MXPA98005648A - Synthetic il-10 analogues - Google Patents

Synthetic il-10 analogues

Info

Publication number
MXPA98005648A
MXPA98005648A MXPA/A/1998/005648A MX9805648A MXPA98005648A MX PA98005648 A MXPA98005648 A MX PA98005648A MX 9805648 A MX9805648 A MX 9805648A MX PA98005648 A MXPA98005648 A MX PA98005648A
Authority
MX
Mexico
Prior art keywords
human
production
peptide
amino acid
substance
Prior art date
Application number
MXPA/A/1998/005648A
Other languages
Spanish (es)
Inventor
Gronhoj Larsen Christian
Gesser Borbala
Original Assignee
Steeno Research Group A/S
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 Steeno Research Group A/S filed Critical Steeno Research Group A/S
Publication of MXPA98005648A publication Critical patent/MXPA98005648A/en

Links

Abstract

La invención se refiere al uso de una sustancia o polipéptído de acuerdo con la fórmula X1-X2-X3-Thr-X4-5 Lys-X5-Arg-X;(SEQ ID NO:22), en donde X, es Ala o Gly, X2 es Tyr o Phe, X3, X4 y X5 se seleccionan independientemente a partir del grupo que consiste de Met,Il-2, Leu y Val;y X6 se selecciona a partir del grupo que consiste de Asn, Asp, Gln y Glu, opcionalmente al menos uno de X1, X2, X3, X4, X5 y X6 esta independiente sustituido son un aminoácido no natural o inusual y/o el péptido esta ciclizado y/o el péptído esta estabilizado y/o el residuo de aminoácido amino-terminal esta acilado y/o el residuo de aminoácido carboxi-terminal esta amidado y los pectidomiméticos modelados en base a la fórmula anterior para la preparación de una composición farmacéutica para la reducción de la producción de TNF-alfa y/o para la profilaxis o tratamiento de la pancreatitis y/o para la profilaxis o tratamiento de infecciones virales tal como el síndrome de inmunodeficiencia adquirida (SIDA) o infección cutánea por HPV. En particular, la invención se refiere análogos de péptidos de la fórmula anterior, en donde al menos uno de XI, X2, X3, X4, X5 y X6 esta independientemente sustituido con un aminoácido no natural 0 inusual y/o el péptido esta ciclizado y/o el péptido esta estabilizado y/o un residuo de aminoácido amino terminal esta acilado y/o el, residuo de aminoácido carboxi terminal esta amídado, y los peptidomiméticos modelados en base a la fórmula anterior.

Description

SYNTHETIC ANALOGUES OF INTERLEUCINE 10 FIELD OF THE INVENTION The present invention relates to the pharmaceutical use of substances that are agonists of interieucin 10 (IL-10). In particular, the invention relates to the use of a substance of the invention for the manufacture of a pharmaceutical composition for the reduction of the production of TNF-a and / or the prophylaxis or treatment of pancreatitis, urinary arthritis (gout), allergy of the skin, allergic reactions on the skin, tissue damage as a result of hypoxia / ischemia, (infarction, reperfusion), inflammatory reactions due to virus infections, and / or for the manufacture of a conceptual agent.
BACKGROUND OF THE INVENTION They have been described, for example, in WO 93/02693 and WO 94/04180, pharmaceutical compositions comprising IL-10 or vIL-10 and the use of hIL-10 or vIL-10 for the manufacture of a pharmaceutical composition REF. 27816 for the treatment of various conditions, and certain IL-10 agonists have been described in WO 96/01318.
BRIEF DESCRIPTION OF THE INVENTION The present invention relates to the use of a substance or polypeptide according to the formula Xi-Xz-Xs-Thr-Xi-Lys-Xs-Arg-Xg (SEQ ID NO: 22), where X2 is Tyr or Phe, X3, X4 and Xs are independently selected from the group consisting of Met, Lie, Leu, and Val; and X6 is selected from the group consisting of Asn, Asp, Gln and Glu, optionally at least one of Xi, X2, X3, X4, X5 and Xe are independently substituted with non-natural or unusual amino acids and / or peptide is cyclized and / or the peptide is stabilized and / or the amino-terminal amino acid residue is acylated and / or the carboxy-terminal amino acid residue is amidated, and the moderate peptidomimetics are based on the above formula for the preparation of a pharmaceutical composition for the reduction of the production of TNF-a and / or for the prophylaxis or treatment of pancreatitis. Additionally, the invention relates to a substance or polypeptide having the formula: X1-X2-X3-Thr-X4-Lys-Xs-Arg-X6 (SEQ ID NO: 22) wherein: X2 is Tyr or Phe, X3, X4 and X5 are independently selected from the group consisting of Met, Lie, Leu, and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu, wherein at least one of Xi, X2, X3, XA, XS and Xs is independently substituted with unnatural or unusual amino acids and / or the peptide is cyclized and / or the peptide is stabilized and / or the amino acid residue at ino-terminal is acylated and / or amino acid residue carboxy terminal is amidated, and moderate peptidomimetics based on the above formula, the substance or polypeptide having at least one of the following properties: a) induces the inhibition of the spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by peripheral blood mononuclear cells , human (PBMC), c) induces production of the protein receptor antagonist interleukin-l (IRAP) by human monocytes, d) induces chemotactic migration of T lymphocytes, CD8 +, in vitro, e) desensitizes T cells CD8 +, human dand or result in insensitivity towards rhIL-10, f) suppresses the chemotactic response of T, human, CD4 + lymphocytes towards IL-8, g) suppresses the chemotactic response of human monocytes towards MCAF / MCP-1 h) inhibits expression of MHC class II molecules in human monocytes stimulated with IFN- ?, i) induces the production of IL-4 by human, normal, cultured CD4 + T cells, j) reduces the production of TNF-a in the reaction of mixed human leukocytes, k) downwardly regulates the production of TNF-a and IL-8 in a rabbit model of acute pancreatitis induced by bile acid and reduced neutrophil infiltration in the lungs of treated rabbits. It is contemplated (as described in detail in the following description of the immunological mechanisms) that the mechanism of action is via interference with the action of mediators of the immune system., in particular cytokines such as onocins, chemokine lymphokines and monocyte receptor antagonists, ie, that the substance of the active ingredient interferes with / suppresses the production of and / or the action of certain cytokines and thus inhibits the pathological processes that lead to tissue damage, and that the substance of the invention induces a production of antagonists of natural monocyte receptors, thus interfering / suppressing with the action of certain cytokines such as TNF-α or IL-1 and inhibiting this way the pathological processes that lead to tissue damage. An important embodiment of the present invention relates in this way to a pharmaceutical composition comprising, as the active ingredient, a substance of the invention. In a further aspect, the present invention relates to the use of a substance of the invention for the manufacture of a pharmaceutical composition for substantially inhibiting a biological effect related to a cytokine, i.e. the use of a substance of the invention as a protein. / IL-1 receptor antagonist peptide, lymphokine, monoclin, interieucin, interferon, chemokine or colony stimulation factor. Another aspect relates to the use of a substance of the invention for the manufacture or preparation of a pharmaceutical composition for the prophylaxis or treatment of a condition related to the disturbance of a cytokine system, ie, the receptor antagonist protein / peptide. from IL; lymphokine, monocin, interieucin, interferon, chemokine or colony stimulation factor system. In another aspect, the invention also relates to a method for treating a condition in a human related to a disturbance in a cytokine system, which method comprises administering to the subject an effective amount of a substance of the invention. The immune, cellular system takes part in the development of these disorders such as infectious, inflammatory and neoplastic diseases. Immunocompetent cells and their products can play important roles in the initiation, progression and possible chronic nature of the development of inflammatory conditions. These disorders are frequently without a known etiology and include common diseases such as diabetes mellitus, rheumatoid arthritis, inflammatory diseases of the gastrointestinal tract and the skin. Apart from these examples, cell-mediated immunity, or pro-inflammatory mediators, however, contribute to many other inflammatory and proliferative diseases (see Table 1).
TABLE I Some diseases where immune reactions mediated by macrophages / T lymphocytes are considered pathogenically important Skin diseases: Psoriasis "Atopic dermatitis Contact Dermatitis T-cell cutaneous lymphoma (CTCL) Sezari syndromes Pemphigus vulgaris Bullous penicillary Erythema nodosum Scleroderma Autoimmune diseases (including rheumatic): Uveitis Bechet disease Sarcoidosis Boeck Sjdgren syndrome Rheumatoid arthritis Juvenile arthritis Reiter syndrome Drop Osteoarthrosis Systemic lupus erythematosus Polymyositis Myocarditis Primary biliary cirrhosis Crohn's disease Ulcerative colitis Multiple sclerosis and other demyelinating diseases Aplastic anemia Purple Idiopathic thrombocytopenia Multiple Myeloma and B-Cell Lymphoma Simmons Panhipopituitarism Serious Diseases and Graves' Eye Disease Subacute Tireoditis and Hashimoto's Disease Addison's Disease Insulin-dependent diabetes mellitus (type 1) Other diseases Several clinical syndromes with vasculitis (for example, polyarteritis nodosa, Wegener's granulomatosis, Giant cell arteritis fever, malaise, Anorexia (for example, in acute and chronic inflammatory and infectious diseases) Disseminated intravascular coagulation (DIC) Arteriesclerosis (atherosclerosis) Shock (for example, in gram-negative sepsis) Cachexia (for example, in cancer, chronic inflammatory and chronic infectious diseases) Rejection to transplant and host disease against the graft Prevention of spontaneous absorption.
Activity, of IL-10 in the production of cytokines: HIL-10 inhibits the production of a number of cytokines including interferon-? (IFN-?), In factor-a Tumor Necrosis (TNF-a), stimulation factor of the Granulocyte Macrophage colony (GM-CSF), Granulocyte-CSF (G-CSF), IL-la, IL- lß, IL-2, IL-6, IL-8 and monocyte chemotactic polypeptide-1 (MCP-1 / MCAF) by monocytes / macrophages and / or T lymphocytes (4, 5). IL-10 also inhibits the ability of monocytes to migrate as a response to the chemokine MCP-1 / MCAF (75). In addition, hIL-10 induces antagonist production (IRAP) of the endogenous receptor, natural interleukin-1 (6), which inhibits IL-la and IL-1β by competing with receptor binding. Since IL-8 is strongly inducible by IL-la and by IL-lβ, IL-10 exerts part of its inhibitory effect on the production of IL-8 by stimulating the production of the IRAP antagonist of the IL-1 receptor. This latter mechanism is of considerable importance for the present invention as described and exemplified in the following. IRAP has anti-inflammatory activities (9), and its therapeutic effect in rheumatoid arthritis has been suggested (10). Also, the IRAP proved to be effective in the treatment of sepsis syndrome and was associated with a 28-day survival benefit dependent on the dose with the IRAP treatment (p = 0.015) in the study by Fisher et al. (11). IRAP exerts part of its anti-inflammatory effects by inhibiting the production of chemokines such as the production of IL-8.
The expression IL-10 and the antigen: IL-10 inhibits the expression of MHC class II in human monocytes (8). The constitutive expression IL-4 or induced by IFN-α of HLA-DR / DP and DQ was inhibited by hIL-10 (12). In addition, monocytes pre-incubated with IL-10 are intractable to the subsequent induction of MHC class II expression by IL-4 or IFN-α. IL-10 inhibits class II expression by human monocytes after activation by LPS (12, 76). BALB / c mice that were given 1 to 10 mg of IL-10 concomitant with a lethal dose of LPS were protected from death (6). IL-10 inhibits intermediate nitrogen compounds and superoxide anions. IL-10 also inhibits the reactive nitrogen intermediate compound (NO) as well as the reactive oxygen intermediate compounds (H202) by macrophages after activation by INF-? (13) Activity, of IL-10 and T cells: IL-10 also has modulatory effects on the functions / activity of T cells. Thus, hIL-10 is a potent chemotactic factor for CD8 + T lymphocytes, while hIL-10 does not show chemotaxis towards T cells CD4 + (14). Additionally, IL-10 suppresses the ability of CD4 + T cells to respond to the chemotactic signals of β-chemokine RANTES as well as a-chemokine and IL-8. HIL-10 also directly inhibits proliferation of T cells from the blood of peripheral, human, and CD4 + T cell clones (14).
Therapeutic considerations: These results / data in vivo and other data summarized for example in WO 96/01318 strongly suggest a homeostatic role of IL-10 in the control of immune inflammation amplified by monocytes and mediated by cells and indicate the wide range of therapeutic applications of IL-10 or a drug with activity similar to IL-10 in the treatment of diseases characterized by decreased / insufficient production and / or activity of IL-10. Tables 1 and 2 list some diseases where an immunomodular similar to IL-10 or an immunomodulator with activity similar to IL-10 is considered to have therapeutic importance.
TABLE 2 Some diseases where an immunomodulator with activity similar to IL-10, due to its induction of IRAP production and / or inhibition of cytokine production and / or cytokine activity may have therapeutic importance (Ref. 20-74 + 109 ) Pre-term labor caused by infection or other conditions Rheumatoid arthritis Lyme arthritis Gout Sepsis syndrome Hyperthermia Colitis or ulcerative enterocolitis Osteoporosis Cytomegalovirus Periodontal disease Glomerulonephritis Chronic, non-infectious inflammation of the lung (eg, sarcoidosis and smoker's disease) Granuloma formation Fibrosis of the liver Fibrosis of the lung Rejection of the transplant Host disease against the graft Chronic myeloid leukemia Acute myeloid leukemia Other neoplastic diseases Bronchial asthma Diabetes mellitus, type I (insulin-ddent) Arteriosclerosis / atherosclerosis Psoriasis Chronic B-cell leukemia Common variable immunodeficiency Lateral effects using other biological response modifiers Disseminated intravascular coagulation Systemic sclerosis Encephalomyelitis Inflammation of the lung Hyper IgE syndrome Enterocolitis Metastasis and cancer growth Adoptive immune therapy Acquired respiratory effort syndrome Sepsis Syndrome rfusion Postsurgical Inflammation Organ transplantation Alopecia AIDS Cutaneous HPV infection DETAILED DESCRIPTION OF THE INVENTION Development of a nonapeptide homologous to IL-10 with activity similar to IL-10 Partial sequences of hIL-10 having a length of 9 amino acids were chosen according to the principles that the sequence should proceed high homology between vIL-10 and hIL-10, but as low homology to mIL-10 as possible. It was found that a nonapeptide, IT9302, possessed some immunosuppressive activities that mimic those of hIL-10 as described in further detail in the following examples. IT9302 corresponds to a nonapeptide sequence from the C-terminal end of hIL-10 with the following amino acid sequence: NH2-Ala-Tyr-Met-Thr-Met-Lys-Ile-Arg-Asn-C00H (SEQ ID NO: 1) The nonapeptide IT9302 is very potent for inducing different functions and is very stable, and it is presumed that it can not be incorrectly attach receivers. A nonapeptide has been chosen because in general the polypeptide sequence of 9 amino acids is unique to a protein. However, it should be noted that the 6 amino acids at the very extreme of hIL-10 seem to be the most important. Within the scope of the present invention, a substance is thus the polypeptide comprising a sequence of the amino acid sequence Ala-Tyr-Met-Thr-Met-Lys-Ile-Arg-Asn (SEQ ID NO: '1) . It is likely that some amino acid substitutions will have no adverse effects on hIL-10 agonist activity as defined herein while threonine, lysine and arginine are present and with an amino acid placed between them.
The present invention relates in particular to the use of a substance or polypeptide according to the formula: X? -X2-X3-Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 22) wherein: Xi is Ala or Gly, X2 is Tyr or Phe, X3, X4 and Xs are independently selected from the group consisting of Met, Lie, Leu, and Val; and X6 is selected from the group consisting of Asn, Asp, Gln and Glu, optionally at least one of Xi, X2, X3, X4, X5 and Xs is independently "substituted with non-natural or unusual amino acids and / or the peptide is cyclized and / or the peptide is stabilized and / or the amino-terminal amino acid residue is acylated and / or the carboxy-terminal amino acid residue is amidated, and the moderate peptidomimetics are based on the above formula for the preparation of a Pharmaceutical composition for the reduction of TNF-α production induced by IL-1 presumably by blocking the IL-1 receptor with IRAP and / or for the prophylaxis or treatment of pancreatitis.
The specific polypeptide examples that are presumed to be useful for the reduction of TNF-a production and / or the prophylaxis or treatment of pancreatitis are as follows: 1. m ^ -Ala-Tyt-lfet-T r-Ilt-Lsrs-l i-Arg-? cn'COOH (SEQ ID NO: 2) 2. M2- a - ^ - ^ t-Thr-Read »Lys-Leu-Arg-Asn-COOH (SEQ ID H0: 3) 3. W ^ -Ala-r-Met-Thr-Met- and B-Val-Arg- ^ ii'COOH. { SEQ ID N0: 4) 4. KH ^ -Gly 'and r-Met-t r-Met-Lyele-Arg-Asp-COOH. { SEQ ID H0: 5) . K ^ -Ala-Phß-Mßt-O? R-Met-Lys-Ile-Arg-Asp-COOH (SEQ ID R0: 6) 6. HHj-Ala-Tyr-Ile-T r-Mat-Lys-Ile-Arg ^ Asp'OOOH (SEQ ID NO: 7) 7. l ^ -JÜ.a-Tyr-eu- hr-Mat-ys-Ile-Arg-Asp-CODH. { SEQ ID NO: 8) a. N% -Ala-tyr -Val -Thr-Mat- Lys-lié -Arg-Asp- COOH. { SEQ ID N0: 9) 9. HHj-Ala-Tyr-K t-Oir-Ile-Lys-Ile-Arg-Asp-COOH (SEQ ID NQ: 10) . 3H2-Ala-Tyr-Mst-T r 'eu-l.ya * lle-Arg-Aßp-COCa. { SEQ ID m-.11} 11. NH2-Ala-Tyr * Met-T r -Val-Lys-Ile? Rg * ABp-C00H (SEQ ID NO: 12) 12. NH2 * Ala-yr-Met-T r-Met-1, and B.Ile-Arg-Asp-COOH (SEQ ID NO: 13) 13. NH2-Ala-Tyr-3Sfet-hr-Met-Lye-lfót-A3rg-ABp-CX30H. { SEQ ID HO? L4) 14. NH2-Ala-Tyr-Ifet-Thr-tet-LyB-Val-Arg-A8p * C00H (SEQ ID NÜ.15J . NH2-Ala-Tyr «lfet-Thr-Ifet-tys-Ild-Arg-Gln-COOB (SEQ ID ND: 1S) 16. W ^ .- Ala-Tyr-Met-T r-Met-Lys-Ilß-Arg-Glu-COOH (SEQ ID NÜ: 17) The present invention relates in particular to a polypeptide having the formula: Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 19), a polypeptide having the formula: Xs-Thr-Xy-Lys-Xs-Arg-Xs (SEQ ID NO: 20) a polypeptide having the formula: X2-X3-Thr-X4-Lys-X5-Arg-Xs (SEQ ID NO: 21) and a polypeptide having the formula Xi-Xz-Xs-Thr-Xi-Lys-Xs-Arg-Xg (SEQ ID NO: 22; wherein: X2 is Tyr or Phe, X3 / X4 and Xs are independently selected from the group consisting of Met, Lie, Leu, and Val; and X6 is selected from the group consisting of Asn, Asp, Gln and Glu, wherein at least one of Xx, X2, X3, X4, X5 and Xs is independently substituted with unnatural or unusual amino acids and / or the The peptide is cyclized and / or the peptide is stabilized and / or the amino-terminal amino acid residue is acylated and / or the carboxy-terminal amino acid residue is aided, and the moderate peptidomimetics are based on the above formula, the analogs have at least one of the following properties: a) induces the inhibition of spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by peripheral blood mononuclear cells, human (PBMC), c) induces the production of the interleukin-1 receptor antagonist protein (IRAP) by human monocytes, d) induces chemotactic migration of lymphocytes T, CD8 +, in vitro, e) desensitizes human CD8 + T cells resulting in an insensitivity to rhIL-10, f) suppresses the chemotactic response of T lymphocytes, human, CD4 + to IL-8, g) suppresses the response Chemotherapy of human monocytes towards MCAF / MCP-1 h) inhibits the expression of MHC class II molecules in human monocytes stimulated with IFN- ?, i) induces the production of IL-4 by normal, human CD4 + T cells , cultured, j) reduces the production of TNF-a in the reaction of mixed leukocytes, humans, k) down-regulates the production of TNF-a and IL-8 in a rabbit model of acute pancreatitis induced by biliary acid and reduces the infiltration of peutrófilos in the lungs of the treated rabbits.
By use of the term "at least one biological activity of 19302" in the present specification and the claims, the reference is to mean that it is at least one of the properties mentioned above. Any of the contemplated peptides of the invention may have an amino-terminal amino acid residue that is acylated such as acetylated or benzoylated. Also, any of the contemplated peptides may have a carboxy-terminal amino acid residue that is amidated. The present invention further contemplates analogues of the peptides formed by other conservative amino acid substitutions than those specific substitutions proposed above, unusual or unnatural amino acid substitutions, peptide stabilization, peptide cyclization, and modeled peptidomimetides in IL-10 agonist peptides. identified. The principle behind conservative amino acid substitution is that certain pairs of amino acids have compatible side chains such that, when one is substituted by another, there will be only minimal changes in the tertiary structure and binding affinity of the peptide. The rules for conservative substitution are explained in (78). "Conservative" as used herein means (i) that the alterations are as structurally neutral as possible, i.e., they are designed to produce minimal changes in the tertiary structure of the mutant polypeptides compared to the native protein, and (ii) that the alterations are as antigenically neutral as possible, which are designed to produce minimal changes in the antigenic determinants of the mutant polypeptides compared to the native protein. Structural neutrality is desirable to preserve biological activity and antigenic neutrality is desirable to avoid activation of immunogenic responses in patients or in animals treated with the substance of the invention. Although it is difficult to select with absolute certainty which alternatives will be structurally and antigenically neutral, there are rules that can guide those skilled in the art to make alterations that are highly likely to be structurally and antigenically neutral, for example (77) and (78) . Some of the most important rules include (1) replacement of hydrophobic residues is less likely to produce changes in antigenicity because they are likely to be located inside the protein, for example, Berzofsky (cited above) and Bowie et al. (cited above); (2) replacement of physicochemically similar residues, that is, synonyms, is less likely to produce structural changes because the replication amino acid can play the same structural role as the replacement amino acid; and (3) the alteration of the evolutionary conserved sequences will probably lead to detrimental structural effects because the evolutionary conservation suggests that the sequences may be functionally important. In addition, these basic rules for selecting mutein sequences, assays are available to confirm the biological activity and structure of molecules engineered by genetic engineering. Changes in the structure can be tested by at least two well-known assays: the method of microcomplement fixation, for example (79) and (80) widely used in evolutionary studies of tertiary structures of proteins; and affinities to sets of monoclonal antibodies specific to the structure, for example (81). Biological assays for the substances of the invention are not fully described in the examples: The inhibition of the spontaneous production of IL-8 by human monocytes is tested, as summarized in Example 1, using the synthesized substance or peptide instead of IT9302. If the production of IL-8 is suppressed to be no more than 50% when 1 ng / ml is used in the substance or peptide, then the substance or peptide is within the scope of the present invention. The inhibition of IL-8 production induced by IL-1β is tested by peripheral blood human mononuclear cells (PBMC) as summarized in Example 2, using the synthesized substance or peptide in place of IT9302. If the percent inhibition of IL-8 production is at least 50% when 1 ng / ml of the substance or peptide is used, then the substance or peptide is within the scope of the present invention. The production of the antagonistic protein of the interleukin-1 receptor (IRAP) by human monocytes is tested, as summarized in Example 3 using the synthesized substance or peptide in place of IT9302. If the IRAP induction is at least 30 ng / ml when 10 ng / ml of the substance or peptide is used, then the substance or peptide is within the scope of the present invention. The induction of chemotactic migration of the human, CD8 +, T lymphocytes is tested in vitro, as summarized in Example 4, using the substance or peptide in place of IT932. If the potency of the substance or peptide when a concentration of 10 ng / ml is used, is present, ie, 2 or more, then the substance or peptide is within the scope of the present invention. The desensitization of human CD8 + T cells which result in an insensitivity to rhIL-10 is tested, as summarized in Example 5, using the synthesized substance or peptide in place of IT9302. If the preincubation of the cells with the substance or peptide results in a sensitivity totally suppressed substantially from the CD8 + cells towards hrIL-10, ie, which gives a value of about 1, such as from 0.8 to 1.2, to a concentration of the substance or peptide of 10 ng / ml, then the substance or peptide is within the scope of the present invention. The suppression of the chemotactic response of human, CD4 + T lymphocytes to IL-8 is tested as summarized in Example 6, using the synthesized substance or peptide in place of IT9302. If the addition of the substance or peptide to a suspension of human CD4 + T lymphocytes results in a substantially total inhibition of the response of the CD4 + cell to IL-8, ie, that gives a value of about 1, such as 0.8 at 1.2, a concentration of the substance or peptide of 10 ng / ml, then the substance or peptide is within the scope of the present invention. The suppression of the chemotactic response of human monocytes to MCAP / MCP-1 is tested as summarized in Example 7 using the synthesized substance or peptide in place of IT9302. If the addition of the substance or peptide to a suspension of human monocytes results in a substantially total inhibition of the monocyte chemotactic response towards MCAF / MCP-1, ie, which gives a value of about 1, such as 0.8 to 1.2, at a concentration of the substance or peptide of 10 ng / ml, then the substance or peptide is within the scope of the present invention. Inhibition of the expression of MHC class II molecules in human monocytes stimulated with IFN-α it is tested as summarized in Example 8 using the synthesized substance or peptide in place of IT9302. The INF-? it upregulates the expression of the MHC II antigen in the cell population from 36.8% to 58.4%, and this stimulation was blocked or down-regulated to 25.2% by 10 ng / ml of rhIL-10 and 31.2% by 1 ng / ml of IT9302 ( Figure 12). If a substance or peptide blocks or downregulates the expression of MHC class II and monocytes at the unstimulated level, when added in an amount of 1-10 ng / ml, then the substance or peptide is within the scope of the present invention. . If the addition of the substance or peptide is able to block the stimulation effect of IFN-? at a concentration of the substance or peptide of 10 ng / ml, then the substance or peptide is within the scope of the present invention.
The induction of IL-4 production is tested by human, normal, cultured CD4 + T cells as summarized in Example 9, using the synthesized substance or peptide in place of IT9302. If the addition of substance or peptide induces the production of IL-4 in CD4 + T lymphocytes at a concentration of the substance or peptide of 10 ng / ml, then the substance or peptide is within the scope of the present invention. The reduction of the production of TNF-a in the reaction of mixed leukocytes, human, is tested as summarized in Example 10 using the synthesized substance or peptide in place of IT93-02. If the addition of the substance or peptide significantly reduces the production of TNF-a in the reaction of mixed human leukocytes in the space of 24 hours at a concentration of the substance or peptide 10 ng / ml, then the substance or peptide is within the scope of the present invention. The down-regulation of TNF-a and the production of IL-8 in a rabbit model of acute biliary acid-induced pancreatitis and reduction of neutrophil infiltration in the lungs of treated rabbits is tested as summarized in Example 14 using the synthesized substance or peptide in place of IT9302. If the addition of the substance or peptide significantly reduces the mortality of the test animals, when the substance or peptide is added at a concentration of 100 μg / kg, then the substance or peptide is within the scope of the present invention. An important embodiment of the present invention thus relates to a polypeptide in which at least one amino acid residue has been substituted with a different amino acid residue and / or in which at least one amino acid residue has been deleted or added. to result in a polypeptide comprising an amino acid sequence that is different from the amino acid sequence or an amino acid sequence sequence as defined in the following, but having essentially hIL-10 agonist activity as defined above. Synthetic peptide analogs can also be made by substituting individual residues with unnatural or unusual amino acids. The sequences of the bioactive peptides are originally derived from proteins that are constituted by the residues of the twenty naturally occurring L-amino acids. However, a chemical synthesis process used to construct the synthetic peptides allows the substitution of alternative residues including D-amino acids, β-amino acids, N-substituted amino acids, naturally occurring amino acids infrequently, or amino acid analogs synthetic, not natural (93). Non-limiting examples of the amino acids useful in the present invention are: Aad 2-aminoadiphatic acid bAad 3-aminoadiphatic acid bAla Beta-alanine, beta-aminopropionic acid Abu-2-aminobutyric acid 4Abu 4-aminobutyric acid, piperidinic acid Acp 6- aminocaproic acid Ahe 2-aminoheptanoic acid Aib 2-aminoisobutyric acid bAib 3-aminoisobutyric acid Ap 2-aminopimelic acid Dbu 2,4-diaminobutyric acid Des Desmosin Dp 2,2 '-diaminopimelic acid Dpr 2,3-diaminopropionic acid EtGly N-ethylglycine EtAsn N-ethylapagina Hyl Hydroxylysine aHyl Alo-hydroxylysine 3Hyp 3-hydroxyproline 4Hyp 4-hydroxyproline Ide Isodesmosine alie Alo-isoleucine MeGly N-methylglycine, sarcosine Melle N-methylisoleucine MeLys 6-N-methyllisine MeVal N-methyllisine N a Norvaline Nle Norleucine Orn Ornithine Additional non-limiting examples of unnatural amino acids occurring infrequently or building blocks are listed as follows: Novabiochera 1994/95 Catalog (Calbiochem-Novabiochem AG, Weidenmattweg 4, CH-4448 Laufelfingen / S itzerland), pp . 65-125; Bachem Feinkemikalien AG 1995 Catalog (Bachem Feinkemikalien AG, Hauptstrasse 144, CH-4416 Bubendoft / S itzerland), p. 753-831; Neosystem Laboratoire Catalog 1997/98 (Neosystem Laboratoire, 7 rue de Boulogne, 67100 Strasbourg, France), p. 131-176. The alternative residues, described above can be used (a) to replace chemically reactive residues and improve the stability of the synthetic peptide towards, for example, enzymatic and proteolytic degradation, (b) to provide analytical labels useful in the detection of the synthetic peptide, and (c) to modulate the synthetic peptide bioactivity by increasing or decreasing the binding affinity of the peptide to the IL-10 receptor, for example by introducing structural constraints that reduce rotational freedom for specific chemical bonds. Within the scope of the present invention are additional substances wherein one or more residues Thr, Lys or Arg in the above formula are substituted with unnatural or unusual amino acids as proposed above. All polypeptide sequences in the following specification and claims are described, also when not stated explicitly, from the N-ter end to the C-terminal end in the conventional format.
The method of choice for the synthesis of peptides and their analogs is the synthesis of solid phase peptides (SPPS). This method was introduced by Marrifield (100) in 1963, and numerous peptides have been synthesized since then with this technique. An excellent review of the current chemical synthesis of peptides and proteins is given by S.B.H. Kent (101). In practice, peptides are mounted by gradual SPPS, the C-terminal amino acid in the form of a reactive N-alpha-protected derivative, if necessary, protected in the side chain, is covalently coupled either directly or by means of a suitable linker to a solid support, for example, a polymeric resin, which is swelled in an organic solvent. The N-alpha-protecting group is then removed and the subsequent protected amino acids according to the desired sequence are joined in a gradual manner. After assembly of the entire peptide chain, the side chain protecting groups are removed, and the peptide is cleaved from the resin, which can be done simultaneously or in separate steps.
Among the various different coupling strategies that have emerged over the years, two are generally in common use, based on the different N-alpha-protecting groups and the equalization of the side chain protecting groups. Merrifield used tert-butyloxycarbonyl (Boc) as the N-alpha-protecting group, while 9-fluoromethylmethoxycarbonyl (Fmoc) was introduced by Carpino and Han (102). The practical application of these two strategies including the reaction of solid supports, side chain protective groups, activation procedures, cleavage procedures, instrumentation, and analytical and monitoring techniques have been given in several articles among which they should mention the following: Stewart and Young (103), Atherton and Sheppard (104), and Pennington and Dunn (105). Peptides and their analogues with unusual or unnatural amino acids in the present invention are conveniently synthesized according to these protocols. Analogs of linear, synthetic peptides can be made by chemically converting structures to cyclic forms. Cyclization of linear peptides can modulate bioactivity by increasing or decreasing the binding affinity of the peptide for the objective protein (94). Linear peptides are very flexible and tend to adopt many different structures in solution. The current cyclization to restrict the number of structures available and in this way favor the most active or inactive structures of the peptide. The immunogenicity of the peptides has been correlated with structural preferences, observed experimentally in solution (95). The differences in immunogenicity may be indicative of differences in the binding affinity of the cyclic peptide-specific antibodies. Cyclization of linear peptides is achieved either by forming a peptide joined between the N-terminal and C-terminal (homodéticos cyclopeptides) or by forming a new covalent bond between the amino acid structure and / or the side chain groups (cyclopeptides heterodéticos) (93). These latter cyclizations use alternative chemical strategies to form covalent bonds, - for example, disulfides, lactones (both also present in natural peptides), ethers, or thioethers. Linear peptides of more than five residues can be cyclized relatively easily. The predisposition of the peptide to form a beta-turn structure in the four central residues facilitates the formation of both homo- and heterodétic cyclopeptides. The presence of proline or glycine residues at the N- or C-terminal ends also facilitate the formation of cyclopeptides, especially of linear peptides shorter than six residues in length. Examples of protocols for the formation of disulfide bonds and for other peptide cyclization reactions are given in Pennington and Dunn (105), chapter 7 and 11. The technology of peptidomimetics is the design of the molecular mimetics of the peptides. The ability to successfully design these molecules depends on the understanding of the properties of the peptide linear sequence and the structure in which it is presented to the IL-10 receptor. The synthesis of mimetics can provide compounds that exhibit higher biological activity, improved solubility, and stability (96). As an example, the following peptidomimetic has been derived based on the α-helical templates in the C-terminal peptide mimetics of cytokines described in US 5,446,128 (97) in combination with the knowledge that the C-terminal end of IL- 10 exists as an a-helix (98).
By this binding of a small molecule to the N-terminus of the peptide, an a-helical structure of the synthetic peptide is stabilized, and the peptide becomes more resistant to proteolytic degradation. Other peptidomimetics can be derived based on the description in US 5,446,128. These substances wherein the substitution has taken place in other residues different from Xi and / or where the substitution has taken place with other molecules than the N-terminal molecule shown in the above formula are within the scope of the present invention. According to the present invention, the term "a peptide analogue" comprises any pharmaceutically active and acceptable compound derived on the basis of the above formulas exhibiting at least one biological activity similar to IT9302, including derivatives of these analogs, especially pharmaceutically acceptable salts. , esters and solvates thereof. The following terms: "cytokine", "lymphokine", "interieucine", "monocyte", "chemokine", "interferon", "colony stimulation factor", and "polypeptides" are used as defined in WO 96/01318. One embodiment of interest of the invention relates to a polypeptide of the invention where the amino acids account for a total of 6, 7, 8, 9 or 10 up to about 100 amino acids, for example, 11, 12, 13, 14, or 15 amino acids, or even larger such as 20 amino acids or 30 amino acids. In a preferred embodiment of the invention, the substance or polypeptide is used in the substantially pure form. To obtain this, purification of the polypeptide may be required. Examples of the methods employed for the purification of the polypeptides are: (i) immunoprecipitation or affinity chromatography with antibodies, (ii) affinity chromatography with a suitable ligand, (iii) other chromatography methods such as gel filtration, exchange ion or high-performance liquid chromatography or derivatives of any of the foregoing, (iv) electrophoretic procedures similar to polyacrylamide gel electrophoresis, denaturing polyacrylamide gel electrophoresis, agarose gel electrophoresis and isoelectric focusing, (v) any other solubilization technique and / or specific purification. Also within the scope of the present invention is a pharmaceutical composition comprising a polypeptide substance of the invention and a pharmaceutically acceptable excipient. The composition may comprise, for example, synthesized, purified protein, or a recombinant, purified polypeptide. The IL-10 agonist used in this invention can be prepared as formulations in a pharmaceutically acceptable medium, for example, saline, phosphate buffered saline (PBS), Ringer's solution, dextrose / saline, Hank's solution, and glucose. The compositions may contain pharmaceutically acceptable excipients as required to approximate physiological conditions, such as buffering agents, tonicity adjusting agents, wetting agents, detergents and the like. The additives may also include additional active ingredients, for example, bactericidal agents, or stabilizers. The amount administered to the patient will vary depending on what is being treated, the purpose of the administration, such as prophylaxis or therapy, the state of the host, the manner of administration, and the like. Pharmaceutical compositions are typically proposed for transcutaneous or parenteral administration, for example, intravenously, subcutaneously, or intramuscularly. Orally administrable forms are also desired and can be provided by modifying the composition by deriving the stomach environment. The composition can be used for prophylactic and / or therapeutic treatment. The pharmaceutical compositions of the invention suitable for topical administration can be, for example, creams, ointments, lotions, ointments, gels, solutions, suspensions, pastes, bars, sprays or powders. The composition can be impregnated or distributed over eg pads, plasters or strips and is conveniently applied 1-10 times in a day. Topical compositions will generally comprise from 1-80% of the active compound by weight, based on the total weight of the preparations, such as 0.001-25% w / w of the active compound, for example, 0.1-10%, 0.5 -5%, or 2-5%. The composition can be formulated in accordance with conventional pharmaceutical practice with pharmaceutical excipients conventionally used for topical applications. Vehicles other than water that may be used for the compositions may include solids or liquids such as emollients, solvents, humectants, thickeners and powders. The pH of the composition can in principle be within a very broad range such as 3-9, although a pH of about 4 to 8 is preferred. Conventional buffering agents can be used to obtain the desired pH. As an example, a composition for transcutaneous administration may contain 1 mg of substance (IT9302) dissolved in 1 g of a cream base such as neutral cream of Moistion with 0.05% s_alicilic acid (the pharmacy of Árhus Kommunehospital) and applied in an amount of 0.4-0.5 mg under plastic in the skin. The composition is used in Examples 16 and 17. The pharmaceutical compositions may alternatively be administered intravenously.
In this manner, the invention provides compositions comprising an IL-10 agonist substance dissolved or dispersed in an acceptable carrier, preferably an aqueous carrier. These compositions can be sterilized by conventional sterilization techniques, or they can be sterile filtered. The resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation that is combined with a sterile aqueous carrier prior to administration. The IL-10 agonist can also be administered with a second biologically active agent, such as a chemotherapeutic agent, normal. These agents include, but are not limited to, vincristine, daunorubicin, L-asparaginase, mitoxantrone, and sacrum. In therapeutic applications, the pharmaceutical compositions are administered to a patient in an amount sufficient to produce the desired effect, defined as a "therapeutically effective dose". The therapeutically effective dose of an IL-10 agonist will vary according to, for example, the particular use for which the treatment is made, the manner of administration, the health and condition of the patient, and the judgment of the practitioner.
For example, the dose for continuous infection will typically be between 500 ng / kg / day and 50 μg / kg / day. This dose is calculated based on a randomized, controlled trial of human IL-10 (90). The concentration of the IL-10 agonist in the pharmaceutical formulations can vary widely, ie, from about 10 μg to about 5 mg / ml, preferably between about 100 μg and about 2 mg / ml. The concentration will usually be selected primarily by fluid volumes, viscosities, etc., according to the particular mode of the selected administration. In this manner, a typical pharmaceutical composition for intravenous infection could be made to contain 250 ml of dextrose / saline solution and 2.5 μg of the IL-10 agonist. For solid compositions, conventional non-toxic solid carriers can be used, including, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, solid saccharine, talc, cellulose, glucose, sucrose, magnesium carbonate, and Similar. For oral administration, a non-toxic, pharmaceutically acceptable composition is formed by incorporating excipients normally employed, such as those carriers listed previously and generally 10-95% active ingredient, ie, an IL-10 agonist substance, preferably of 25-75%. For aerosol administration, the IL-10 agonist is preferably delivered in finely divided form together with a surfactant and a propellant. Typical percentages of the IL-10 agonist are 0.01-20% by weight, preferably 1-10%. The surfactant must of course be non-toxic, and preferably soluble in the propellant. Representative of these agents are partial esters or esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic acid, octanoic acid, lauric acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, oleic acid and oleic acid with an aliphatic polyhydric alcohol or its cyclic anhydride such as, for example, ethylene glycol, glycerol, erythritol, arbitol, mannitol, sorbitol, the hexitol anhydrides derived from sorbitol, and the polyoxyethylene and polyoxypropylene derivatives of these esters. Mixed esters, such as mixed or natural glycerides may also be employed. The surfactant may constitute 0.1-20% by weight of the composition, preferably 0.25-5%. The rest of the composition is ordinarily the propellant. The liquefied propellants are typically gases at ambient conditions, and they condense under pressure. Among the suitable liquefied propellants are lower alkanes containing up to 5 carbon atoms, such as butane and propane; and preferably fluorinated and fluorochlorinated alkanes. Mixtures of the above can also be used. In the production of aerosol, a container equipped with a suitable valve is filled with the appropriate propellant, which contains the finely divided polypeptide (s) and the surfactant. In this way, the ingredients are maintained at an elevated temperature until released by the action of the valve. To improve serum half-life, the IL-10 agonist can be encapsulated, introduced into the liposome lumen, prepared as a colloid, or other conventional techniques that can be used to provide a prolonged lifetime of the polypeptides. Thus, in certain embodiments, the IL-10 agonist can be encapsulated in a liposome. A variety of methods are available for preparing liposomes, as described, for example, in (83), (84), (85) and (86).
As described above, it has been found that IT9302 and analogs or variants thereof are useful for preventing the effects of known cytokines that are pathogenically comprised in the pathological conditions previously described. Therefore, therapy potentials when using the polypeptide of the invention or analogs and derivatives thereof are contemplated and should be investigated in all diseases where the therapeutic effect of hIL-10 and / or IRAP is suspected (see above, Tables 1 and 2) .
DESCRIPTION OF THE FIGURES Figure 1 is a diagram showing that IT9302 inhibits the spontaneous production of IL-8 by cultured, purified human monocytes.
Figure 2 is a diagram showing that the IT9302 inhibits the production of IL-1 induced by IL-1 (1 ng / ml) by human peripheral blood mononuclear cells.
Figure 3 illustrates the production of IRAP by human monocytes stimulated by IT9302.
Figure 4 illustrates the production of IRAP by human monocytes stimulated with IL-10.
Figure 5 illustrates the chemotactic activity of IT9302 on CD8 + T cells.
Figure 6 illustrates desensitization of CD8 + T cells by IT9302, which results in insensitivity of CD8 + T cells to chemotaxis induced by IL-10 (10 ng / ml).
- Figure 7 illustrates the suppression of IL-8 activity in CD4 + T cells by IT9302.
Figure 8 is a diagram showing that IT9302 inhibits monocyte chemotaxis induced by MCAF / MCP-1.
Figure 9 shows the production of IL-4 in cells and cytosolic fractions of CD4 + T cells by Western ECL transfer.
Figure 10 shows the production of TNF-a as the cytosolic fractions of mixed lymphocyte cultures, human by Western blot ECL. The Western blot of TNF-α as and IL-4 described in materials and methods was carried out, but using a rabbit anti-human TNF-α antibody (Pepro Tech. Inc., London, England) and the secondary antibody labeled with horseradish peroxidase (Catalog No. P 217, Dako, Denmark).
Figure 11 shows the regulation of T cell proliferation by IL-10 and IT9302.
Figure 12 shows the expression of HLA-DR in human monocytes (flow cytometry).
Figure 13 shows that the shock induced by LPS and leukopenia are modulated by IT9302, shown by the total count of leukocytes.
Figure 14 shows that injection of murine IT9302 in rabbits before the produced pancreatitis prevents leukopenia.
EXAMPLES Materials and Methods atocinas y qu mioa trayentes Recombinant hIL-10 was obtained from Pepro Tech Inc., NJ. (Catalog No. 200 10). Recombinant hIL-lβ and recombinant hIL-8 were a class donated from Dainippon Pharmaceutical Company, Osaka, Japan. The culture medium was RPMI 1640 GIBCO, free of LPS according to the Amoebocy Liming Lysate test (Sigma E-TOXATE Kit Catalog No. 210-Al). rhMCAF / MCP-1 was a class bestowed by Professor Kouj i Matsushima, Kanazawa, Japan.
Leukocyte chemotaxis assay T cell chemotaxis Subsets of CD4 + and CD8 + T lymphocytes characterized by expressing antigens of either CD4 or CD8 were purified from the heparinized blood of normal donors. In this way, peripheral blood mononuclear cells were purified (PBMC) from the heparinized blood by diluting 100 ml of the blood with the Hanks balanced salt solution (HBSS) 1: 1 and then separating by stratification of the cells in Linfopac ^ (Nycomed Phar a, Oslo, Norway) followed by gradient centrifugation at 2000 rpm for 20 minutes. The mononuclear cells were washed 3 times in HBSS and the cell pellet was diluted in 4 ml of HBSS containing 1% fetal calf serum and separated at 4 ° C by using Dynabeads coated with monoclonal antibody to the CD4 or CD8 antigen ( Dynabeads M-450 CD4 Catalog No. 111.16, Dynabeads M-450 CD8 Catalog No. 111.08, DETACHaBEAD Catalog No. 125.904). The relationship counts: cell was 10: 1 and the incubation time was 1 hour. The beads were discarded by adding the polyclonal anti-mouse antibody according to the manufacturer's instructions. The chemotaxis assay was a 48-well microcamera technique (Neuroprobe, Rockville, MD) as previously described (74; see reference 3 and reference 14). The chemoattractants were diluted in RPMI 1640 (GIBCO Catalog No. 61870-010) with fetal calf serum, filtered, sterile 1% and placed in a lower chamber of 25 μl. In the case of terminating T-cell chemotaxis, T cells (5 x 10 ml) were dispersed in the medium and 50 μl was placed in the upper chamber separated from the lower chamber by a polyvinylpyrrolidone-free, polycarbonate-sized filter. 5 μm pore (Nucleopore Corp., Pleasanton, CA) coated with type IV collagen (Sigma Catalog No. C 0543). The cells were allowed to migrate for 2 hours at 37 ° C in 5% C02. The filters were then carefully removed, fixed in 70% methanol, and stained for 5 minutes in Coomassie brilliant blue. The cells attached to the lower surface of the filter were counted by measuring their area using a video camera in the microscope connected to a computer system for digital analysis and supported by programs for the objective determination of chemotactic migration. Approximately 5% of the T cells will migrate spontaneously, corresponding between 12,000 and 13,000 cells; This can vary from day to day, but very little in the same day experiments. As previously described (ref 13 and ref 14), it was therefore chosen to report the results as a relation between the number and cells that migrate in the sample and in the negative control, which reflects spontaneous migration. This relationship is referred to as the chemotactic index (Cl). All samples were analyzed in triplicate and cell migration in each well was measured in three fields before the average value of the area was estimated. In some experiments, the chemotaxis membrane was not coated with collagen, and in the present assay system the IgG cells will therefore fall into the bottom of the lower well of the chemotaxis chamber. In one experiment, the chemotactic activity of IT9302 on CD8 + T cells was performed by testing serial dilutions of IT9302 added to the lower chamber and evaluating chemotaxis as described above. In a second experiment, the ability of IT93Q2 to desensitize the migration of CD8 + T cells as a response to rHIL-10 (10 ng / ml) was studied by adding IT9302 to target cells 30 minutes before chemotaxis. IT9302 was added in serial concentrations and the chemotactic response of rhJL-10 was evaluated as described above. In a third experiment, the ability of IT9302 to suppress chemotactic responses of CD4 + T cells towards rhIL-8 (10 ng / ml) was studied by adding IT9302 to target cells minutes before chemotaxis was performed. IT9302 was added in serial concentrations and the chemotactic response of rhIL-8 was evaluated as described above.
Chemotaxis of Monocytes Monocyte chemotaxis was measured using the same Boyden camera equipment as described for the previous T cells. The MCAF / MCAF-1 chemoattractant was diluted in RPMI 1640 medium with 0.5% BSA and a concentration of 10 ng / ml was added to the lower chamber. Monocytes, purified by the normal plastic adhesion technique, from normal, human PBMCs obtained as described above were dispersed in RPMI 1640 medium with 0.5% BSA and then incubated for 30 minutes in the presence of IT9302 at different concentrations; the cells were added to the upper chambers of chemotaxis at a concentration of 10 cells / ml. The upper and lower chambers were separated by a polyvinylpyrrolidone-free polycarbonate filter with a pore size of 8 μm or (Nucleopore, Pleasanton, CA). The camera was incubated 37 ° C for 90 minutes. The membranes containing the migrating cells were tested as described above and a chemotactic index was calculated according to the technique described above.
Production of IL-8 by mononuclear, peripheral, human, normal cells (PBMC) PBMC were purified from the heparinized blood of normal human donors. After gradient centrifugation with Lymphoprep1 ^ (Nycomed Phar a, Oslo, Norway), the mononuclear cells were diluted to 2 × 10 6 cells / ml in the RPMI 1640 free LPS medium (Gibco Catalog No. 6187-010) contains heat inactivated fetal calf serum, filtered, sterile 1% and penicillin (10,000 IE / ml), streptomycin (10 mg / ml) and gentamicin (2.5 mg / ml). Cells were grown in Nunc Micro Plates 24-well micro plates (Nunc, Denmark) and in the presence of different IT9302 concentrations (0.1 μg, 100 ng, 10 ng, 1 ng, 0.1 ng, 0.01 ng / ml) for 24 hours. After 24 hours of incubation, another dose of IT9302 was added once more, and 1 hour later r-hLL-lβ (1 ng / nml) was added to the cell cultures. Supernatants were collected after a total of 48 hours of incubation, and the concentration of IL-8 secreted by IL-8 ELISA was measured using an IL-8 ELISA kit (Dainippon Pharmaceutical, Co. Ltd., Osaka, Japan) . Briefly, normal and cellular supernatants were incubated for one hour and in duplicate at 20 ° C on a microplate shaker. After washing, a second antibody was added for one hour, followed by one hour of incubation with goat anti-rabbit IgG labeled with peroxidase. After washing, the reaction was developed with 0-phenylenediamine. Thirty minutes later the reaction was stopped with 1.6 N sulfuric acid. The optical density (OD) was measured in an ELISA vector at 490 nm. The concentration of IL-8 was calculated by an absorbance calibration curve of known concentration against concentrations of IL-8 standards.
Determination of the concentration of IRAP PBMC were purified as described above. The PBMC were cultured in RPMI and 1640, heat inactivated fetal calf serum, sterile 10% (including penicillin 100.00 IE / ml, streptomycin 10 mg / ml, gentamicin 2.5 mg / ml) and the cell concentration was 5 x 10 cells / ml. The monocytes were then purified by the normal plastic adhesion technique. Then, the monocytes were cultured in RPMI 1640 with 2% FCS (2.5 x 10 cells / ml) with different dilutions of rhIL-10 or IT9302. The cells were stimulated for 24 hours and the supernatants were collected for the determination of IRAP. The IRAP ELISA was carried out using the Quantikin Human Immunoassay Kit from R &D Systems Europe Ltd. (Catalog No. DRA 00, Abingdon, Oxon, UK).
Determination of T cell proliferation Proliferation assay. PBMC (2 x 10) were cultured in 200 μl of RPMI with 10% FCS for 72 hours with 0.5 μl / ml PHA and rhIL-10 (1, 10, 100 ng / ml) or IT9302 (0.1, 1, 10 ng). / ml) in triplicate. The last 48 hours, 3H-thymidine was added with 0.5 μCi / well (Amersham, Denmark). The cells were harvested on a filter (Glass Microfibre Filters, Whatman, Catalog No. 1822 849) and scintillation fluid (Ultima, Gold MV, Packard) was added. The scintillation count was done on a Tri-Carb model 1600 TR, Packard.
Determination of 2nd expression of the MHC class II antigen in monocytes The expression of HLA-DR in human monocytes. Monocytes were isolated from heparinized, fresh blood by adhesion to plastic at 37 ° C in RPMI 1640 containing 10% FCS. After incubation, the supernatant was removed, Hank's solution was added (4 ° C with 1% FCS) and the cells were discarded on cooling at -20 ° C for 15 minutes and shaking gently on the table. Monocytes (2 x 10 6 cells / ml) were stimulated in RPMI 1640 supplemented with 2% FCS, with IFN-α. (10 ng / ml), or rhIL-10 (100, 10, 1 ng / ml) added before IFN-? or IT9302 (10, 1, 0.1 ng / ml) added before IFN-? for 40 hours. The cells were discarded from the wells when cooling as before. Fresh, non-fixed cells were used for surface typing by the anti-human antibody for HLA-DR. The cells were resuspended in Hank's solution with 1% FACS, 1 x 10 cells / ml and mouse anti-HLA DR, DP, DK antibodies, conjugated with FITC were added (F 0817, DAKI Denmark) for 45 minutes. The cells were washed three times in Hank solution and a FACS analysis was performed on a coulter-s XL-MCL flow cytometer at a wavelength of 488 nm. The non-specific binding was determined with a non-relevant Fitch conjugated antibody (DAKO F 479 anti-goat mouse). Alternatively, cells were fixed in % DMSO, 40% RPMI 1640 and 50% sterile FCS, and stored at -80 ° C and used for DNA typing.
Of ermination of apoptosis in monocytes DNA typing of stimulated monocytes. The fixed cells were incubated in 70% ethanol for 60 minutes and washed twice in Hank's solution. 1 x 106 cells were resuspended in 250 μl of 1 μg / ml RNase in 1.12% sodium citrate, pH 8.4 (ribonuclease A, Pharmacy No. 17-0442-01) and incubated at 37 ° C for 3 minutes. Subsequently, 250 μl of propiodiumiodide (50 μg / ml) was added in Hank's solution), and the cells were incubated in the dark for 30 minutes. After this, the cells were washed twice in Hank's solution (Jensen et al., (110)). The DNA content was measured by flow cytometry in a coulter-s XL-MCL at a wavelength of 550 nm.
Resolved Regulation of 2nd pro-proliferation of T cells by IL-10 and IT9302 PBMC were stimulated by PHA as described above and rhIL-10 and IT9302 were added for 72 hours, and incorporation of 3H-thymidine was measured during the last 18 hours. Both rhIL-10 and IT9302 down-regulated cell purification at an optimal concentration of 100 ng / ml of rhIL-10 and 10 ng / ml of IT9302, as seen in Figure 11.
Expression of HLA-DR in human monocytes The monocytes were purified as described above and stimulated for 40 hours by IFN-? (10 ng / ml) and / or rhIL-10 and / or IT9302 added 30 minutes before IFN- ?. The expression of the MHC II antigen was studied by incubation with. the human anti-HLA DR, DP, DK mouse antibody, conjugated to FITC. The expression of the MHC II antigen was upregulated by IFN-α. as seen in Figure 12, and 10 ng / ml of rhIL-10 and 1 ng / ml of IT9302 down-regulated the expression of the MHC II antigen in a similar manner.
DNA typing of stimulated monocytes Monocytes stimulated with IFN-? from the above, they were incubated with propidium iodide and the DNA content was measured by flow cytometry. The fraction of cells expressing DNA in the Gi or G2 phase of cell proliferation and also the fraction expressing apoptosis was measured. As seen in Figure 12, unstimulated monocytes that express 6.6% apoptosis, stimulation of IFN-? it down-regulated apoptosis to 4.1%, while the stimulation of both IL-10, IFN-? and IT9302 + IFN-? of the cells induced the apoptotic fraction at 10.3% and 9.3%, respectively.
Determination of IL-4 production by CD4 + T lymphocytes Cell cultures CD4 + T lymphocytes were purified from normal, heparinized human blood. After gradient centrifugation with Lymphoprep ™ (Nycomed Pharma, Oslo, Norway), the mononuclear cells were further separated 4 ° C using Dynabeads (Dynal AS, Norway) coated with monoclonal anti-CD4 antibodies. The beads were discarded when adding polyclonal anti-mouse antibody (Dinal, AS, Norway). The purity of the positively selected cells was higher than 99% as judged by the FACS analysis. When it is examined that the production of new IL-4 by T cells stimulated with IL-8 T cells were cultures, 5 x 106 cells / ml of an RPMI 1640 free of LPS (Gibco Catalog No. 61870-010) contains heat inactivated fetal calf serum, filtered, sterile 1% (FCS) penicillin (10,000 IU / l), streptomycin (10 mg / ml) and gentamicin (2.5 mg / ml). T cells were stimulated for 3 days using rIL-8 (100 ng / ml), rIL-10 (100 ng / ml), IT9302 (10 ng / ml) and IFN-? (10 ng / ml). Human, recombinant IL-8 (rh IL-8) was a class of Dainippon Pharmaceuticals, Co. Ltd., Osaka Japan), and the IFN-? was purchased from Boehringer Ingelheim Am Rhein, Germany. To obtain specific inhibition of IL-8 stimulation, a neutralizing monoclonal anti-IL-8 antibody (WS.4) was used, a class of Dr. K. Matsushima, Japan). Recombinant IL-10 was purchased from Pepro Tech,? Nc. (London England).
Preparation of cell material and culture supernatant for gel electrophoresis.
The cultured T cells and the culture medium were separated by centrifugation at 2000 rpm for 5 minutes. The supernatants were freeze-dried and dissolved in 100 μl of the lysis buffer. The cells were directly redispersed in 100 μl of the gel lysis buffer (9). The material was maintained at -80 ° C until further examination.
ECL-Western transfer of proteins derived from CD4 + T cells.
Cells or supernatants of the freeze-dried cell cultures were used for the determination of the protein content of IL-4. Proteins from 15% SDS-PAGE gels, one-dimensional, were transferred by transfer onto Hybond-ECL nitrocellulose membranes (Amersham RPN 2020D, UK) and blocked with 5% bovine serum albumin (Sigma) in saline pH 7.8. of Tris buffer containing 0.1% T een-20. The blots were then incubated with a polyclonal goat anti-human IL-4 antibody (R & D Systems, UK) followed by a secondary antibody labeled with horseradish peroxidase (Catalog No. RPN 2106 ECL, Amersham, UK), and immunostaining was detected by arranging a film (Kodak X-OMAT-S, USA) for 90 seconds.
EXAMPLE 1 Inhibition of the spontaneous production of IL-8 by human monocytes The test was performed as described in "Production of IL-8 by peripheral, human, normal mononuclear cells (PBMC)". The monocytes were purified by the plastic adhesion technique and 3.0 x 10 cells / ml were stimulated for 40 hours. As is known in Figure 1, IT9302 inhibited the production of IL-8 by monocytes, and at 0.1 ng / ml of IT9302, the production of IL-8 was suppressed to 35% of spontaneous production in vitro. The viability of the cells always exceeded 80% after 1 day in culture and the addition of IT9302 did not affect, in this or in other examples, the viability at any IT9302 concentration between 0.1 and 1000 ng / ml (IT9302 PM: 1,127 dalton, rhIL-10 predicted PM: 18, 400 dalton).
EXAMPLE 2 Inhibition of IL-8 production induced by IL-1β by human peripheral blood mononuclear cells (PBMC) The test was performed as described in "Production of IL-8 by human mononuclear cells, normal peripheral blood (PBMC)". As shown in Figure 2, IT9302, in a dose-dependent manner, inhibited the IL-8 production induced by IL-lβ by human peripheral blood mononuclear cells in vitro. The suppression of stabilized IL-8 production at IT9302 concentrations between 0.01 and 100 ng / ml.
EXAMPLE 3 Production of the interleukin-1 receptor antagonist protein (IRAP) by human monocytes The test was performed as described in "Determination of the concentration of IRAP". As shown in Figure 3, IT9302 induced the production of IRAP by human monocytes dependent on the dose. Production was increased basically when concentrations of IT9302 above 10 ng / ml were used. Figure 4 shows the induction of IRAP by rhIL-10 and since hIL-10 is approximately 20 times larger than IT9302, 5 ng / ml of IT9302 equal to 100 ng / ml of IL-10 in molarity. Therefore, at the powers of IT9302 and rhIL-10 they are comparable and approximately equal with respect to the induction of IRAP at lower concentrations. At concentrations of IT9302 exceeding 10 ng / ml, the induction of IRAP increased and reached a maximum level of 60 ng / ml. In addition, the specificity of this induction was tested by the antibody that specifically targets IT9302. In a separate experiment, where monocytes spontaneously produced 3.5 ng / ml of IRAP and were induced by 1-10 ng / ml of IT9302 at a maximum IRAP production of 10.6 + 0.6 ng / ml, this production could be blocked by an antibody polyclonal for IT9302, 2 μg / ml added 30 minutes before 10 ng / mg of IT9302, so that the level of IRAP was down-regulated to 2.9 + 0.3 ng / ml. These results in contrast to the result contained with the antibody and chosen against IL-10, 19F1, which was added in the same manner (2 μg / ml 30 minutes before 10 ng / ml of IT9302) did not block the production of IRAP but at otherwise they up-regulated this to 22 ng / ml. This result can be explained because this antibody is capable of neutralizing endogenous IL-10 but not IT9302. IL-10 produced spontaneously in cell culture has a negative autoregulatory effect on the production of IRAP by IT9302.
EXAMPLE 4 The chemotactic effect in CD8 + T lymphocytes, human The experiment was carried out as described in "Leukocyte chemotaxis assay". As shown in Figure 5, IT9302 induced the chemotactic migration of human T cells, CD8 + in vitro, whereas it did not affect CD4 + T cells (data not shown). Again, the power in IT9302 shown in this experiment is comparable to that of rhIL-10 shown previously (14).
EXAMPLE 5 Desensitization of human CD8 + T cells resulting in insensitivity to rhIL-10 The experiment was carried out as described in "leukocyte chemotaxis assay". IT9302 was added to a suspension of CD8 + T cells 30 minutes before these cells were tested for their chemotactic response to rhIL-10. As shown in Figure 6, pre-incubation of cells with IT9302 results in a suspended sensitivity of CD8 + T cells to hrIL-10. This indicates that IT9302 can affect the binding of rhIL-10 to the IL-10 receptor.
EXAMPLE 6 Suppression of the chemotactic response of CD4 + T lymphocytes to IL-8 The experiment was carried out as described in "leukocyte chemotaxis assay". As shown in Figure 7, IT9302, in a dose-dependent manner and added to a suspension of human CD4 + T lymphocytes, inhibited the response of CD4 + T cells to IL-8.
EXAMPLE 7 Suppression of the chemotactic response of human monocytes towards MCAF / CP1 The experiment was carried out as described in "monocyte chemotaxis". As shown in Figure 8, IT9302, in a dose dependent manner of adding a suspension of human monocytes, "inhibits the monocyte chemotactic response towards MCAF / MCPl.
EXAMPLE 8 Inhibition of the expression of MHC class II molecules of human monocytes when stimulated with IFN-α In a new experimental model (flow citrometry), it has been shown that monocytes stimulated with IFN-α upregulate their MHC II antigen expression, and this induction can be blocked by IL-10 and IT9302 in a similar manner. At the same time, both peptides up-regulate the number of cells showing apoptosis in monocytes (see Figure 12). These experiments indicate that IL-10 and IT9302 can inhibit T-cell proliferation dependent on monocytes / macrophages by decreasing the antigen exhibiting the capacity of monocytes via down-regulation with the expression of MHC class II (see 12). The PMBC stimulated by PHA began to proliferate as shown by the incorporation of 3H-thymidine, which can also be down-regulated by IL-10 and IT9302. PHA stimulates the proliferation of T cells through the activation of Ca2 + -dependent channels. Ca2 + fluxes were decreased in human T cell clones when cells were pre-incubated with IL-10 (Taga, K. et al., 1992). The present experiment shows that the proliferation of T cells is down-regulated by IT9302 also by the control of the Ca2 + -dependent channels.
Discussion related to the experiments.
The present data show a dose-dependent inhibitory effect of the synthetic nonapeptide, IT9302, in processes that reflect pro-inflammatory activity, including the production of IL-8 and the migration of monocytes and / or T cells. Thus, IT9302 was able to suppress the spontaneous production of IL-8 by human monocytes cultured overnight. This could be explained by a direct inhibitory effect on the production of IL-8 mRNA and / or the subsequent protein production and / or release. Another mechanism could be explained by the fact that monocytes cultured in vitro will express and produce IL-1, which then induces the production of IL-8. This is supported by the fact that IT9302 has been shown to potently induce IRAP production of monocytes. Therefore, IT9302 also inhibits the spontaneous production of IL-8 by interfering with the activity of IL-1. The induction of IRAP observed by IT9302 seems to induce a biologically active IRAP, since the IT9302 added to the cultures counteracts the production of IL-8 induced by IL-1, but only when it is added at least 16 hours before adding IL-1. 1 to the crops. This could mean that IT9302 inhibits the IL-1 production induced by IL-1 by inducing the production of IRAP, which then in turn blocks the activity of IL-1 through its receptor. If IT9302 directly inhibited the production of IL-8, it would have been expected that the addition of IT9302 to the cultures 1 hour before adding IL-1 should inhibit the production of IL-8, which was not the case (data not shown) . Therefore, the observed inhibition of IL-8 production of IT9302 will probably be due to an induction of IRAP production instead of a direct inhibition of IL-8 production. IT9302 specifically blocks the IL-1 receptor by the induction of IRAP, and thus the induction of other cytokines that are induced by IL-1, similar to TNF-a, IL-8 and probably many other cytokines and factors. The specificity of the induction was confirmed because the antibody to IT9302 could block the induction of IRAP. Another 19F1 antibody of IL-10 did not block the production of IRAP induced by IT9302. IT9302 also mimics the activity of IL-10 by suppressing the ability of CD4 + T cells to migrate as a response to IL-8. Since IL-8 is related to many different states of inflammation and since CD4 + cells appear early in the infiltration of T cell-mediated immune inflammation such as skin allergy, this characteristic may prove to have a significant therapeutic value for control of T cell-mediated immune inflammation. The chemotactic activity of CD8 + T cells, demonstrated by IT9302, is also parallel to that of IL-10, and IT9302 can thus activate T-cell populations with suppressive activity that contributes to the completion of T cell-mediated immune inflammation. Therefore, IT9302 according to the examples shown above, has therapeutic value in diseases where IL-10 and / or IRAP may also have therapeutic value. Additionally, IT9302 may have therapeutic value in diseases where IL-8 and / or MCAF and / or IL-1 are believed to have pathogenetic patents. The present invention describes analogues of IT9302, ie, substances or peptides having at least some of the properties described above.
EXAMPLE 9 Jnducsión of 2nd production of IL-4 in CD4 + T lymphocytes A tends Similar to IL-10, IL-4 is a product of CD4 + T cells of the TH2 type. It was observed that recombinant human IL-10 induces the production of IL-10 by cultured human CD4 + T cells. This means that IL-10, in addition to its own immunosuppressive functions, also induces the production of another immunosuppressive cytokine, IL-4. Therefore, it was tested whether IT9302 also induces the production of IL-4 by CD4 + T cells. In this way, CD4 + T cells, purified as described in "Methods for T cell chemotaxis", and cultured as described in the section "Determination of IL-4 production by CD4 + T lymphocytes", were stimulated for 3 days with IT9302 ( 10 ng / ml) or IL-10 (100 ng / ml). The cytosolic fractions were collected and analyzed for their IL-4 content using the Western blot (Figure 9) and a goat anti-human IL-4 polyclonal antibody. As shown in Figure 9, it was observed that 11-10 as well as IT9302 induce the production of IL-4 by human, normal, cultured CD4 + T cells.
EXAMPLE 10 Inhibition of TNF-α production during a mixed leukocyte reaction (MLR).
It has been shown that the reaction of mixed leukocytes is partially dependent on the production of TNF-a during the reaction. It has been shown that the IT9302 does not significantly reduce the MLR, but it was found that there is a significant reduction in the production of TNF-a during the MLR. In this way, MLR was performed by purifying human leukocytes and then culturing one million cells / ml of allogeneic donors for 4 days. Before establishing the cultures, a group of cells was irradiated for 2 minutes using beta irradiation. The cytosolic protein fractions were purified as described in the section "Determination of IL-4 production by CD4 + T lymphocytes", and Western blotting was performed using rabbit anti-human TNF-α antibody. As shown in Figure 10, a significant reduction in TNF-α production was observed during a reaction of mixed, human leukocytes.
EXAMPLE 11 The modulation of the shock induced by LPS and leucopenia in pigs.
Since it was found that IT9302 modulates the production of cytokines, including TNF-a and IL_8, and was supported by the published sequence of porcine IL-10 (88), it was tested whether IT9302 was able to modulate the course of the Leukopenia induced by LPS in pigs (Figure 13). In a preliminary experiment, it was tested how the intravenous injection of 0.1 mg / kg of IT9302 modulated the effect of the intravenous injection of 2 μg / kg of LPS in pigs (N = 3). IT9302 was injected 30 minutes before the LPS injection, and blood samples were drawn as described in Figure 13. The total leukocyte count as well as the cell differential gutter were determined, and the total number of neutrophilic leukocytes was calculated. based on these results. As shown, it was observed that the injection of LPS caused momentary leukopenia. However, the injection of IT9302 prevented leukopenia as shown in the Figure.
EXAMPLE 12 The effect of IT9302 murine homologue to human 119302 on the release of TNF-a in plasma after 2a administration of LPS in mice.
It has been shown in animals that murine, recombinant IL-10 can protect mice from lethal endotoxemia (89) and further, it has been shown that IL-10 administered in humans has inhibitory effects on T cells and suppresses reproduction of proinflammatory cytokines (90). The purpose of the study was to test whether IT9302 can suppress the production of TNF-a in mice exposed to endotoxin-induced shock after administration of LPS (Figure 14). BALB / C mice were used for eight weeks of age (obtained from the center for research animals, Bomholtgaard, Bomholtvej 10, DK-8680 Ry, Denmark). LPS from E. coli was sigma (lot 3444114) and the murine IT9302 was from Schafer-N, Lers0n Parkallé 42, DK-2100 Copenhagen 0, Denmark. The mice were injected intraperitoneally with 100 μg of LPS in 1 ml of 0.9 M NACÍ. 60 minutes before the injection of LPS, the mice were given IT9302 murine equivalent to 0.1 μg, 1 μg and 10 μg of IL- 10 human Groups of 5 animals (Imobilon, Pherovet, Malmo, Sweden) were anesthetized after 1, 2, 3 hours, respectively, after which maximum whole blood was removed through a cardiac perforation. The blood was centrifuged cold and the sera were collected and stored at -70 ° C for subsequent analysis of the TNF_a content by ELISA (TNF-a Mouse Immuno Assay Diagnostic kits, Genzyme, Cambrige, MA, USA). As shown, it was observed that 10 μg of murine IT9302 clearly suppressed TNF-a in the mouse sera compared to the total group not treated with mIT9302 (see Table 3).
TABLE 3 m-TNF-a ng / ml + SEM in murine IT9302 mouse serum equivalent to hIL-10 added 60 min before LPS 0 0.1 μg 1 μg 10 μg 1 h 2.48 ± 0.16 1.92 ± 0.48 2. .64 + 0.38 1.84 + 0.16 2 hs 1.43 + 0.17 1.68 + 0.27 1., 16 ± 0.21 0.62 ± 0.14 3 hs 0.48 + 0.04 0.05 + 0.02 0., 18 ± 0.05 0.0 4 hs 0.09 + 0.06 0.06 ± 0.02 0. .20 + 0.04 0.05 ± 0.01 EXAMPLE 13 Modulation of acute pancreatitis induced by bile acid using murine IT9302 Rodents, mice and rabbits are often used in animal models, and recently Genzyme Diagnostics has announced cross-reactivity kits for cytokine search products. Especially, it was possible to measure TNF-a and IFN-? of rabbit by mouse immunoassay equipment. As we were working on a rabbit model, the study of the pathophysiological role of IL-8 in experimental acute pancreatitis, we investigated the effect of murine IT9302 on induced leukopenia (Figure 14). The hypothesis was that even IL-10 could be identical in mice and rabbits and could have effects on the invasion of leukocytes induced by IL-8 in the pancreas during acute pancreatitis.
Experimental model White rabbits of the New Zealand race (Oryctolagus cuniculus) weighing 1.7.4.0 kg for 18-24 hours were aided. The operative processes according to Benerjee et al., 1994 (91) were followed, and acute pancreatitis was induced in rabbits by 5% bile acid given in the pancreatic duct. Murine IT9302 (100 μg / kg) was given through a central vein 30 minutes before bile acid and subsequently at a dose of 100 μg / kg in rabbits immediately thereafter. Plasma samples were collected according to Figure 14. As demonstrated, it was observed that injection of murine IT9302 in rabbits before induced pancreatitis prevented leukopenia as demonstrated in the Figure.
EXAMPLE 14 Modulation of 2nd acute pancreatitis in rabbits induced by bile acid using IT9302 The same experimental model was used as in Example 13 with the exception that the murine IT9302 was changed to human IT9302. Acute pancreatitis was induced by the intraductal injection of 2.0 ml of 5% chemodeoxycholine bile acid (10 animals). Another group of animals was treated with human IT9302 (8 animals), the first dose (100 μg / kg) which was injected subcutaneously and the second dose (100 μg / ml) intravenously, half an hour before the induction of pancreatitis acute Serum samples were collected for cytokine measurements with time intervals of 0, 1, 3, 6, and 12 hours after the induction of acute pancreatitis.
Methods for the measurement of TNF-a and IL-8 in rabbit serum.
The ELISA kit of mouse TNF-a Genzyme code number 80-3807-00 was used in a modified version of the TNF-a measurements in the serum of rabbits. This equipment is constituted by the solid phase monoclonal anti-TNF-α antibody that captures the TNF-a present in the serum, and a polyclonal goat anti-TNF-α antibody, conjugated with peroxidase, substrate and TMB chromagen. The incubation time for serum and normal TNF-a was extended for 4 hours and the development time for substrate incubation was 30 minutes. Measurements of rabbit IL-8 in rabbit serum were carried out by a special IL-8 ELISA kit, a grant from Professor Kouj i Matsushima, Tokyo, Japan. rabbit guinea pig, anti-IL-8, monoclonal, and rabbit anti-guinea pig immunoglobulin G antibodies conjugated with alkaline phosphatase were used as capture, second antibodies and detection, respectively For methods, see Ikeda et al. (106 ).
Result TABLE 4 Acute pancreatitis in rabbit, by 5% bile acid, serum TNF-a content (pg / ml ± SEM) 0 1 3 6 9 12 1210 ± 396 1206 + 239 1918 + 374 1662 + 357 1884 ± 698 915 ± 431 Acute pancreatitis, treated with human IT9302, 30 min before 5% bile acid, serum TNF-a content (pg / ml ± SEM) 0 1 3 6 9 12 347 ± 77 739 ± 339 697 + 146 658 ± 156 872 ± 594 493 ± 203 TABLE 5 Acute pancreatitis, by 5% bile acid, serum IL-8 content (pg / ml ± SEM) 0 1 3 6 9 12 1154 ± 351 780 ± 153 2210 ± 459 2690 ± 468 2196 + 1058 1833 ± 11 Acute pancreatitis, treated with human IT9302, 30 min before 5% bile acid, IL-8 content in sue (pg / ml + SEM) 0 1 3 6 9 12 875 ± 181 695 ± 205 900 ± 178 1037 ± 244 1207 ± 210 1087 ± 21 COMPENDIUM Maximal induction of TNF-a was achieved at 3 hours, and maximal IL-8 at 6 hours. The levels of both TNF-a and IL-8 in the blood circulation were down-regulated markedly from 4 to 12 hours. The levels of pancreatic enzymes in the blood were also measured (amylase, lipase and tryptase) and showed all peaks after 3-6 hours after the induction of acute pancreatitis, but none of these enzymes seemed to be affected by the treatment with IT9302. The histologically similar pancreatic changes were observed in the two groups while there was a significant reduction in the infiltration of intertiscial neutrophils in the lungs. conclusion It downregulates the production of TNF-α and IL-8 in a model of acute pancreatitis induced by biliary acid, and blocks the infiltration of neutrophils in the lungs of these treated rabbits, thus preventing the development of the ARDS-like syndrome in these animals, resulting in a reduction in mortality from 60% to 0% after 6 hours. Interieucine 1 s5J3 was considered as an important inducer of acute pancreatitis (see reference 107) and thus supporting the hypothesis that IT9302 can block all effects inducible by IL-1. The reason why the human IT9302 can be used in a rabbit model could be explained by the observation made by Dan Gaur et al. And published this year in Nature (108) where it showed that rabbit proteins (Lagomorfa) are phylogenetically more close to primate proteins (human) than to other rodent proteins (mouse, rat, ...).
EXAMPLE 15 IT9302 as a cancer treatment and in the prevention of metastasis.
It was recently demonstrated (111) that the systemic administration of cellular IL-10 induces an effective, specific and long-lasting immune response against established tumors in mice in vivo. It was suggested that this effect be partially explained by the renewed effects of IL-10 on various cell types, including co-stimulation of T cell proliferation, chemoattraction of CD8 + T cells, and stimulation of killer cell activity activated with lymphokine. It was also observed that human IL-10 can reverse the local immunosuppressive effect of viral IL-10. Another research group confirmed the potential therapeutic role of IL-10 administration in cancer (112) because they observed that IL-10 inhibits metastasis through a mechanism dependent on natural killer cells in an in vivo experiment with mice that have different tumors, including malignant melanoma of metastasis. Kundu et al. (1996) (113) reported that IL-10 exerts anti-metastatic and antitumor effects in a murine model of human breast cancer. These observations indicated a potential role for IL-10 and for the administration of IT9302 in the biological therapy of cancer.
EXAMPLE 16 IT9302 as an immune adjuvant in the treatment of viral infections.
It is known that IL-10 possesses certain antiviral capabilities. In this way, Kollmann et al. (1996) (114) found that IL-10 inhibits acute HIV infection in vivo from SCID mice with human fetal thyroid and liver. It was observed that two of the applicants / researchers, when applying IT9302 in a cream formulation (total dose of 400 to 500 μg) under plastic wrap occlusion in the skin of the back for 24 hours, developed an increase in their number total CD8 + T cells (60% and 90%, respectively) and an increase in serum IRAP concentration from 1 ng / ml to ng / mml. In seven untreated volunteers, the IRAP concentration never exceeded 1.1 ng / ml. As an accidental and unexpected observation, both researchers observed that common warts (due to cutaneous HPV infection) showed clear signs of inflammation with redness and itching around the warts 3 to 4 days after application and a cream containing IT9302. The inflammatory reaction occurred in a synchronized manner on several fingers for one of the test persons, while the other person only had one wart on one finger. In both cases, the warts gradually decreased in size gradually over the next 4 to 7 days so that there were no clinical signs of remaining wart infection after 10 to 11 days. In a follow-up 2 months later, there were no signs of infection recurrence. Therefore, it was found that IT9302, possibly through a systemic activation of NK cells and / or the activity of cytotoxic CD8 + T cells, is able to elicit a latent immunoresponse against cutaneous HPV infection, resulting in eventually the clinical remission of the virus infection. In this way, IT9302 is a possible therapeutic alternative for the treatment of virus infections such as HIV and HPV infections.
EXAMPLE 17 I 9302 as an immune adjuvant in the treatment of inflammatory diseases of the joints (arthritis).
Anti-IL-1 therapy as well as TNF-a therapy appear to have significant clinical potential in the treatment of arthritis (Maini, 1996) (115). As described elsewhere in this document, it was found that IT9302 is an inhibitor of TNF-a production as well as an IRAP stimulator.
(IL-1 receptor antagonist protein) of human mononuclear cells. In this way, IT9302 is a potential treatment modality of arthritis. This was supported by an observation of one of the applicants of this invention who, after applying IT9302 (approximately 500 μg) in a cream base on the skin for 24 hours, observed a strong reduction in chronic joint pains due to to arthritis. This observation was made three times and in each case the symptoms were gradually reduced during the following week after the removal of the application of IT9302. In this way, in vivo support was found for the arguments for using IT9302 in the treatment of acute or chronic inflammatory reactions such as arthritis or other autoimmune diseases.
REFERENCES Bendtzen K. Lymphokines in inflamation. Inflammation Basic Mechanisms Tissue Injuring Principles and Clinical Models (P Venge &A Lindbom eds) 1985, Almqvist & Wikselle International. Stockholm: 187-217.
Bendtzen K, Interleukin-1, Interleukin-6, and tumor necrosis factor in infection, inflammation and immunity. Immunol Lett 1988; 19: 183-192.
Larsen C.G. Leukocyte activating and chemotatic cytokines in cell-mediated immune reactions of the human skin. Dermatovenerol Act. 1991; suppl. 160: 1-48 Florentino D. F., M. Bond W., and T. R. Mosmann, 1989. Two types of mouse helper T cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Thl clones. J. Exp. Med., 170: 2081. viera P., R. de Wall-Malefyt, M. -N. Dang, K. E. Johnson, R. Katelein, D. F. Fiorentiono, J. E. de Vries, M. -G. Roncarolo, T. R. Mosmann, and K. W. Moore, 1991. Isolotion and expression of human cytokine synthesis inhibitory factor (CSFI / IL-10) cDNA clones: homology to Epstein-10 Barr virus open reading frame BCRF. Proc. Nati Acad. Sci. (USA), 88: 1172.
Moore, K.W., O 'Garra A., de Waal Malefyt R., Vieira, Mossmann T.R. 1993. Interleukin-10, Annu Rev. Immunol, 11: 165-90.
Kim, J.M., Brannan, C.l. Copeland N.G., Jenkins, N.A., Khan, T.H., Moore, K.W. 1992. Structure of the mouse interleukin-10 gene and chromosomal localization of the mouse and human genes. J. Immunol 148: 3618-23.
Carter, D.B., Deibel, M.R-Jr, Dunn, C.J. et al 1990. Purification, cloning, expression and biological characterization of an interleukin-1 receptor antagonist protein. NATURE 344: 633-638. 9. Hannum, C.H., Wilcox, C.J., Arend, W.P. and 5 co-workers. 1990. Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor. Nature 343: 336-40.
. Firestein, G.S., Boyle, D.L., Yu, C, et al. 10 1994. Synovial interleukin-1 receptor antagonist and interleukin-1- balance in rheumatoid arthritis. Arthritis Rheum 37: 644-652. 11. Fisher, C.J. -Jr., Slotman, G.J., Opal, S.M., 15 Pribble, J.P. et al. 1994. Initial evaluation of recombinant interleukin-1 receptor antagonist in the treatment of sepsis syndrome: a randomized, open-label, placebo-controlled multicenter trial. The IL-1RA Sepsis syndrome Study Group. 20 Crit-Care-Med. 22: 12-21. 12. de Waal-Malefyt, R., Haanen J., Spits, J., et al. 1991. IL-10 and viral IL-10 strongly reduces antigen-specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via down-regulation of class II MHC expression. J. Exp. Med. 174: 915-24: Gazzinelli, R.T., Oswald, I.P., James, S.L., Sher, .A., 1992. IL-10 inhibits parasite killing and nitric oxide production by IFN - \ - activated macrophages. J. Immunol. 148-1792-96.
Jinquan, T., Larsen, C.G., Gesser, B., Matsushima, K., Thestrup-Pedersen, K. 1993.
Human IL-10 is a chemoattractant for CD8 + T lymphocytes and an inhibitor of IL-8-induced CD4 + T lymphocyte Migratian. Journal of Immunology, 151: 4545-4551.
Rousset F., E. Garcia, T. Defrance, C. Peronne, D. -H. Hsu, R. Kastelein, K. W. Moore, and J. Banchereau, 1992. IL-10 is a potent growth and differentiation factor for activated human B lymphocytes. Proc. Nati Acad. Sci. USA, 175: 671.
Howard, M., 0 'Garra, A., Ishida, H., de Waal Malefyt, R., de Vries, J. 1992. Biological properties of Interleukin-10, J. Clin. Immunol 12: 239-47. 17. Kuhn, R., Lohler, J., Rennick, D., Rajewsky, K., 5 Muller, W. 1993. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75: 263-74. 18. Sher, A., Florentino, D.F., Caspar, P., Pearce,. E., Mosmann, T. 1991. Production of IL-10 by CD4 + lymphocytes correlates with down-regulation of Thl cytokine synthesis in helminth infection. J. Immunol. 147: 2713-16. 19. Clerici, M., Shearer, G.M. 1993 Immunology Today. 14: 107-111.
. Bry, K., Lappalainen, U. 1994. Interleukin-4 and transforming growth factor-beta 1 modulate the production of interleukin-1 receptor antagonist and prostaglandin E2 by decidual cells. Am-J-obstet-Gynecol 170 (4): 1194-1198. 21. Firestein, G., S., Boyle, D.L., Yu, C, Paine, M.M., Whisenand, T.D., Zvaifler, N.J., Arend, W. P. 1994, Synovial interleukin-1 receptor antagonist and interleukin-1 balance in rheumatoid arthritis. Arthritis Riieu, 37/5: 644-652. 22. Roberge, C.J., De-Medicis, R., Dayer, J.M., Rola-Pleszcyczynski, M., Naccahe, P.H., Poubelle, P.E. 1994. Cystal-induced neutrophil activation: V. Differential production of biologically active IL-1 receptor antagonist. J.
Immunol 152/11: 5485-5494. 23. McCall, R.D., Haskill, S., Zimmermann, E.M., Lund, P.K., Thompson, R.C., Sartor, R.B. 1994. Tissue interleukin 1 and interkeukin-1 receptor antagonist expression in entercolitis in resistant and susceptible rats. Gastroendterology (4): 960-72.
. Kimble, R.B., Vannice, J.L, Bloedow, D.C., Thompson, R. C, Hopfer, W., Kung, V. T., Brownfield, C, Pacifici, R. 1994. Interleukin- 1 receptor antagonist decreases bone loss and bone resorption in ovariectomized rats. J. Clin Invest. 93/5: 1959-1967. 25 Kline, J. N., Geist, L.J., Monick, M.M., Stinski, M.F., Hunninghake, G.W., 1994. J. Immuno1. 152 (5): 2351-7.
Tompkins, R. G. 1994. Human recombinant interleukin-1 receptor antagonist in the treatment of sepsis syndrome (editorial; comment). Crit-Care-Med. 22 (1): 3, 22 (1): 12-21.
Everaerdt, B., Brouckaert, P., Fiers, W. 1994. Recombination IL-1 receptor antagonist protects against TNF-induced lethality in mice. J. Immunol. 152/10: 5041-5049.
Fisher, C. J. Jr. , Slotman, GJ, Opal, SM, Pribble, JP, Bone, R. C, Emmanuel, G., Ng, D., Bloedow, D. C, Catalano, MA 1994. Initial evaluation of human recombination interleukin-1 receptor antagonist in the tratment of sepsis syndrome: a randomized, open-label, placebo-controlled multicenter trial. The IL-1RA Sepsis Syndrome Study Group (see comments). Crit-Care-Med. 22 (1): 12-21, 22 (1): 3 30. Gomez-Reino-Carnoto, J. J. 1994. New therapies in rheumatoid arthritis. Med-Clin 543-545 32. Nyshihara, T., Ohsaki, Y., Ueda, N., Saito, N., Mundi, GR 1994. Mouse interleukin-1 receptor antagonist induced by actinobacillus actinomycetemcomitans lipopolysaccharide blocks the effects of interleukin-1- bone resorption and osteoclast- like cell formation. Infect- 10 Immun. 62 (2): 390-7. 33. Simon, C, French A., Piquette, G. N., el- Danasouri, I., Zurawski, G., Dang, W., Polan, M.L. 1994. Embryonic implantation in mice is blocked by interleukin-1 receptor antagonist (see comments). Endocrinology 134 (2): 521-8, 134 (2): 519-20. 34. Baergen, R., Benirschke, K., Ulich, T. R., 1994.
Cytocine expression in the placenta. The role of interleukin 1 and interleukin 1 receptor antagonist expression in chorioamnionitis and parturition. Arch-Pathol-Lab-Med. 118 (1): 52-5 35. Tang, W.W., Feng, L., Vannice, J.L., Wilson, C.B. 1994. Interleukin-1 receptor antagonist ameliorates experimental antigomerular basement membrane antibody-associated flomeulonephritis.
J. Clin-Invest. 93 (1): 279-9. 36. Cassatella, M.A., Meda, L., Gasperini, S., Calzetti, F., Bonara, S. 1994. 37. Interleukin 10 (IL-10) upregulates IL-1 receptor antagonist production from lipopolysaccharide-stimulated human polymorphonuclear leukocytes by delaying mRNA degradation. J. Exp-Med. 179/5: 1695-1699. I5 38. Mancini, R., Benndetti, A., Jezequel, A.M. 1994. An interleukin-1 receptor antagonist decrease fibrosis induced by dimethylnitrorsamine in rat liver. Virchows- 20 Arch. 424/1: 25-31. 39. Lukacs, N. W., Kunkel, S.L., Burdick, M.D. Lincoln, P.M., Strieter, R. M. 1993. 40. Interleukin-1 receptor antagonist blocks che okine production in the mixed lymphocyte reaction. Blood, 82 (12): 3668-74 41. Bandara, G., Mueller, G.N., Galea-Lauri, J., Tindal, M.H., Georgescu, H.I., Suchanek, M.K., Hung, G.L., Gloriso, J.C., Robbins, P.D., Evans, C.H. 1993. 42. Intraarticular expression of bislogically active interleukin 1-receptor-antagonist protein by ex vivo transfer. Proc-Natl-Acad-Sci-U-S-A-. 90 (22): 10764-8 43. Dinarello, C. A. 1994. Anti-interleukin-1 strategies in the treatment of septic shock syndrome. Can-J-infect-Dis. 5 (suppl A): 9A- 16A. 44. Oelmann, E., Topp, M. S. Reufi, B., Papadimitriiou, C, Koenings ann, M., 'Oberberg, D. Thiel, E., Berdel, W. E. 1994. Int-J-Oncol 4/3: 555-551 45. Estrov, Z. 1993. Interruption of autocrine and paracrine growth-stimolatory mechaisms: a new therapeutic stratefy for chronic myelogenous leukemia. Semin-Hematol. 30 (3 suppl 3): 35-6 46. Wooley, P.H., Whalen, J.D., Chapman, D.L., Berger, A.E., Richard, K.A., Aspar, D.G., Staite, N.D. 1993. The effect of an interleukin- 1 receptor antagonist protein on type II collagen-induced arthritis and antigen-induces arthritis in mice. Arthritis Rheum. 36 (9): 1305-1314. 47. Peterson, C.M., Hales, H.A., Hatasaka, H.H., 15 Mitchell, M.D., Rittenhouse, L., Jones, K.P. 1993. Interleukin-1 beta (IL-1 beta) modulates prostaglandin production and the natural IL-1 receptor antagonist inhibits ovulation in the optimally stimulated rat ovarian ~~ perfusion 20 - model. Endocrinology 133 (5): 2301-2306. 48. Estrov, Z., Kurzrock, R., Talpaz, M. 1993. Role of interleukin-1 inhibitory molecules in therapy of acute and chronic myelogenous leukemia.
Leuk. Lymphoma 10 (6): 407-418. 49. Chensue, S.W., Bienkowski, M., Eessalu, T.E., Warmington, K.S., Hershey, S.D., Lukacs, N.W., Kunkel, S.L. 1993. nous IL-1 receptor antagonist protein (IRAP) regulates schistosome egg granuloma formation and the regional lymphoid response. J. Immunol. 151 (7): 3654-3662. 50. Bowyer, J.F., Davies, D.L., Schmued, L., Broening, H.W., Newport, G.D., Slikker, W. Jr. , Holson, R.R. 1994. Further studies of the role of hyperthermia in metham-pehtamine neurotoxicity. J. Pharmacol. Exp. Ther 268/3: 1571-1580. 51. Cole, O.F., Sullivan, M.H.F., Eider, M.G. 1993. The? Interleukin-l- receptor antagonist is a partial agonist of prostaglandin synthesis by human decidual cells. Prostaglandins 46/6: 493-498. 52. Jenkins, J.K., Arend, W.P. 1993. Interleukin 1 receptor antagonist production in human monocytes is induced by IL-lalfa, IL-3, and IL-4 and GM-CSF, Cytokine 5/5: 407-415 Coceani, F., Lees, J., Redford, J., Bishai, I. 1992. Interleukin-1 receptor antagonist: effectiveness against interleukin-1 fever. Dog. J. Pharmacol. 70 (12): 1590-1596.
Schiro, R., Longoni, D., Rossi, V., Maglia, O., Doni, A., Arsura, M., Carrara, G., Masera, G., Vannier, E., Dinarello, C.A., Rambaldi, A., Biondi, A. 1994. Suppression of juvenile chronic myelogenous leukemia colony growth by interleukin-1- receptor antagonist. Blood 83/2: 460-465.
Watson, ML, Smith, D., Bourne, AD, Thompson, RC, Westwick, J. 1993. Cytokines contribute to airway dysfunction hyperreactivity, pulmonary eosinophil accumulation and tumor necrosis factor generation by pre-treatment with and interleukin-1- receptor antagonist . Am. J. Respir. Cell. Mol. Biol. 8 (4): 365-369.
Abhyankar, S., Gilliland, D.G., Ferrara, J.L.M. 1993. Interlukin-1- is a critical effector molecule during cytokine dysregulation in graft-versus-host disease to minor histocompatibility antigens. Transplantation 56/6: 1518-1523.
Lan, H.Y., Nikolic Paterson, D.J., Zara a, M., Vannice, J.L., Atkins, R.C. 1993. Suppression of experimental crescentic glomerulonephitis by the interleukin-1 receptor antagonist., Kidney Int. 43 (2): 479-485.
Herve, P. 1993. Prevention and treatment of acute GvHD -New modalities. Nouv. Rev. Fr. Hematol. 3/3: 295-297 Conti, P., Panara, MR, Barbacane, RC, Placido, FC, Bongrazio, M., Reale, M., Dempsey, RA, Fiore, S. 1992. Blocking the interleukin-1 receptor inhibits leukotriene B4 and prostaglandin E2 generation in human monocyte cultures. Cell Immunol. 145 (1): 199-209. 60. Kristensen, M., Deleuran, B., Eedy, D.J., Feldmann, M., Breathnach, S.M., Brennan, F.M. 1992. Distribution of interleukin-1 receptor antagonist 'protein (IRAP), interleukin-1 receptor, and interleukin-1 alpha of IRAP in psoriatic lesional epidermis. Br. J. Dermatol. 127 (4): 305-311". 61. Romero, R., Sepulveda, W., Mazor, M., Brandt, F., Cotton, D.B., Dinarello, C.A .:, Mitchell, M.D. 1992. The natural interleukin-1 receptor antagonist in term and pre-term perturbation. Am. J. Obstet. Gyencol 164 (4 Pt 1): 863-872. 62. Dinarello, C.A. 1992. Reduction of inflammation by decreasing production of interleukin-1 or by specific receptor antagonism. Int. J. Tissue. React. 14 (2): 65-75. 63. Conti, P., Panara, MR, Barbacane, RC, Bongrazio, M: Dempsey, RA, Reale, M. 1993. Human recombinant IL.l receptor antagonist (IL-lRa) inhibits leukotriene B4 generation from human monocyte suspensions stimulated by lipopolysaccharide (LPS) Clin. Exp. Immunol. 91/3: 526-531. 64. DeForge, L.E., Tracey, D.E., Kenney, J.S., Remick, D.G. 1992. Interleukin-1 receptor antagonist protein inhibits interleukin-8 expression in lipopolysaccharide-stimuled human whole blood. Am. J. Pathol. 140 (5): 1045-1054. 65. Porat, R., Poutsiaka, D.D., Miller, L.C., Granowitz, E.V., Dinarello, C.A. 1992. Interleukin-1 (IL-1) blockade receptor reduces endotoxin and Borrealia burgdorferi-stimulated IL-8 synthesis in human monoclear cells. Faseb. J. 6 (7): 2482-2486. 66 Boermeester, M.A., van Leeuwen, P.A.M., Scheneider, A.J., Houdijk, A.P.J., Ferwerda, C.C., Wesdorp, R.I.C. 1993. Interleukin-1 receptor antagonist: A new therapeutic agent in the treatment of septic syndrome. Ned. Tijdschr. Geneesks 137/7: 337-342. 67. Smith, R.J., Chin. J.E., Sam, L.M., Justen, J.M. 1991. Biologic effects of an interleukin-1 receptor antagonist protein on interleukin-1-stimulated cartilage erosion and chondrocyte responsiveness. Arthsitis rehum. 34 (1): 78-83.
Conti, P. Barbacane, R.C., Panara, M.R., Reale, M., Placido, F.C., Fridas, S., Bongrazio, M., Dempsey, R.A. 1992. Human recombinant interleukin-1 receptor antagonist (hrlL-lra) enhances the stimulatory effect of interleukin-2 on natural killer cell activity against MOLT-4 target cells. Int. J. Immunopharm. 14/6: 987-993 Selig, W., Tocker, J. 1992. Effect of interleukin-1 receptor antagonist on antigen-induced pul onary responses in guinea pigs. Eur. J. Pharmacol. 213/3: 331-336.
McCarthy, P.L. Jr. , Abhyankar, S., Neben, S., Newman, G., Sieff, C, Thompson, R.C., Burakoff, S.J., Ferrara, J.L.M. 1991. Inhibition of interleukin-1 by an interleukin-1 receptor antagonist prevents graft-versus-host diseases. Blood 78/8: 1915-1918. 71. Estrov, Z, Kurzrock, R., Wetzler, M., Kantarjian, H., Blake, M., 'Harris, D., Gutterman, JU, Talpaz, M. 1991. Suppression of chronic myelongenous leukemia colony growth by interleukin- 1 (IL-1) receptor antagonist and soluble IL-1 receptors: A novel application for inhibitors of IL-1 activity. Blood 78/6: 1476-1484 72. Thomas, TK, Will, PC, Srivastava A., Wilson, CL, Harbison, M., Little, J., Chesonis, RS, Pignatello, M., Schmolze, D., Symington, J., Kilin, PL, Thompson , RC 1991. Evaluation of an interleukin-1 receptor antagonist in the mouse acetic acid-induced colitis model. Agents Actions 34 / 1-2: 187-190. 73. Cárter, D.B., Deibel, M.R. Jr. , Dunn, C.J., Tomich, C.S.C., Laborde, A.L., Slightom, J.L., Berger, A.E., Bienkowski, M.J., Sun, F.F., McEwan, R.N., Harris, P.K.W., Yem, A.W., Waszak, G.A., Chosay, J.G., Sieu, L.C., Hardee, M.M., Zurcher Neely, H.A., Reardon, I.M., Heinrinkson, R.L. et al. 1990. Purification, cloning expression and biological characterization of an interleukin-1 receptor antagonist protein. Nature 344/6267: 633-638. 74. Larsen C.G, Anderson A.O, Apella E., Oppenheim J.J., Matsushima K., 1989. Science 243: 1464; 75. Larsen C.G., Jinquan T., Deleurant B., Thestrup-Pedersen K. 1993, IL-10 is a potent regulator of the chemotactic response of mononuclear cells, but not of granulocytes. J. Invest. Dermatol. Vol. 100, No. 6 76. Sankoff and Kruskal in chapter 1 of "Time Warps, string Edits, and Macromolecules: The theory and Practice of Edits, and Macromolecules: The Theory and Practice of Sequence Comparison "(Addison-Wesley, Reading, Mass. 1983). 77 Berzofsky, Science 229, (1985) 932-940 78. Bowie et al., Science 247, (1990) 1306-1310 79. Wasserman et al., J. Immunol. 87, 1961, 290-295 80. Levine et al., Methods in Enzymology 11, 1967, 928-936. 81. Lewis et al., Biochemistry 22, 1983, 948-954 82. Rene de Waal Maletyt, John Abrahams, Bruce Bennett, Cari G. Figdor and Jan E. de Vries (1991), Interleukin 10 (IL-10) Inhibits Cytokine Synthesis by Human Monocytes: An Autoregulatory Role of IL-10 Produced by Monocytes. J. Exp. Med. 174, 1209-1220 3. Szoka et al., Ann. Rev. Biophys, Bioeng. 9, 1980, 467 !4. U.S. Patent No. 4,235,871 . US 4,501.72! 86 US 4,837.02! 87. Walter H. Gotlieb, John S. Abrams, Joanna M. Watson, Thierry J. Velu, Jonathan S. Berek, Otoniel Martinez-Meza (1992), Presence of interleukin 10 (IL-10) in the ascites of patients with ovarian and other intra-abdominal cancers. Cytokine 4, No. 5, 385-390 88. Blancho G., P. Gianello, Sh. Germana, M. Baetscher, D.H. Sachs and Chr. LeGuern (1995), Molecular identification of porcine interleukin : Regulation of expression in kidney allograf model. Proc. Nati Acad. Sci. USA 92, 2800-2804 83. Howard, M., T. Muchamuel, S. Andrade, S. Menon (1993), Interleukin 10 protects mice from lethal endotoxemia. J. Exp. Med. 177, 1205-1208. 90. Chernoff, A.E., E.V. Granowitz, L. Shapiro, E. Vannier, G. Lonnemann, J.B. Angel, J.S. Kennedy, A.R. Rabson, S. Wolff, C.A. Dinarello (1995), A randomized, controlled trial of IL-10 in humans. Inhibition of inflamatory cytokine production and immune responses. J. Immunol. 154, 5492-5499. 91. Banerjee, A.K., S.W. Galloway and A.N. Kingsnorth (1994), Experimental models of acure pancreatitis. Br. J. Surg. 81, 1096-1103 92. Hong, S.S., DS. S. Chin, T.S. Cho, S.E. Kim (1962), Experimental pancreatitis induced by alcohol and bile in rabbits. Annals of Surgery 156 (6), 929-939. 93. Bodansky, M. (1984), Principies of Peptide Synthesis, Springer-Verlang, Berlin 94. Pelton, J.T., et al., Proc. Nati Acad. Sci. USA 82, 233-239. 95. Dyson, * H. et al. (1988), Annual Review of Biophysics and Biophysical Chemistry 17, 305-324) 96. Nakanishi, H., et al. (1993), Peptidomimetics of the immunoglobulin supergene fa ily a review. Gene 137, 51-56. 97 US 5,446,128 98. Walter et al. (1995), Biochemistry 34, 12118-25 99. Marshall, G.R. (1993), Tetrahedron 49, 3457-3558 100. Merrifield, R.B. (1963), J. Amer. Chem. Soc. 85, 2149-2154). 101. Kent, S.B.H. (1988), Annu. Rev. Biochem. 57, 957-989 102. Carpino, L.A. and Han, G.Y. (1972), J. Org. Chem. 37, 3404-3409 103. Stewart, J.M. and Young, J.D. (1983), "Solid Phase Peptide Synthesis", Pierce Chemical Company, Rockford, Illinois 104. Atherton, E. and Sheppard, R.C. (1989), "Solid Phase Peptide Synthesis", IRL Press at Oxford University Press 105. Pennington, M.W. and Dunn, B.M. (eds.) (1994), "Peptide Synthesis Protocols", Humana Press, totowa, New Jersey 106. Ikeda, N. et al. (1995), Infection and Immunity, 4812-4817. 107. Fink, G.W. and Norman, J.G. (1996), "Intrapancreatic Interleukin-lβ Gene Expression by Specific Leukocyte Populations during Acute Pancreatitis", J. Surgical Research 63, 369-373. 108. Gaur, D. et al. (1996), "Phylogenetic position of the order Lagomorpha (rabbits, hares and allies)", Natue, 379, 333-335 109. Poli, G. et al. (1994), Proc. Nati Acad. Sci. USA 91, 108-112 110. Jensen, I.M. et al. (1993), Analyt, Cell. Pathol. 5, 213-223 111. Berman, R.M., Suzuki, T. et al. (1996), "Systemic administration of cellular IL-10 induces in effective, specific, longlived immune response against established tumors in mice", J. Immunol, 157, 231-238. 112. Zheng, L.M., Ojcius, D.M. et al. (1996), "IL-10 inhibits tumor metastasis through an NK cell-dependent mechanism", J. Exp. Med. 184, 579-584.
Kundu, N., Beaty, T.L. et al. (1996), "Antimetastatic and anti-tumor activities of IL-10 in a murine model of breast cancer", J. Nati. Cancer Inst. 88, 479-480 Kollmann, T.R., Pettoello-Mantovani, M. et al (1996), "Inhibition of acute in vivo HIV infection by human IL-10 treatment of SCID mice with planted with human fetal thymus and liver", Proc. Nati Acad. Sci. USA 93, 3126-3131 Maini, R.N. (1996), "A perspective on anti-cytokine, and anti T cell directed therapies in rheumatoid arthritis", Clin, Exp. Rheumatol, 13, suppl. 12, S35-40.
LIST OF SEQUENCES (1) GENERAL INFORMATION: (i) APPLICANT: (A) RECIPIENT: Steeno Research Group A / S (B) STREET: Dunbirkevej 6 (C) CITY: Odense SV (E) COUNTRY: Denmark (F) POSTAL CODE: 5250 ii) TITLE OF THE INVENTION: Immunomodulators Iii) SEQUENCE NUMBER: 23 (iv) COMPUTER LEGIBLE FORM: (A) TYPE OF MEDIUM: Flexible disk (B) COMPUTER: compatible with IBM PC (C) OPERATING SYSTEM: PC-DOS / MS-DOS (D) PROGRAM: Patentln Relay # 1.0, Version # 1.30 (EPO) 2) INFORMATION FOR SEQ ID NO: 1: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 1: Wing Tyr Met Thr Met Lys lie Arg Asn 1 5 (2) INFORMATION FOR SEQ ID NO: 2: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETIC: NO (xi) DESCRIPTION FOR SEQ. ID No: 2 Wing Tyr Met Thr lie Lys Met Arg Asn 1 5 (2) INFORMATION FOR SEQ ID NO: 3: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 3; Wing Phe Met Thr Leu Lys Leu Arg Asn 1 5 (2) INFORMATION FOR SEQ ID NO: 4 (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 4 Wing Tyr Met Thr Met Lys Val Arg Glu 1 5 ) INFORMATION FOR SEQ ID NO: 5: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETIC: NO (xi) DESCRIPTION FOR SEQ. ID No: 5 Gly Tyr Met Thr Met Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 6: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide 'iii) HYPOTHETICAL: NO [xi) DESCRIPTION FOR SEQ. ID No: 6: Wing Phe Met Tyr Met Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 7: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 7 Wing Tyr lie Thr Met Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 8 (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 8: Wing Tyr Leu Thr Met Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 9: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide iii) HYPOTHETICAL: NO [xi) DESCRIPTION FOR SEQ. ID No: 9: Wing Tyr Val Thr Met Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 10: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 10 Wing Tyr Met Thr lie Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 11: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 11: Wing Tyr Met Thr Leu Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 12: i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO ; xi) DESCRIPTION FOR SEQ. ID No: 12 Wing Tyr Met Thr Val Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 13: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) ) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 13 Wing Tyr Met Thr Met Lys lie Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 14: ;) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 14 Wing Tyr Met Thr Met Lys Met Arg Asp 1 5 (2) INFORMATION FOR SEQ ID NO: 15: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 15 Wing Tyr Met Thr Met Lys Val Arg Asp 1 5 ; 2) INFORMATION FOR SEQ ID NO: 16: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear iii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO [xi) DESCRIPTION FOR SEQ. ID No: 16: Wing Tyr Met Thr Met Lys lie Arg Gln 1 5 ; 2) INFORMATION FOR SEQ ID NO: 17: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear ; ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 17 Wing Tyr Met Thr Met Lys lie Arg Glu 1 5 ) INFORMATION FOR SEQ ID NO: 18: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear ; ii) TYPE OF MOLECULE: peptide iii) HYPOTHETIC: NO (xi) DESCRIPTION FOR SEQ. ID No: l Ser Pro Gly Gln Gly Thr Gln Ser Glu 1 5 (2) INFORMATION FOR SEQ ID NO: 19: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 6 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 19: Thr Xaa Lys Xaa Arg Xaa 1 5 (2) INFORMATION FOR SEQ ID NO: 20 (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 7 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 20: Xaa Thr Xaa Lys Xaa Arg Xaa 1 5 ) INFORMATION FOR SEQ ID NO: 21: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 8 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETIC: NO (xi) DESCRIPTION FOR SEQ. ID No: 21: Xaa Xaa Thr Xaa Lys Xaa Arg Xaa 1 5 (2) INFORMATION FOR SEQ ID NO: 22: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 9 amino acids (B) TYPE: amino acid (C) TYPE OF HEBRA: D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 22 Xaa Xaa Xaa Thr Xaa Lys Xaa Arg Xaa 1 5 (2) INFORMATION FOR SEQ ID NO: 23: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 27 base pairs (B) TYPE: nucleic acid (C) TYPE OF HEBRA: simple D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: DNA (genomic) (iii) HYPOTHETICAL: NO (xi) DESCRIPTION FOR SEQ. ID No: 23: GCCTACATGA CAATGAAGAT ACGAAAC It is noted that in relation to this date, the best method known by the applicant to carry out the present invention is that which is clear from the present description of the invention.
Having described the invention as above, the content of the following is claimed as property:

Claims (17)

1. The use of a substance or polypeptide according to the formula: Xi-Xs-Xs-Thr-Xy-Lys-Xs-Arg-Xs (SEQ ID NO: 22), wherein X2 is Tyr or Phe, X3, X4 and Xs are independently selected from the group consisting of Met, Lie, Leu and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu, optionally at least one of Xi, X2, X3, X4, X5 and Xs is independently substituted with an unnatural or unusual amino acid and / or the peptide is cyclized and / or the peptide is stabilized and / or the amino acid residue is amino acid. terminal is acylated and / or the carboxy-terminal amino acid residue is amidated, and the peptidomimetics are modeled on the basis of the above formula for the preparation of a pharmaceutical composition for the reduction of TNFa production.
2. The use of a substance or polypeptide according to the formula Xi-Xa-Xs-Thr-Xy-Lys-Xs-Arg-Xs (SEQ ID NO: 22), wherein Xi is Ala or Gly, X2 is Tyr or Phe, X3 , X4 and Xs are independently selected from the group consisting of Met, Lie, Leu and Val; and X6 is selected from the group consisting of Asn, Asp, Gln and Glu optionally at least one of Xi, X2, X3, X4, X5 and X6 is independently substituted with an unnatural or unusual amino acid and / or the peptide this cyclized and / or the peptide is stabilized and / or the amino-ternal amino acid residue is acylated and / or the carboxy-terminal amino acid residue is amidated, and the peptidomimetics are modeled on the basis of the above formula for the preparation of a pharmaceutical composition for prophylaxis or treatment of pancreatitis.
3. The use of a substance or polypeptide according to the formula X? -X2-X3-Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 22), wherein X2 is Tyr or Phe, X3, X and Xs is independently selected from the group consisting of Met, Lie, Leu and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu, optionally at least one of Xi, X2, X3, X, Xs and Xβ is independently substituted with an unnatural or unusual amino acid and / or the The peptide is cyclized and / or the peptide is stabilized and / or the amino-terminal amino acid residue is acylated and / or the carboxy-terminal amino acid residue is amidated, and the peptidomimetics are modeled on the basis of the above formula for the preparation of a pharmaceutical composition for prophylaxis of treatment of viral infections such as acquired immunodeficiency syndrome (AIDS) or cutaneous HPV infection.
4. A substance or polypeptide having the formula X? -X2-X3-Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 22), characterized in that Xi is Ala or Gly, X2 is Tyr or Phe, X3 X4 and Xs are independently selected from the group consisting of Met, Lie, Leu and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu wherein at least one of Xi, X2, X3, X, X5 and X6 is independently substituted with an unnatural or unusual amino acid, this substance or polypeptide that has at least one of the following properties. a) induces the inhibition of spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by mononuclear cells, peripheral blood, human (PBMC), c) induces the production of interleukin-1 receptor antagonistic protein (IRAP) by human monocytes, d) induces chemotactic migration of human CD8 + T lymphocytes in vitro, e) desensitizes human CD8 + T cells resulting in an insensitivity to rhIL-10, f) suppresses the chemotactic response of human CD4 + T lymphocytes towards IL-8, g) suppresses the chemotactic response of human monocytes towards MCAF / MCP-1, h) inhibits the expression of MHC molecules Class II in human monocytes stimulated by IFN- ?, i) induces IL-4 production by human, normal, cultured CD4 + T cells, j) reduces the production of TNFa in the reaction of mixed leukocytes, human, k) regulates falling The production of TNFa and IL-8 in the rabbit model of acute pancreatitis induced by biliary acid and reduces the infiltration of neutrophils in the lungs of treated rabbits,
5. A substance or polypeptide having the formula X2-X3-Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 21), characterized in that X2 is Tyr or Phe, X3, X and = are independently selected from the group consisting of Met, Lie, Leu and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu wherein at least one of Xi, X2, X3, X4, X5 and X6 is independently substituted with an unnatural or unusual amino acid, the substance or polypeptide having at least one of the following properties a) induces the inhibition of the spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by mononuclear cells, of blood peripheral, human (PMBC), p 3 c) induces interleukin-1 receptor (IRAP) antagonism by human monocytes, d) induces chemotactic migration of CD-8 + human T lymphocytes in vitro, e) desensitizes human CD8 + T cells resulting in an insensitivity to rhIL-10, f) suppresses the chemotactic response of human CD4 + T lymphocytes towards IL-8, g) suppresses the chemotactic response of human monocytes towards MCAF / MCP-1, h ) inhibits the expression of molecules MHC class II cells in human monocytes stimulated by IFN- ?, i) induces IL-4 production by human, normal, cultured CD4 + T cells, j) reduces the production of TNFa in the reaction of mixed leukocytes, human, ) downregulates the production of TNFa and IL-8 in the rabbit model of acute pancreatitis induced by bile acid and reduces the infiltration of neutrophils in the lungs of treated rabbits.
6. A substance or polypeptide having the formula X3-Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 20), characterized in that X3, X4 and X5 are independently selected from the group consisting of Met, lie, Leu and Val; and X6 is selected from the group consisting of Asn, Asp, Gln and Glu, wherein at least one of X3, X4, X5 and Xe is independently substituted with a non-natural or unusual amino acid, this substance or polypeptide having the less one of the following properties a) induces the inhibition of the spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by mononuclear, peripheral blood, human cells (PMBC), c) induces the production of interleukin-1 receptor antagonist protein (IRAP) by human monocytes, d) induces chemotactic migration of human CD-8 + T lymphocytes in vitro, e) desensitizes cells Human CD8 + T resulting in an insensitivity to rhIL-10, f) suppresses the chemotactic response of human CD4 + T lymphocytes towards IL-8, g) suppresses the chemotactic response of human monocytes towards MCAF / MCP-1, h) inhibits the expression of MHC class II molecules in human monocytes stimulated by IFN- ?, i) induces IL-4 production by human, normal, cultured CD4 + T cells, j) reduces the production of TNFa in the reaction of mixed leukocytes, human, k) downregulates the production of TNFa and IL-8 in the rabbit model of acute pancreatitis induced by bile acid and reduces the infiltration of neutrophils in the lungs of treated rabbits.
7. A substance or polypeptide having the formula Thr-X4-Lys-X5-Arg-Xs (SEQ ID NO: 19), characterized in that X4 and Xs are independently selected from the group consisting of Met, Lie, Leu and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu, wherein at least one of X3, X4 / Xs and Xe is independently substituted with a non-natural or unusual amino acid, this substance or polypeptide having the less one of the following properties a) induces the inhibition of spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by mononuclear cells, peripheral blood, human (PMBC), c) induces the production of the interleukin-1 receptor antagonistic protein (IRAP) by human monocytes, d) induces chemotactic migration of human CD-8 + T lymphocytes in vitro , e) desensitizes human CD8 + T cells resulting in insensitivity to rhIL-10, f) suppresses the chemotactic response of human CD4 + T lymphocytes towards IL-8, g) suppresses the chemotactic response of human monocytes towards MCAF / MCP -1, h) inhibits the expression of MHC class II molecules in human monocytes stimulated by IFN- ?, i) induces IL-4 production by human, normal, cultured CD4 + T cells, j) reduces the production of TNFa in the reaction of mixed leukocytes, humans, k) down-regulates the production of TNFa and IL-8 in the rabbit model of acute pancreatitis induced by bile acid and reduces the infiltration of neutrophils in the lungs of rabbits. ados.
8. A substance or peptide according to any of claims 4-7, characterized in that it is cyclized.
9. A substance or peptide according to any of claims 4-7, characterized in that it is stabilized.
10. A substance or peptide according to any of claims 4-7, characterized in that the amino-terminal amino acid residue is acylated.
11. A substance or peptide according to any of claims 4-7, characterized in that the carboxy-terminal amino acid residue is amidated.
12. A polypeptide modeled on the formula X? -X2-X3-Thr-X4-Lys-X5-Arg-Xs (SEQ ID NO: 22), caracerized because: Xi is Ala or Gly, X2 is Tyr or Phe, X3 , X and X5 are independently selected from the group consisting of Met, Lie, Leu and Val; and Xs is selected from the group consisting of Asn, Asp, Gln and Glu the peptidomimetics having at least one of the following properties a) induces the inhibition of the spontaneous production of IL-8 by human monocytes, b) induces the inhibition of IL-8 production induced by IL-1β by mononuclear, peripheral blood, human (PMBC) cells, c) induces the production of the interleukin-1 receptor antagonistic protein (IRAP) by human monocytes, d) induces chemotactic migration of human CD8 + T lymphocytes in vitro, e) desensitizes human CD8 + T cells resulting in an insensitivity to rhIL-10, f) suppresses the chemotactic response of human CD4 + T lymphocytes towards IL -8, g) suppresses the chemotactic response of human monocytes towards MCAF / MCP-1, h) inhibits the expression of MHC class II molecules in human monocytes stimulated by IFN- ?, i) induces IL-4 production by cell s human CD4 +, normal, cultured, j) reduces the production of TNFa in the reaction of mixed leukocytes, humans, k) down-regulates the production of TNFa and IL-8 in the rabbit model of acid-induced acute pancreatitis and reduces the infiltration of neutrophils in the lungs of treated rabbits.
13. A pharmaceutical composition, characterized in that it comprises a substance or polypeptide according to any of claims 4-12.
14. The use of a substance or polypeptide according to any of claims 4-12 for the treatment or prophylaxis of one or more of the diseases mentioned in Tables 1 and 2.
15. The use of a substance or polypeptide according to claim 4-12 for the manufacture of a pharmaceutical composition with the treatment or prophylaxis of one or more of the diseases mentioned in Tables 1 and 2.
16. A method for treating and / or preventing one or more of the diseases mentioned in Tables 1 and 2, the method is characterized in that it comprises administering to a patient in need thereof, a therapeutically or prophylactically effective amount in a substance or polypeptide in accordance with any of claims 4-12.
17. The synthesis of a substance or peptide according to any of claims 4-12, characterized in that it is by the use of solid phase peptide synthesis (SPPS), the process comprising the following steps: a) covalently coupling the amino acid C-terminal in the form of a reactive derivative, optionally protected in the side chain, N-alpha-protected, either directly or by means of a linker ^ suitable for a solid support, b) remove the N-alpha-protecting group , c) add the protected, subsequent amino acids according to the desired sequence in a gradual manner, d) remove the side chain protective groups, if any, e) in the assembly of the complete chain of peptides, cleave the peptide of the resin, and optionally f) cyclizing and / or stabilizing the peptide and / or acylating the amino-terminal amino acid residue and / or amidating the carboxy-terminal amino acid residue. SUMMARY OF THE INVENTION The invention relates to the use of a substance or polypeptide according to the formula X? -X2-X3-Thr-X4-Lys-X5-Arg-X6 (SEQ ID NO: 22), wherein Xi is Ala or Gly, X2 is Tyr or Phe, X3 / X4 and X5 are independently selected from the group consisting of Met, Lie, Leu and Val; and X6 is selected from the group consisting of Asn, Asp, Gln and Glu, optionally at least one of Xi, X2, X3, X4 ^ Xs and β is independently substituted with an unnatural or unusual amino acid and / or the peptide this cyclized and / or the peptide is stabilized and / or the amino-terminal amino acid residue is acylated and / or the carboxy-terminal amino acid residue is amidated and the peptidomimetics modeled on the basis of the above formula for the preparation of a pharmaceutical composition for the reduction of the production of TNF-a and / or for the prophylaxis or treatment of pancreatitis and / or for the prophylaxis or treatment of viral infections such as the acquired immunodeficiency syndrome (AIDS) or cutaneous HPV infection. In particular, the invention relates to peptide analogues of the above formula, wherein at least one of Xi, X2, X3, X4, X5 and Xs is independently substituted with an unnatural or unusual amino acid and / or the peptide is cyclized and or the peptide is stabilized and / or an amino acid residue of the terminal amino acid is acylated and / or the carboxy-terminal peptidomimetics are aminated, and modeled on the basis of the formula previous.
MXPA/A/1998/005648A 1996-01-18 1998-07-13 Synthetic il-10 analogues MXPA98005648A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCPCT/DK1996/000029 1996-01-18

Publications (1)

Publication Number Publication Date
MXPA98005648A true MXPA98005648A (en) 2001-09-07

Family

ID=

Similar Documents

Publication Publication Date Title
AU718476B2 (en) Synthetic IL-10 analogues
US6168791B1 (en) Antibodies that bind immunomodulators
CA2781173C (en) Immunomodulatory polypeptides derived from il-2 and their use thereof in the therapeutic of cancer and chronic infections
AU703371B2 (en) Enhancement of xenograft tolerance and porcine cytokines therefor
RU98115293A (en) SYNTHETIC ANALOGUES IL-10
JP2008500289A (en) Treatment of inflammatory airway diseases
Rhyne et al. Characterization of the human interleukin 1 receptor on human polymorphonuclear leukocytes
Schmidt et al. Interleukin-1
Lipnick et al. Immune abnormalities in the pathogenesis of juvenile rheumatoid arthritis
MXPA98005648A (en) Synthetic il-10 analogues
JP2818834B2 (en) IL-1α stabilized pharmaceutical preparation
Hofstetter et al. Human FcεRII. Molecular, biological and clinical aspects
Boraschi et al. Structure-function relationship of interleukin-1 giving new insights for its therapeutic potential
CN116615221A (en) Antagonistic peptides targeting IL-2, IL-9 and IL-15 signaling for the treatment of cytokine release syndrome and cytokine storm related disorders
WO1993020102A1 (en) Peptide t and related peptides in the treatment of inflammation, including multiple sclerosis
Boger et al. Immunomodulatory approaches to the treatment of inflammation
Reddy Recombinant bovine interleukin-1beta and interleukin-2 mediated immunomodulation in cattle
JPH06506918A (en) Peptide MB-4 promotes interleukin-1 production and modulates age-related immune responses