only inhibited by the superantigens at high molar ratios of superantigen: for example, fifty times the amount of superantigen is needed to inhibit TAL binding (Fig. 4A). However, binding of TGA and GAL is inhibited more significantly by the superantigens (Fig.
4B and indicating that these polypeptides bound the HLA antigens with lower affinity.
EXAMPLE 6 INHIBITION OF MBP-INDUCED T CELL PROLIFERATION This example illustrates that the proliferation of T cells which are normally 15: activated by myelin basic protein (MBP) can be inhibited by simultaneous exposure to the present polypeptides.
METHODS
Copolymer 1 i Copolymer 1 batches 02095 and 55495, with average molecular weights of 6000 Da and 5800 Da, respectively, were obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel).
Myelin Basic Protein Peptides were synthesized on an Applied Biosystems Peptide Synthesizer using solid phase techniques. Barany et al., THE PEPTIDES 1 (1979). Peptides purified by reversed-phase HPLC. The peptides used were HA 306-318, having the sequence PKYVKQNTLKLAT (MW 1718)(SEQ ID NO: and MBP 84 102, having the sequence DENPWHFFKNIVTPRTPP
(MW
2529) (SEQ ID NO: 1).
Terpolymers GAL, TGA, TAL, GTL and control polypeptides are as described above under Example 3.
T cell lines and clones Myelin Basic Protein (MBP) specific T-cell lines originated from spleens of mice which, ten days earlier, were immunized with 20 pg of the 84-102 peptide of MBP emulsified in complete Freund's adjuvant supplemented with 4 mg/ml of Mycobacterium tuberculosis H37Ra. Cells were cultured and selected in vitro using the immunizing antigen (0.2-1 mg/plate), in culture medium (RPMI, 2 mM glutamine, 1 mM sodium pyruvate, non essential amino acids, 5 x10- M 2-mercaptoethanol, 100 Ag/ml penicillin, 100 pg/ml streptomycin), supplemented with 1% autologous serum. After i: 4 days, cells were transferred to culture medium containing 10% FCS and supplemented with 10% supernatant of Con A activated normal spleen cells as T cell growth factor (TCGF). Every fourteen to twenty one days, cells were stimulated by exposure to the immunizing antigen presented on 1. syngeneic irradiated (3000 rad) spleen cells (50 x 10 6 /plate) for 3 days, followed by propagation in TCGF medium. Cloning of T cell lines is performed by limiting dilution at 0.3 cells/well in microliter plate in the cl"'s presence of the antigen (2-10 pg/well) and irradiated syngeneic spleen cells (5 xI 0 6 /well).
Human T cell lines were derived from peripheral blood mononuclear cells according to Teitelbaum et al., 89 PROC. NATL. ACAD. SCI. USA 137 (1992).
Approximately 5 x 106 cells were incubated in each well of a 24-well culture plate with Copolymer 1 or MBP (50 mg/ml) in culture medium supplemented with 10% heat-inactivated autologous serum. After 7 days of culture, the cells were transferred to culture medium containing 10% fetal calf serum and recombinant human interleukin 2 (20 units/ml). The cells were grown continuously in this medium with periodic exposure to antigen presented on irradiated (3000 rad), autologous mononuclear cells, every 14-18 days.
Proliferation assay Cells of T lines or clones were tested for their specific proliferative response 10-21 days after antigenic stimulation. T Cells (1.5 x 10 4 were cultured in triplicates with I' 5 x 105 irradiated spleen cells and with the indicated antigens in a final volume of 0.2 ml in 10% FCS culture medium. At the end of 48 hr incubation, cultures were pulsed with 1 4Ci[ 3 H]-thymidine (standard deviation <20% of the mean cpm), and harvested 6-12h later.
Inhibition studies Inhibition of the T-cell proliferative response is studied by adding various concentrations of Copolymer 1 and Terpolymers plus the stimulating MBP antigen to the proliferation assay system. Inhibition is calculated as percent inhibition using equation 2:
I
Inhibition [1 (cpm with inhibitor cpm without inhibitor)] X 100. 2
RESULTS
Figure 1 illustrates how copolymer 1 and terpolymers effect the proliferation of T i cells which are specific for certain myelin basic protein (MBP) peptides. In general, T cells proliferate when exposed to the antigen to which they were sensitized. Thus, MBP and, in particular, certain antigenic peptides from MBP 20 stimulate the proliferation of MBP-specific T cells.
Copolymer 1 and Terpolymers did not stimulate proliferation of T cells which were specific for the 84-102 peptide of MBP. Instead, they significantly inhibited the proliferation of T cells exposed to this MBP antigen.
TAL exhibited the most efficient inhibition of proliferation in T cells specific for the MBP 84-102 antigen. Inhibition by TAL is even greater than that provided by Copolymer 1. TGA induced lower inhibition of T cell proliferation. GAL and GTL inhibited T cell proliferation in a dose responsive manner similar to Copolymer 1.
EXAMPLE 7 TERPOLYMERS ARE RECOGNIZED BY SOME COPOLYMER 1-SPECIFIC T CELLS Terpolymers can stimulate some Copolymer 1-specific T cell lines to .I proliferate and secrete the cytokine, IL-4.
METHODS
Copolymer 1 Copolymer 1 batches 02095 and 55495, with average molecular weight of 6000 Da and 5800 Da, respectively, were obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel).
9* Myelin Basic Protein Peptides were synthesized on an Applied Biosystems S: Peptide Synthesizer using solid phase techniques. Barany et al., THE PEPTIDES 1(1979). Peptides purified by reversed-phase HPLC. The peptides used were i HA 306-318,- having-the- sequence PKYVKQNTLKLAT-(MW 1718) (SEQ ID NO: 2),-and MBP 84102, having the sequence DENPWHFFKNIVTPRTPP
(MW
2529) (SEQ ID NO: 1).
Terpolymers GAL, TGA, TAL, GTL and control polypeptides are as described above under Example 3.
T cell lines and clones Mouse T cell lines and clones were established according to Aharoni et al., 23 Eur. J. Immunol. 17 (1993); Aharoni et al., 94 PROC. NATL. ACAD.
Sci. USA 10821 (1997). Copolymer 1 specific lines were originated from spleens of mice which, 15 to 35 days earlier, had been rendered unresponsive to EAE by subcutaneously injecting each mouse with 5-10 mg Copolymer 1, emulsified in incomplete Freund's adjuvant (Difco).
Alternatively, Copolymer 1 specific lines were obtained from the lymph nodes of mice which had been immunized ten days earlier with 200 1g Copolymer 1 emulsified in complete Freund's adjuvant (Difco) supplemented with 4 mg/ml of Mycobacterium tuberculosis H37Ra (Difco).
Copolymer 1-specific T cell lines were used and T cell clones LN-1, LN-2, S-3, 5-22-5, C-14, and C-52. The LN-2 antibodies were derived from lymph nodes of (SJLJ x BALB/c) F, mice injected with Copolymer 1 in CFA. The 5-22-5 antibodies were obtained from spleens of (SJL/J x BALB/C) F, mice injected with Copolymer 1 in CFA, according to Aharoni et al., 23 EUR. J. IMMUNOL. 17 (1993); Aharoni et al., PROC. NATL. ACAD. Sci.
USA 10821 (1997).
Human T cell clones were derived from peripheral blood mononuclear cells according Teitelbaum et al., 89 PROC. NATL. ACAD. Sci. USA 137 (1992). Approximately 5 x 106 cells were incubated in each well of a 24well culture plate with Copolymer 1 or MBP (50 pg/ml) in culture medium supplemented with 10% heat-inactivated autologous serum. After 7 days of culture, the cells were transferred to culture medium containing i: fetal calf serum and recombinant human- interleukin 2 (20 units/ml). The cells were grown-continuously in this medium with periodic exposure to antigen presented on irradiated (3000 rad), autologous mononuclear cells, every 14-18 days. The C-52 T-cell clone was derived from a DR7,w11 donor, also according to Teitelbaum et al., 89 PROC. NATL. ACAD. SCl. USA 137 (1992).
Proliferation assay Cells ofT lines or clones were tested for their specific proliferative response 10-21 days after antigenic stimulation. T Cells (1.5 x 104) were cultured in triplicates with x l0 5 irradiated spleen cells or with human EBV-transformed B cells (5x104), and with the indicated antigens in a final volume of 0.2 ml in 10% FCS culture medium.
At the end of 48 hr incubation, cultures were pulsed with 1 ACirH]-thymidine and then harvested 6-12 hr later. The variations of triplicates from their mean were under 15 cell line cross-reacted only with TAL. No cell lines cross reacted with GfL or with AGT which has the same amino acids as TGA but in a different mole fraction than TGA or Copolymer 1.
Table 6 Cross reactivity of Terpolymers with Copolymer 1 for Copolymer I specific T-cell lines and clones 1. Murine T-cell lines and clones Cross reactivity with Copolymer 1 T-cells I LN -3 1 S-3 S-22-5 Polypeptide prol* L-4* prl L- prol*r L* SD-1689 -GAL 130 139 49 11 0 1 0 0 SD-1690 -TGA 3 6 102 107 0 1 0 0 SD-i 691 -TAL 3 7 2 3 64 120 0 0 SD-1697 -GTL 2 4 12 6 0 1 0 1:1:1IAGT 2 0 2 2 1 0 2 OT *prol as measured by proliferation,- *IlL7-= as measured by lnterl eukin-4.
II. Human T-cell Clones Cross reactivity with Copolymer
I(%
T-cells 1C-14 C-52 Polypeptide proliferation poieaion SD-I1689 GAL 4 SD-1690 -TGA 1 58 SD-1691 -TAL 0 0 SD-1697 GTL 0 EXAMPLE 8 CROS-EACT WITH ANTI-COPOLYMER I ANTIBODIES T~0DAI YMFRS I ~r Antibodies directed against Copolymer I cross-react with Terpolymers which lack either tyrosine, glutamic acid or alanine. However, Terpolymers lacking lysine are not recognized efficiently by anti-Copolymer 1 antibodies.
Antibodies Mouse anti-MBP and anti-Copolymer 1 monoclonal antibodies were obtained by fusion of MBP- or Copolymer 1-immunized spleen cells from SJUJ mice, with the NSO/1 murine plasmacytoma cell line. Teitelbaum et al., Proc. Natl. Acad. Sci.
USA 9528 (1991).
Radioimmunoassay Flexible plastic microtiter plates were coated with Copolymer 1 or Terpolymers (2 g/ml). After 16 hr incubation at room temperature, plates were washed three times and saturated for 2 hr with PBS containing 2% bovine serum albumin, 0.05% Tween 20, 0.1% sodium azide, 10 mM EDTA, and heparin at 5 units/ml S: ('PBS buffer"). Monoclonal antibody supernatants (50 ul), were added to the wells for a 2 hr incubation, and the wells were washed again with PBS buffer. 125l labeled goat anti-mouse Fab antibodies (1 x 10 5 cpm/well) were added for :overnight incubation at 4=C. After extensive washing, radioactivity is measured in a gamma counter.
METHODS
ELISA Assay for Antibody Cross-Reactivity A standard ELISA assay is employed using anti-Copolymer 1 polyclonal antibodies and microtiter plates coated with 2 /g/ml of terpolymer preparation.
Copolymer 1 Copolymer 1 with the following amino acid composition is obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel).
Amino Acid Molar Fraction I=sR- L-glutamic acid 0.141 l L-alanine 0.427 L-tyrosine 0.095 L-lysine 0.338 Terpolymers IThe four Terpolymers of Example 1 were used.
RESULTS
Table 7 indicated that anti-Copolymer 1 polyclonal antibodies cross react with Terpolymers which lack either tyrosine, glutamic acid or alanine. The relatively high percentage binding of the terpolymers lacking glutamic acid (TAL) might be explained by its high average molecular weight. Terpolymers which lack lysine are not efficiently recognized by anti-Copolymer 1 antibodies. These data :6 suggest that charged amino acids like lysine may play a role in the recognition and binding of Copolymer 1 and Terpolymers.
Table 7 o 25 30 a Percent Binding in separate experiments Terpolymers MW* 1 2 3 3 Mean S.D.
TAL 20,000 111.1 130.1 115.8 114.0 117.8 8.48 GAL 8,850 7.5 10.8 9.3 9.2 9.2 1.35 GTL 11,050 98.7 87.0 n.d.s 92.9 8.30 TGA 7,600 87.9 79.3 83.6 6.12 *daltons. n.d. not determined.
Cross reactivity of Terpolymers with Copolymer 1-reactive monoclonal antibodies The cross reactivity of the Terpolymers with Copolymer 1 at the level of B cell response is tested using monoclonal antibodies (mAbs), that are either reactive with both Copolymer 1 and MBP (mAbs 2-2-18 and 3-1-45), or are reactive with only Copolymer I (mAbs 3-3-9 and See Teitelbaum et al., 88 Proc. Natl.
Acad. Sci. USA 9528 (1991).
Table 8 illustrates that the Terpolymers differed in their ability to bind these mAbs.
TGA and GTL were not recognized by any of the Copolymer 1 specific mAbs. On the other hand, TAL and GAL bound to Copolymer 1 and MBP specific mAbs with an affinity which is similar to that of Copolymer 1. GAL and TAL differed only in the binding to one mAb i.e. 5-7-2 which bound to TAL and not to GAL.
Table 8 Cross reactivity of Terpolymers with MBP- and Copolymer 1-reactive B cell antibodies
S*
S. S
S
S*
S...0 Cross reactivity with Copolymer 1 Antibody 2-2-18 3-1-45 3-3-9 5-7-2 Polypeptide SD-1689- GAL 96 98 107 SD-1690 TGA -3 3 2 1 SD-1691 TAL 96 -98 103 106 SD-1697 GTL 1 2 1 1 2-2-18 is anti mouse MBP monoclonal antibody cross reactive with Copplymer 1 3-1-45is anti Copolymer 1 monoclonal antibody cross reactive with MBP 3-3-9 is anti Copolymer 1 monoclonal antibody non cross reactive with MBP 5-7-2 is anti Copolymer 1 monoclonal antibody non cross reactive with MBP EXAMPLE 9 COPOLYMER 1 AND TERPOLYMERS COMPETE WITH COLLAGEN FOR BINDING TO HUMAN LEUKOCYTE ANTIGENS AND INHIBIT COLLAGEN-SPECIFIC T-CELL RESPONSE This example illustrates that Copolymer 1, TAL, GAL, GTL, and TGA compete for binding to MHC proteins with the rheumatoid arthritis-associated immunodominant collagen antigen, CII 261-273 (SEQ ID NO: 3).
METHODS
Protein Expression and Purification Recombinant HLA-DR1 and HLA-DR4 molecules (encoded by DRA/DRB I *0101 5 and *0401, respectively) were expressed in Drosophila S2 cells as described in Stem, L. et al. 68 CELL 465 (1992) and Dessen, A. et al. 7 IMMUNrTY 473 (1997).
Cells were grown in roller bottles at 26C in Excell 401 medium (Sigma, St. Louis, MO) supplemented with 0-5% fetal bovine serum (Sigma). Cells were induced by addition of CuSO to 1mM final concentration, then incubated an additional 20 days. Immunoaffinity purification of recombinant HLA-DR1 and HLA-DR4 is performed as previously reported by Stem, L. et al. 68 CELL 465 (1992) and Dessen, A. et al. 7 IMMUNITY 473 (1997). Supernatant from harvested cells is sequentially passed through Protein A, Protein G and Protein A-LB3.1 columns, followed by elution of the bound HLA-DR with 50 mM 3cyclohexylamino-1propane sulfonic acid (CAPS), pH 11.5, and neutralized with 200 mm phosphate (pH The eluate is concentrated on a Centriprep 10 membrane (Amicon).
Protein concentrations were determined by bicinchoninic acid assay (Pierce Chemical Co.).
Polypeptide labeling The present polypeptides and the HA 306-318 peptide were biotinylated as in Examples 4 and 7.
Class II MHC protein binding assay In this assay, water-soluble recombinantly-produced proteins were incubated with biotinylated polypeptides of the present invention and varying quantities of unlabeled competitor polypeptides, collagen CII peptides or influenza virus HA peptides. Assays were performed in 96-well microliter immunoassay plates (PRO-
BOND
M
Falcon) which were coated with affinity-purified LB3.1 monoclonal antibodies. Antibody coating is performed by placing 100 Pl of 10.0 mg/ml LB3.1 monoclonal antibodies in each well and incubating at 4"C for 18 hrs. Microtiter wells were then blocked with Tris buffered saline (TBS) containing 3% bovine serum albumin (BSA) for 1 hr at 37°C and washed three times with TTBS. Before sample addition, 50 Al of TBS containing 1% BSA is added to each well.
Phosphate buffered saline (PBS) is 150 mM sodium chloride, 7.5 mM sodium phosphate dibasic, 2.5 mM sodium phosphate monobasic, pH 7.2. Tris buffered saline (TBS) is 137 mM sodium chloride, 25 mM TRIS pH 8.0, 2.7 mM potassium chloride. TTBS is TBS with 0.05% Tween-2G. Other solutions used in this assay are described in Fridkis-Hareli, M. et al., 160 J. IMMUNOL. 4386 (1998).
Binding analysis is performed by incubating the water soluble DR molecules with biotinylated polypeptides of the present invention and varying concentrations of unlabeled inhibitors (Copolymer 1, TAL, GAL, GTL, TGA, Collagen CII 261-273 peptide or HA 306-318 peptide). The collagen type CII peptide 261-273, has SEQ "ID NO: 3 (AGFKGEQGPKGEP) and a molecular weight of 1516. The concentration of DR employed-isrO-t5-zM-The final concentration of biotinylated Copolymer 1 or terpolymers is 1.5 pM. Incubation is for 40 hr at 37°C in 50 pl binding buffer at pH Bound label is detected using streptavidin-conjugated alkaline phosphatase as follows. Plates were washed three times with TTBS and incubated with 100 l of streptavidin-conjugated alkaline phosphatase (1:3000, BioRad, Richmond,
VA)
for 1 hr at 37°C, followed by addition of p-nitrophenyl phosphate in triethanolamine buffer (BioRad). The absorbency at 410 nm is monitored by a microplate reader (model MR4000, Dynatech, Chantilly, VA).
T cell hybridoma and antigen presenting cell (APC) binding assays Copolymer 1 and Terpolymers were tested to ascertain if they could inhibit activation of T cells responsive to the collagen CII peptide. Mouse DR1-restricted 3.19 and 19.3 T cell hybridomas and mouse DR4-restricted 3838 and D3 T cell l hybridomas were used. Rosloniec, E. et al., 185 J. Exp. MED. 1113-1122 S(1997); Andersson, E. et al., PROC. NATL. ACAD. Sci. USA (1998). Antigen presenting cells (APCs) were L cells transfected with DR1 (the L57.23 cell line provided by Rosloniec, E. et al., 185 J. Exp. Med. 1113-1122 (1997)), L cells transfected with DR4, and Priess cells (DRB1*0401/DRB4*0101). T cell stimulation experiments were performed in 96-well microliter plates in a total *i volume of 0.2 ml. Irradiated (3000-rad) APC (2.5 x 10^/well) were coincubated with CII 261-273 (40 mg/ml) and varying concentrations of the present polypeptides for 2 hr at 37"C, then T cells (5 x 10 4 /well) were added and the 15 incubation is continued for 24 hr at 37°C. Supernatants (30 l) were removed and incubated with IL-2 dependent CTL-L (5 x 10'/well) for 12 hr, followed by labeling with 3 H-thymidine (1 /Ci/well) for 12 hr. Plates were harvested and the radioactivity-is monitored using a 1450 microbeta Plus liquid scintillation counter (Wallac, Gaithersburg, MD).
RESULTS
g Class II MHC protein binding assay a The recombinant water-soluble HLA-DR1 and -DR4 proteins produced in insect cells were largely free of bound autoantigens or other peptides. Hence, data obtained from insect cell produced proteins can more accurately indicate the actual binding affinities for polypeptides. Fridkis-Hareli et al., 160 J. IMMUNOL.
4386 (1998) (the entire contents of which are hereby incorporated herein by reference).
Competitive binding of each of the Copolymer 1 and Terpolymers to HLA-DR1, HLA-DR2 and HLA-DR4 molecules is depicted in Figure 5. Binding of each of the Copolymer 1 (YEAK, top panel Fig. 5A) and Terpolymers is substantially R -4reater than that of the CII 261-273 (SEQ ID NO: 3) peptide, as judged by SEC 104 54 ,-y quantity of CII 261-273 (SEQ ID NO: 3) peptide required for 50% inhibition. As also observed above, TAL bound HLA-DR1 and HLA-DR4 with greater affinity than did Copolymer 1. The kinetics of inhibition by unlabeled TAL (YAK, bottom panel Fig. 5A) polypeptides were also somewhat superior to that of the influenza virus peptide HA 306-318 (SEQ ID NO: However, the influenza virus peptide HA 306-318 inhibited the binding of Copolymer 1 and of Terpolymers more efficiently than the CII 261-273 peptide.
T cell Hybridoma and antigen presenting cell binding assay results Copolymer 1 and Terpolymers also inhibited DR1-restricted T cell activation by CII collagen peptide (Fig. T cell activation was detected by observing IL-2 production by DR1-restricted-CII-specific T cell hybridomas. Collagen peptide CII 261-273 (SEQ ID NO: 3) at varying concentrations was coincubated with one of the present polypeptides and then T cells (clone 3.19 or 19.3 as indicated) were added, and the mixtures were further incubated. The supematants from these incubated cells are removed, and were assayed for IL-2 by observing whether the 2 supernatant induced proliferation of IL-2-dependent cytotoxic T lymphocytes (CTL-L). The extent of-inhibition by TAL is shown as solid circles by TGA as solid triangles by GTL as open triangles and by Copolymer 1 as solid squares Percent inhibition of CTL-L proliferation shown on the ordinate was calculated using equation 1.
9 Again, TAL is the most potent inhibitor. However, GTL-andcCopolymer 1 were also potent inhibitors of T cell activation by the Cil collagen peptide. TGA inhibited activation less efficiently. Similar results were obtained with other batches of Copolymer 1 and Terpolymers.
A similar inhibition of activation of DR4-restricted T cells was observed, as shown in Figure 7. IL-2 production was used to assess activation of DR4-restricted
CII-
specific T cell hybridomas (3838 and D3). The presence of different polypeptides inhibited IL-2 production, indicating that they inhibited DR4-restricted T cell activation. Fig. 7A shows the effects of coincubating irradiated 3838 or D3 Priess cells with collagen peptide CII 261-273 (SEQ ID NO: 3) at the fixed concentration of 40 /g/ml, and with varying concentrations of polypeptides, for 2 hr at 37°C.
Fig. 7B shows the effects of incubating L cells transfected with a gene encoding HLA-DR4 with collagen peptide CII 261-273 (SEQ ID NO: 3) at the fixed concentration of 40 pg/ml, and with varying concentrations of GAL, TAL, GTL, TGA, and Copolymer 1, for 2 hr at 37°C. T cells were then added (clones 3838 or D3 as indicated), and samples were further incubated for 24 hr at 370C.
l Supernatants (30 were then removed, and were assayed for activation by IL-2induced proliferation of IL-2-dependent cytotoxic T lymphocytes (CTL-L). Each polypeptide mixture was tested in duplicate. The concentration of the present polypeptides is indicated on the abscissa. The extent of inhibition by TAL is shown as solid circles by TGA as solid triangles by GTL as open triangles(A), and by Copolymer 1 as solid squares Percent inhibition of CTL- L proliferation shown on the ordinate was calculated using equation 1.
EXAMPLE COPOLYMER 1 INHIBITS ACTIVATION OF T CELLS RESPONSIVE TO A MYASTHENIA GRAVIS ANTIGENIC PEPTIDE .i METHODS- Copolymer 1 Copolymer 1 was obtained from Teva Pharmaceutical Industries (Petach Tikva, Israel).
Myasthenia Gravis-Related Peptides were synthesized on an Applied Biosystems Peptide Synthesizer using solid phase techniques. Barany et al., THE PEPTIDES 1 (1979). Peptides were purified by reversed-phase HPLC. The peptides used were the p259 peptide, see Zisman et al., Hum. Immuol. 1995 Nov.; 44(3):121-30; Brocke et al., Immunology 1990 Apr.; 69(4):495-500.
IL-2 secretion Secretion of IL-2 by the cell line WCB AB in response to the myasthenia gravis peptides andlor Copolymer 1 were evaluated. Cells (1.5 x 10 4 were incubated with the indicated antigen. Secretion of IL-2 by the cell line WCB AB was used as a measure of activation of that T cell line.
3 SEC 56 M 104 v_
RESULTS
Table 9 indicates that the p259 peptide stimulates T cell secretion. However, when Copolymer 1 is incubated with the p259 peptide, T cell secretion of IL-2 is inhibited in a dose-related fashion. At 100 /AM Copolymer 1 inhibits about 91% of IL-2 secretion (Table 10), indicating that Copolymer 1 is a potent inhibitor of T cell activation.
Table 9 IL-2 secretion from WCB AB line in response to p259 259 conc. Avg OD SD %CV IL-2 (AM) (450 nm) (pg/ml) 0 0.077 0.01 0 ."t5 0.25 0.135 0.00 24 0.227 0.01 3.9 72 1 0.387 0.01 2.9 159 2 0.725 0.02 2.4 347
S
a go oj Table IL-2 secretion from WCB AB line in response to Copi alone or 2/iM p259 Copi Cop1 Avg OD IL-2 SD %CV Inhibition (iM) (450 nm) (pg/ml) 0 0.725 0.02 2.4 347 1 0.086 0.01 0 2 0.079 0.00 0 0.086 0.00 0 0.266 0.01 8.0 93 73 0.166 0.01 40 88 100 0.151 0.01 11.2 32 91 Absorbance is the average of 3 samples. Confidence values were calculated relative to the absorbance of a blank (%CV SDI(Avg-blank)*100).
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form or suggestion that that prior art forms part of the common general knowledge in Australia.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
EDITORIAL NOTE APPLICATION NUMBER 52262/99 The following Sequence Listing pages 1/16 to 16/16 are part of the description. The claims pages follow on pages 59 to 66.
WO 00/05250 PCT/US99/16747 SEQUENCE LISTING <110> Aharoni, Rina Teitelbaum, Dvora Arnon, Ruth Sela, Michael Fridkis-Harelli, Masha Strominger, Jack <120> Treatment of Autoimmune Conditions with Copolymer 1 and Related Copolymers and Peptides <130> 1662/493762 <140> filed herewith <141> 1999-07-23 <150> US 60/093,859 <151> 1998-07-23 <150> US 60/101,825 <151> 1998-09-25 <150> US 60/102,960 <151> 1998-10-02 <150> US 60/106,350 <151> 1998-10-30 <150> US 60/108,184 <151> 1998-11-12 <150> US 60/123,675 <151> 1999-03-09 <160> 57 <170> WordPerfect 8.0 for Windows <210> 1 <211> 19 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide (MBP residues 84-102) <400> 19 Asp Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr Pro Arg 1 5 10 Page 1 of 16 WO 00/05250 PCT/US99/16747 Thr Pro Pro 19 <210> 2 <211> 13 <212> PRT <213> Artificia <220> 1 Sequence <223> <400> Pro Ly 1 synthetic peptide (HA residues 306-318) 2 's Tyr Val Lys Gin Asn Thr Leu Lys Leu Ala Thr 5 10 13 <210> 3 <211> 13 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide (CII amino acids 261-273) <400> 3 Ala Gly Phe Lys Gly Glu Gin Gly Pro Lys Gly Glu Pro 1 5 10 13 <210> 4 <211> <212> PRT <213> Artificial Se< <220> <223> Synthetic pepi <400> 4 Ala Glu Lys Tyr Ala quence tide 1 <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide Page 2 of 16 WO 00/05250 PCT/US99/16747 <400> Ala Glu Lys Val Ala 1 <210> 6 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 6 Ala Glu Lys Phe Ala 1 <210> 7 <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 7 Lys Glu Tyr Ala 1 4 <210> 8 <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 8 Lys Tyr Ala Glu 1 4 <210> 9 <211> 4 <212> PRT <213> Artificial Sequence <220> Page 3 of 16 WO 00/05250 PCT/US99/16747 <223> synthetic peptide <400> 9 Lys Glu Val Ala <210> <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400>.10 Lys Val Ala Glu <210> 11 <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 11 Lys Glu Phe Ala <210> 12 <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 12 Lys Phe Ala Glu <210> 13 <211> 4 <212> PRT <213> Artificial Sequence Page 4 of 16 WO 00/05250 PCT/US99/16747 <220> <223> synthetic peptide <400> 13 Lys Tyr Ala Ala <210> 14 <211> 4 <212> PRT <213> Artificial Sequence <220> <223>.synthetic peptide <400> 14 Lys Lys Tyr Ala 1 4 <210> <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Lys Val Ala Ala <210> 16 <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 16 Lys Lys Val Ala 1 4 <210> <211> <212> 17 4
PRT
Page 5 of 16 WO 00/05250 PCT/US99/16747 <213> Artificial Sequence <220> <223> synthetic peptide <400> 17 Lys Phe Ala Ala <210> 18 <211> 4 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 18 Lys Lys Phe Ala <210> 19 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 19 Ala Lys Tyr Ala Glu <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Glu Ala Lys Tyr Ala <210> 21 <211> Page 6 of 16 WO 00/05250 PCT/US99/16747 <212> PRT <213> Artificial Sei <220> <223> synthetic pep <400> 21 Ala Lys Val Ala Glu quence tide <210> 22 <211> <212> PRT <213>.Artificial Sequence <220> <223> synthetic peptide <400> 22 Glu Ala Lys Val Ala <210> 23 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 23 Ala Lys Phe Ala Glu 1 <210> 24 <211> <212> PRT <213> Artificial Sequen <220> <223> synthetic peptide <400> 24 Glu Ala Lys Phe Ala 1 ce <210> Page 7 of 16 WO 00/05250 PCT/US99/16747 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Ala Lys Glu Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 26 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 26 Ala Ala Glu Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 27 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 27 Ala Ala Lys Tyr Ala Glu Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 28 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 28 Glu Ala Lys Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 Page 8 of 16 WO 00/05250 PCT/US99/16747 <210> 29 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 29 Ala Glu Lys Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> <211>.15 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Lys Glu Ala Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 31 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 31 Ala Glu Glu Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 32 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 32 Glu Lys Ala Tyr Ala Ala Ala Ala Ala Ala. Lys Ala Ala Ala Ala 1 5 10 Page 9 of 16 WO 00/05250 PCT/US99/16747 <210> 33 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 33 Ala Ala Lys Tyr Glu Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 34 <211>.15 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 34 Glu Ala Ala Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Glu Lys Lys Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 36 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 36 Ala Lys Lys Tyr Glu Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 Page 10 of 16 WO 00/05250 PCT/US99/16747 <210> 37 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 37 Ala Ala Glu Tyr Lys Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 38 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 38 Ala Ala Lys Tyr Glu Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 39 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 39 Ala Ala Lys Tyr Ala Glu Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Ala Glu Tyr Ala Lys Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 Page 11 of 16 WO 00/05250 PCT/US99/16747 <210> 41 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 41 Ala Glu Lys Ala Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 42 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 42 Ala Tyr Lys Ala Glu Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 43 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 43 Ala Lys Tyr Ala Glu Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 44 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 44 Ala Pro Lys Tyr Val Lys Gln Asn Thr Leu Lys Leu Ala Thr Ala 1 5 10 Page 12 of 16 WO 00/05250 PCT/US99/16747 <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Ala Ala Ala Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 46 <211>.15 <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 46 Ala Lys Lys Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 47 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 47 Ala Glu Ala Tyr Ala Ala Ala Ala Ala Ala Lys Ala Ala Ala Ala 1 5 10 <210> 48 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 48 Ala Glu Lys Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 Page 13 of 16 WO 00/05250 PCT/US99/16747 <210> 49 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 49 Ala Lys Glu Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Ala Lys Lys Tyr Ala Glu Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 51 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 51 Ala Glu Ala Tyr Lys Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 52 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 52 Lys Glu Ala Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 Page 14 of 16 WO 00/05250 PCT/US99/16747 <210> 53 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 53 Ala Glu Glu Tyr Lys Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 54 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 54 Glu Lys Ala Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> Glu Lys Lys Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 <210> 56 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 56 Glu Ala Lys Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 Page 15 of 16 WO 00/05250 PCT/US99/16747 <210> 57 <211> <212> PRT <213> Artificial Sequence <220> <223> synthetic peptide <400> 57 Glu Lys Tyr Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala Ala 1 5 10 Page 16 of 16

Claims (49)

1. A terpolymer consisting essentially of tyrosine, alanine and lysine randomly polymerized into a polypeptide.
2. The terpolymer of Claim 1 which is substantially free of glutamic acid.
3. The terpolymer of Claim 1 wherein said tyrosine is present in a mole fraction of about 0.005 to about 0.250; said alanine is present in a mole fraction of about 0.3 to about 0.6; and lysine is present in a mole fraction of about 0.1 to about
4. The terpolymer of Claim 1 wherein said tyrosine is present in a mole fraction of about 0.10, said alanine is present in a mole fraction of about 0.54, and said lysine is present in a mole fraction of about 0.35. 15 5. A terpolymer consisting essentially of glutamic acid in a mole fraction of about 0.005 to about 0.300; tyrosine in a mole fraction of about 0.005 to about 0.250; and lysine in a mole fraction of about 0.3 to about 0.7. v*
6. The terpolymer of Claim 5 wherein said glutamic acid is present in a mole fraction of about 0.26, said tyrosine is present in a mole fraction of about 0.16 and said lysine is present in a mole fraction of about 0.58.
7. A terpolymer consisting essentially of glutamic acid in a mole fraction of about 0.005 to about 0.300; alanine in a mole fraction of about 0.005 to about 0.600; and lysine in a S* mole fraction of about 0.2 to about 0.7. o P:\OPER\P\k 2.186990l-174clails.doc-3 1/10/03
8. The terpolymer of Claim 7, wherein said glutamic acid is present in a mole fraction of about 0.15, said alanine is present in a mole fraction of about 0.48 and said lysine is present in a mole fraction of about 0.36.
9. A terpolymer consisting essentially of tyrosine, glutamic acid and alanine randomly polymerized into a polypeptide, wherein said tyrosine is present in a mole fraction of about 0.005 to about 0.250; said glutamic acid is present in a mole fraction of about 0.005 to about 0.300, and said alanine is present in a mole fraction of about 0.005 to about 0.800. The terpolymer of Claim 9, wherein said tyrosine is present in a mole fraction of about 0.14, said glutamic acid is present in a mole fraction of about 0.21, and said alanine is present in a mole fraction of about 0.65.
11. The terpolymer of Claim 9 or 10, which is substantially .free of lysine.
12. A pharmaceutical composition for the treatment of an autoimmune disease, comprising a therapeutically effective amount of a terpolymer according to any one of claims 1-11 and a pharmaceutically acceptable carrier.
13. The pharmaceutical composition of Claim 12, wherein said terpolymer has a molecular weight of about 2,000 to about 40,000 daltons. 25 14. The pharmaceutical composition of Claim 13, wherein said terpolymer has a molecular weight of about 4,000 to about 9,000 daltons. P:\OPER\Pxk\236990-174claimsii.dc.3 1/100113 -61 The pharmaceutical composition of Claim 12, wherein said autoimmune disease is a B cell mediated autoimmune disease.
16. The pharmaceutical composition of Claim 12, wherein said autoimmune disease is a T cell mediated autoimmune disease.
17. The pharmaceutical composition of Claim 12, wherein said autoimmune disease is an arthritic condition.
18. The pharmaceutical composition of Claim 12, wherein said autoimmune disease is a demyelinating disease.
19. The pharmaceutical composition of Claim 12, wherein said autoimmune disease is an inflammatory disease. The pharmaceutical composition of Claim 12, wherein said autoimmune disease is multiple sclerosis, autoimmune S 15 hemolytic anemia, autoimmune oophoritis, autoimmune thyroiditis, autoimmune uveoretinitis, chronic immune thrombocytopenic purpura, colitis, contact sensitivity disease, diabetes mellitus, Graves disease, Guillain-Barre's syndrome, Hashimoto's disease, idiopathic myxedema, myasthenia gravis, psoriasis, pemphigus vulgaris, rheumatoid arthritis, or systemic lupus erythematosus.
21. The use of a therapeutically effective amount of the terpolymer according to any one of claims 1-11 for the manufacture of a medicament for the treatment of an 25 autoimmune disease in a mammal.
22. The use of a therapeutically effective amount of a polypeptide consisting essentially of amino acids tyrosine, P:\OPER\Pxk\23g6990-174claims.doc-31/10/03 -62- glutamic acid, alanine and lysine for the preparation of a pharmaceutical composition for the treatment of an autoimmune disease, wherein said autoimmune disease is not multiple sclerosis.
23. The use of Claim 21 or 22, wherein said terpolymer or polypeptide inhibits activation of T cells.
24. The use of Claim 21 or 22, wherein said terpolymer or polypeptide activates T cell suppressor mechanisms. The use of Claim 21, wherein said terpolymer or polypeptide inhibits activation of T cells responsive to myelin basic protein.
26. The use of Claim 21 or 22, wherein said terpolymer or polypeptide inhibits activation of T cells responsive to collagen type II peptide. 15 27. The use of Claim 21 or 22, wherein said terpolymer or polypeptide binds to a class II MHC protein.
28.. The use of Claim 21 or 22, wherein said terpolymer or polypeptide binds to HLA-DR1.
29. The use of Claim 21 or 22, wherein said terpolymer or polypeptide binds to HLA-DR2.
30. The use of Claim 21 or 22, wherein said terpolymer or polypeptide binds to HLA-DR4.
31. The use of Claim 21 or 22, wherein said terpolymer or polypeptide binds to an antigen presenting cell. S 25 32. The use of Claim 21, wherein said autoimmune disease is multiple sclerosis. P:\OPER\P.sk\233.'90'-174cl;inis.doc-31/I1/03 -63-
33. The use of Claim 21 or 22, wherein said autoimmune disease is autoimmune hemolytic anemia, autoimmune oophoritis, autoimmune thyroiditis, autoimmune uveoretinitis, chronic imm-une thrombocytopenic purpura, colitis, contact sensitivity disease, diabetes mellitus, Graves disease, Guillain-Barre's syndrome, Hashimoto's disease, idiopathic myxedema, myasthenia gravis, psoriasis, pemphigus vulgaris, rheumatoid arthritis, or systemic lupus erythematosus.
34. The use of Claim 21 or 22, wherein said terpolymer or polypeptide has a molecular weight of about 2,000 to about 40,000 daltons. The use of Claim 34, wherein said terpolymer or polypeptide has a molecular weight of from about 4,000 to about 12,000 daltons.
36. The use of claim 22, wherein the polypeptide is glatiramer acetate.
37. The use according to Claim 21, wherein the terpolymer o I contains lysine, alanine, and tyrosine polymerized in a molar ratio of at least 3 moles of lysine per mole of tyrosine, and at least 4 moles of alanine per mole of tyrosine.
38. The use according to Claim 37, wherein the molar ratio of lysine alanine tyrosine is 4.0 5.0 o 25 39. The use according to Claim 21, wherein the terpolymer contains the amino acids glutamic acid :lysine alanine polymerized in a molar ratio of 1.5 4.0 P:\OPER\Pk\23 99-174clliims.doc-31/10/03 -64- The use according to Claim 21, wherein the terpolymer contains the amino acids glutamic acid alanine tyrosine polymerized in a molar ratio of 1.5 5.0
41. The use according to Claim 21, wherein the terpolymer contains the amino acids glutamic acid :lysine tyrosine polymerized in a molar ratio of 1.5 4.0
42. The use according to Claim 21 or 22, wherein the terpolymer or polypeptide inhibits binding of an antigenic peptide to an MHC class II protein.
43.' The use according to Claim 21 or 22, wherein further the terpolymer inhibits class II-specific T cell responses to an MHC class II protein-peptide complex.
44. The use according to Claim 42, wherein the antigenic peptide is associated with an autoimmune disease.
45. The use according to Claim 43, wherein the MHC class II protein is associated with an autoimmune disease.
46. The use according to Claim 21 or 22, wherein the medicament or pharmaceutical composition further comprises at least an additional therapeutic agent.
47. The use according to Claim 46, wherein the additional therapeutic agent is selected from the group consisting of an antibody, an enzyme inhibitor, an antibacterial agent an antiviral agent, a steroid, a nonsteroidal anti-inflammatory agent, an antimetabolite, a cytokine, and a soluble cytokine 25 receptor.
48. The use according to Claim 46, wherein said additional therapeutic agent is an inducer of synthesis of a cytokine P:\OPER\P.\k23X86990I-174cl.lims.doc-3 1/l((03 in a subject.
49. The use according to Claim 47, wherein said cytokine is selected from the group consisting of interferon-p, interleukin-4 and
50. The use according to Claim 47, wherein said enzyme inhibitor is selected from the group consisting of a protease inhibitor and a cyclooxygenase inhibitor.
51. The use according to Claim 12, wherein said pharmaceutically acceptable carrier is selected as suitable for administration to the subject by a route selected from the group consisting of intravenous, intramuscular, intraperitoneal, subcutaneous, oral, and transdermal administration.
52. The use according to Claim 21 or 22, wherein said autoimmune disease is a B cell mediated autoimmune disease.
53. The use according to Claim 21 or 22, wherein said autoimmune disease is a T cell mediated autoimmune disease.
54. The use according to claim 21 or 22 wherein the autoimmune disease is an arthritic condition.
55. The use according to Claim 21, wherein said autoimmune disease is a demyelinating disease.
56.. The use according to Claim 21 or 22, wherein said autoimmune disease is an inflammatory disease. S. 57. The use according to any one of Claims 21-56, wherein *therapeutically effective amount is at least 5 mg per day. 25 therapeutically effective amount is at least 5 mg per day. g P:\OPER\Pxk\236199)I- 174claims.doc-3 1/11003 -66-
58. The use according to Claim 57, wherein the therapeutically effective amount is at least 10 mg per day.
59. The use according to Claim 58, wherein the therapeutically effective amount is at least 15 mg per day.
60. The use according to Claim 59, wherein the therapeutically effective amount is at least 20 mg per day.
61. The use according to any one of Claims 21-56, wherein the therapeutically effective amount is in the range of to 400 pg per kg of a subject per day. DATED this 31 st day of October 2003 Yeda Research and Development Co. Ltd. AND President and Fellows of Harvard College o• by Davies Collison Cave Patent Attorneys for the Applicants
AU52262/99A 1998-07-23 1999-07-23 Treatment of autoimmune conditions with copolymer 1 and related copolymers and peptides Expired AU768782B2 (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US9385998P 1998-07-23 1998-07-23
US60/093859 1998-07-23
US10182598P 1998-09-25 1998-09-25
US60/101825 1998-09-25
US10296098P 1998-10-02 1998-10-02
US60/102960 1998-10-02
US10818498P 1998-11-12 1998-11-12
US60/108184 1998-11-12
PCT/US1999/016747 WO2000005250A1 (en) 1998-07-23 1999-07-23 Treatment of autoimmune conditions with copolymer 1 and related copolymers and peptides

Publications (2)

Publication Number Publication Date
AU5226299A AU5226299A (en) 2000-02-14
AU768782B2 true AU768782B2 (en) 2004-01-08

Family

ID=30449548

Family Applications (1)

Application Number Title Priority Date Filing Date
AU52262/99A Expired AU768782B2 (en) 1998-07-23 1999-07-23 Treatment of autoimmune conditions with copolymer 1 and related copolymers and peptides

Country Status (1)

Country Link
AU (1) AU768782B2 (en)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DEEB ET AL (1992) J. IMMUN METHODS 152(1): 105-113 *
EUR.J.IMMUNLOL (1994) 24 (10): 2497-2505 *
ZISMAN ET AL (1991) PNAS 88(21): 9738-9742 *

Also Published As

Publication number Publication date
AU5226299A (en) 2000-02-14

Similar Documents

Publication Publication Date Title
CA2337688C (en) Treatment of autoimmune conditions with copolymer 1 and related copolymers
US7425332B2 (en) Treatment of autoimmune conditions with Copolymer 1 and related Copolymers
EP2327712B1 (en) Treatment of Crohn&#39;s disease with copolymer 1 and polypeptides
US6800287B2 (en) Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US6514938B1 (en) Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
JP4903886B2 (en) Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic applications
US7585843B2 (en) Treatment of demyelinating autoimmune disease with modified ordered peptides
JP4800481B2 (en) Treatment of autoimmune diseases with Copolymer 1 and related copolymers and peptides
AU768782B2 (en) Treatment of autoimmune conditions with copolymer 1 and related copolymers and peptides

Legal Events

Date Code Title Description
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 20010817

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired