MOLECULES INHIBITING A METABOLIC PATHWAY INVOLVING THE SYK PROTEIN TYROSINE KINASE AND METHOD FOR IDENTIFYING SAID MOLECULES

The present invention relates to the C-13 molecule (methyl 2-{5-[(3-benzyl-4-oxo-2-thioxo-1,3-thiazolidin-5-ylidene)methyl]-2-furyl}-benzoate) and to organic molecules functionally equivalent to the C-13 molecule, capable of inhibiting the binding of an antibody or antibody fragment with the human Syk protein tyrosine kinase, to the use of these molecules for the production of medicaments for the prevention or treatment of diseases dependent on metabolic pathways involving Syk, and also to a method for identifying such molecules.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

The present invention relates to organic molecules capable of inhibiting the binding of an antibody or antibody fragment with human Syk tyrosine kinase protein, the use of said molecules for producing medicinal products for the prevention and treatment of conditions dependent on metabolic pathways involving Syk, and a method for identifying such molecules.

Syk (“Spleen tyrosine kinase”) Tyrosine Kinase protein (PKT) is a cytoplasmic protein which is a key mediator in immunoreceptor-dependent signalling in the cells involved in inflammation such as B lymphocytes, mast cells, macrophages and neutrophils. In mast cells and basophils, cross-linking of the FcεRI receptor (receptor with high affinity for immunoglobulin E) with IgE and antigens induces phosphorylation of FcεRI ITAM (“Immunoreceptor Tyrosine-based Activation Motif”) motifs so as to form a binding site for Syk which is then activated. The activated Syk protein in turn phosphorylates numerous substrates, including LAT (“linker for activation of T cells”), SLP-76 (“Src homology 2 (SH2) domain-containing leukocyte protein of 76 kD”) and Vav adapter proteins, resulting in the activation of a plurality of signalling cascades, such as those of PLC-γ (phospholipase Cγ), PI3K (“phosphatidylinositol 3-kinase”), Erk (“extracellular signal-regulated kinase”), JNK (“c-jun N-terminal kinase”) and p38 (see FIG. 9). These cascades eventually give rise to the degranulation, synthesis and release of lipid mediators and the production and secretion of cytokines, chemokines and growth factors by mast cells and basophils1,2.

Syk protein is thus recognised as a potential pharmaceutical target, particularly for the treatment of type I hypersensitivity reactions including allergic rhinitis, urticaria, asthma and anaphylaxis due to its critical position upstream from immunoreceptor signalling complexes regulating the inflammatory response in leukocytes. The fact that Syk regulates FcεRI signalling positively3, particularly suggests that it could be an excellent target for the treatment of allergic disorders. Furthermore, due to the central role thereof in FcεRI-dependent signalling, interacting pharmaceutically with Syk could prove to be more advantageous than the conventional use of antihistamines or leukotriene receptor agonists inhibiting a single step downstream from the complex cascades contributing to the acute and chronic symptoms associated with allergic conditions.

Pharmacological inhibitors of Syk kinase activity having a therapeutic potential, such as, in particular, Syk-specific anti-sense oligonucleotides in the form of aerosols or small molecules interfering with Syk activity such as ER-27139, BAY-613606, piceatannol and R112 have already been developed1, 4. However, if multiple types of cells expressing Syk are considered, potential side effects associated with systemic exposure of the immune system to medicinal products targeting the Syk kinase domain need to be taken into consideration. Indeed, Syk protein is widely distributed in various cell types, it is thus essential to account for the adverse effects of the inhibition thereof on varied physiological functions such as cell differentiation, adhesion and proliferation5, 6.

The inventors identified Syk protein inhibitors which act by preventing the interaction thereof with the natural cellular partners thereof rather than by targeting the catalytic site thereof, particularly compound C-13 and compounds 1 to 87 given in table 1.

The present invention relates to the molecule C-13 (methyl 2-{5-[(3-benzyl-4-oxo-2-thioxo-1,3-thiazolidin-5-ylidene)methyl]-2-furyl}benzoate) having the formula

as a medicinal product for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk in humans or animals.

The present invention further relates to functionally equivalent organic molecules to molecule C-13, binding with Syk tyrosine kinase protein and particularly molecules capable of inhibiting by at least 5%, preferably at least 10%, for example at least 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80 or 85% in vitro binding of

    • (i) antibody fragment G4G11 (SEQ ID No. 2), or
    • (ii) antibody fragment G4E4 (SEQ ID No. 3), or
    • (iii) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein on an epitope comprising at least one of residues 65 to 74 of the amino acid sequence of human Syk tyrosine kinase protein represented by the sequence SEQ ID No. 1, or
    • (iv) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein and inhibiting by at least 10% the binding of antibody fragments G4G11 or G4E4 with human Syk tyrosine kinase protein (SEQ ID No. 1)
      with human Syk tyrosine kinase protein or with any of the variants thereof in animals, for example murine Syk tyrosine kinase protein wherein the sequence is illustrated in FIG. 8B (SEQ ID No. 4), as a medicinal product for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk in humans or animals.

The term “functionally equivalent molecule” refers to a molecule, for example an organic molecule having a molecular weight between 50 and 2500 Da, capable of resulting in the same effect in vitro in an intra or extracellular medium or in vivo, optionally with a different intensity, than a given molecule. In particular, within the scope of the present invention, the term “functionally equivalent molecule to molecule C-13” refers to a molecule capable of producing the same effect in vitro in an intra or extracellular medium or in vivo, optionally with a different intensity, on human tyrosine kinase protein as represented by sequence SEQ ID No. 1 (FIG. 8A) as molecule C-13. In particular, it refers to molecules inhibiting the binding of Syk with another protein produced on the Syk region comprising the SH2 domains thereof. More specifically, these molecules do not affect the kinase enzyme activity of Syk. For example, this consists of molecules capable of inhibiting the interaction of Syk tyrosine kinase protein with antibody fragment G4G11 (SEQ ID No. 2), antibody fragment G4E4 (SEQ ID No. 3), or an antibody or antibody fragment binding with the same epitope as antibody fragment G4G11 or G4E4 on human Syk protein (SEQ ID No. 1).

The term “percentage of inhibition” of the binding of an antibody or antibody fragment with Syk protein, particularly refers to the ratio [(A−B)/(A×100)], where A consists of the intensity of a signal proportional to the quantity of an antibody or antibody fragment bound with Syk protein in the absence of a molecule according to the invention and B the intensity of the same signal in the presence of a molecule according to the invention under the same conditions. The inhibition of the binding of an antibody or antibody fragments with Syk protein may particularly be demonstrated in vitro by an antibody displacement test based on the ELISA technique as described for example in international application WO 2005106481. This test may be performed for example according to the protocol described in example 3-2) hereinafter.

The term “Syk variants in animals” refers to the genes of various animal species, for example mouse, rat, dog, cat or another mammal, coding for a protein having a strong sequence homology or identity with human Syk protein as represented by the sequence SEQ ID No. 1 (see FIG. 8A), for example a protein having at least 70, 75, 80, 85, 90 or 95% sequence homology or identity with the sequence SEQ ID No. 1 of human Syk protein, having the same tyrosine kinase activity and involved in the same functional cascades as same, particularly in the functional cascade giving rise to mast cell degranulation. It may particularly refer to orthologous genes, i.e. genes found in different organisms, having evolved from the same ancestral gene following speciation events.

The present invention also relates to the pharmaceutically acceptable salts, and if applicable, stereoisomers and racemates of C-13 or of equivalent molecules according to the invention.

The term “pharmaceutically acceptable salts” refers to relatively non-toxic inorganic and organic acid or basic addition salts preserving the biological activity of the molecules according to the invention. Examples of pharmaceutically acceptable salts are particularly described in S. M. Berge et al., “Pharmaceutical Salts”, J. Pharm. Sci, 1977, 66: p. 1-1940. The pharmaceutically acceptable addition salts of molecules according to the invention may for example be hydrobromide, hydrochloride, sulphate, bisulphate, phosphate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptanate, lactobionate, sulphamate, malonate, salicylate, propionate, methylenebis-b-hydroxynaphthoate, gentisic acid, isethionate, di-p-toluoyltartrate, methanesulphonate, ethane-sulphonate, benzenesulphonate, p-toluenesulphonate, cyclohexyl sulphamate and quinateslaurylsulphonate salts, and equivalents. Other pharmaceutically acceptable salts which may be suitable include metal salts, for example pharmaceutically acceptable alkaline metal or alkaline-earth salts, such as sodium, potassium, calcium or magnesium salts.

These pharmaceutically acceptable salts may be prepared in situ during the final molecule isolation and purification. Alternatively, the acid or basic addition salts may be prepared by reacting the purified molecule separately in the acid or basic form thereof with a base or an organic or inorganic acid and by isolating the salt formed. For example, a pharmaceutically acceptable acid addition salt may be prepared by reacting a molecule according to the invention with a suitable organic or inorganic acid (such as for example hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinic, maleic, formic, acetic, propionic, fumaric, citric, tartaric, lactic, benzoic, salicylic, glutamic, aspartic, p-toluene-sulphonic, benzene-sulphonic, methane-sulphonic, ethane-sulphonic, hexanoic or naphthalene-sulphonic acids such as 2-naphthalene sulphonic acid), optionally in a suitable solvent such as an organic solvent. A basic addition salt may, when a suitable acid group is present, be prepared by reacting a molecule according to the invention with a suitable organic or inorganic base (for example triethyl-amine, ethanol-amine, triethanol-amine, choline, arginine, lysine or histidine), optionally in a suitable solvent such as an organic solvent. The salts thus generated may then be isolated by means of crystallisation and filtration.

In some embodiments, pharmaceutically acceptable salts are preferred in that they provide the molecules according to the invention with superior stability or solubility, facilitating the formulation thereof.

According to one particularly preferred embodiment, the organic molecules according to the invention bind with Syk tyrosine kinase protein at a site located outside the catalytic domain thereof.

Also preferably, the organic molecules according to the invention have a molecular weight between 50 and 2500 Dalton, for example between 50 and 2000 Da, between 50 and 1500 Da or between 50 and 1000 Da.

According to one particular embodiment, the molecules according to the invention are capable of inhibiting, by at least 5%, preferably by at least 10%, the binding of an antibody or antibody fragment binding with human Syk tyrosine kinase protein on an epitope comprising at least two, preferably at least 3, 4 or 5, for example 5, 6, 7, 8, 9 or 10 of residues 65 to 74 of the amino acid sequence of human Syk tyrosine kinase protein (SEQ ID No. 1), with human Syk tyrosine kinase. According to one preferred embodiment, the antibody or antibody fragment binds with human Syk tyrosine kinase protein on the same epitope as antibody fragment G4G11 or antibody fragment G4E4 on human Syk tyrosine kinase protein, the sequence of which is illustrated by SEQ ID No. 1.

According to a further particular embodiment, the molecules according to the invention are capable of inhibiting, by at least 5%, preferably by at least 10%, the binding of an antibody or antibody fragment which binds to with Syk tyrosine kinase protein and inhibits, by at least 15%, preferably by at least 20, 30, 40, 50, 60, 70 or 80%, the binding of antibody fragments G4G11 or G4E4 with human Syk tyrosine kinase protein (SEQ ID No. 1), with human Syk tyrosine kinase protein.

Preferably, the molecules according to the invention bind with human Syk protein on a three-dimensional cavity comprising the Arginine residue situated in position 68 and the two glutamic acid residues situated in positions 121 and 155 of the Syk protein, the sequence of which is illustrated by SEQ ID No. 1. Preferably still, the three-dimensional cavity further comprises the Serine residue situated in position 9, the Glutamine residue situated in position 43, the Phenylalanine residue situated in position 51, the Isoleucine residue situated in position 66, the Glutamate residues situated in position 67 and 69, the Leucine residue situated in position 70, the Asparagine residue situated in position 71, the Glycine residue situated in position 72, the Threonine residue situated in position 73, the Tyrosine residue situated in position 74 and the Alanine residue situated in position 75 of human Syk protein, the sequence of which is illustrated by SEQ ID No. 1.

More preferably, the in vitro affinity, measured by the dissociation constant (or Kd), of the molecules according to the invention for Syk protein, is less than 100 more preferably, less than 50 and particularly preferably, less than 25 μM. The affinity of the molecules according to the invention for Syk protein is, for example, between 0.01 and 100 μM, between 0.1 and 50 μM or between 0.5 and 25 μM. The dissociation constant of the molecules according to the invention with respect to Syk protein may particularly be measured in vitro by means of fluorescence spectroscopy (or spectrofluorometry).

The present invention further relates to the use of a molecule according to the invention for producing a medicinal product for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk in humans or animals.

According to one particularly preferred embodiment, the molecules or salts according to the invention are used for producing a medicinal product for the prevention or treatment of type I hypersensitivity reactions.

The term “hypersensitivity” refers to an unsuitable or excessive immune response to an allergen, for example pollen, dust, animal hairs or certain foods, with effects ranging from moderate allergic reaction (skin rash, rhinitis, conjunctivitis, etc.) to severe systemic reactions potentially resulting in anaphylactic shock and potentially life-threatening in some cases. Immediate and delayed hypersensitivity reactions are classified in types I and IV respectively of the classification defined by Gell and Coombs (Gell P G H, Coombs R R A, eds. Clinical Aspects of Immunology. 1st ed. Oxford, England: Blackwell; 196339). According to this classification, “type I (or atopic or anaphylactic) hypersensitivity” is an immediate allergic reaction associated with exposure to a specific antigen or allergen, for example by swallowing, inhalation, injection or direct contact, and the triggering of immunoglobulin E (IgE) secretion by plasma cells. The IgE binds with the Fc receptors found on the surface of tissue mast cells and blood basophils. Subsequent exposure to the sensitised mast cells and basophils to the same allergen gives rise to the degranulation of the cells having the corresponding IgE and the release of mediators such as histamine, leukotriene or prostaglandins acting on the surrounding tissues, particularly giving rise to vasodilation and smooth muscle contraction. The reactions may be local or systemic and the symptoms vary from moderate irritation to sudden death due to anaphylactic shock. Examples of conditions caused by type I hypersensitivity include allergic asthma, allergic conjunctivitis, allergic rhinitis (hay fever), anaphylaxis, angioedema, urticaria, eosinophilia, allergies to antibiotics such as penicillin or cephalosporin. “Type II hypersensitivity” or “antibody-dependent immune response” is a reaction generally requiring from a few hours to one day, associated with interactions between antibodies (IgG, IgM) and an antigen on the surface of the cells of the patient carrying this antigen, giving rise to the destruction of these cells and the proliferation of B lymphocytes, producing antibodies against the antigen. “Type III hypersensitivity” or “immune complex disease” is a reaction developing over a number of hours, days or weeks, associated with the presence of similar quantities of antibodies and antigens giving rise to the formation of immune complex not suitable for evacuation circulating in the vessels, the deposition thereof on the walls of said vessels and giving rise to local or systemic inflammatory responses. “Type IV hypersensitivity” or “cell-mediated immunity” or “delayed hypersensitivity reaction” is an immune reaction generally requiring two to three days to develop and not associated with an antibody response but with the formation of a complex between cells which express a major histocompatibility complex I or II antigen and T lymphocytes giving rise to the release of lymphokines and/or cytotoxicity mediated by T lymphocytes.

Preferably, the molecules or salts according to the invention are used for producing medicinal products for the prevention or treatment of type I hypersensitivity reactions which inhibit IgE-dependent mast cell degranulation. More preferably, the molecules according to the invention are capable of inhibiting by 50% in vitro mast cell degranulation, at a concentration (IC50) between 1 ng/ml and 1 mg/ml, for example at a concentration between 1 ng/ml and 500 μg/ml, between 1 ng/ml and 250 μg/ml, between 1 ng/ml and 100 μg/ml, between 1 ng/ml and 50 μg/ml, between 1 ng/ml and 10 μg/ml, between 1 ng/ml and 5 μg/ml or between 1 ng/ml and 2 μg/ml. Also preferably, a quantity between 1 nM and 1 mM, for example between 1 nM and 100 nM, between 10 nM and 100 nM or between 1 nM and 10 nM, of a molecule according to the invention is capable of inhibiting mast cell degranulation by 50% in vitro.

More preferably, the metabolic pathway involving Syk on which the molecules or salts according to the invention is a mast cell or basophil activation pathway.

More preferably, the condition on which the molecules or salts according to the invention act is allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia or an allergy to an antibiotic.

According to one preferred embodiment, the molecules or salts according to the invention have no effect on the metabolic pathways involving human Syk protein (SECT ID No. 1) other than those giving rise to mast cell degranulation and/or type I hypersensitivity reactions. More preferably, the molecules or salts according to the invention have no effect on the antibody response following immunisation by a thymus-dependent antigen or on Syk-dependent neutrophil recruitment.

Syk tyrosine kinase protein is also found on the surface of B lymphocytes, T lymphocytes, neutrophils, eosinophils, NK cells, platelets, erythrocytes, osteoclasts, epithelial cells or cancer cells. According to one alternative embodiment, the metabolic pathway involving Syk on which the molecules or salts according to the invention act is a B lymphocyte, T lymphocyte, neutrophil, eosinophil, NK cell, platelet, erythrocyte, osteoclast, epithelial cell or cancer cell activation pathway. According to this embodiment, the condition on which the molecules or salts according to the invention act may thus be rheumatoid arthritis, an autoimmune disease, inflammation or cancer.

According to one particular embodiment, the molecules or salts according to the invention may be used in combination with another therapeutic molecule. For example, it may consist of a therapeutic molecule also used for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk or, on the other hand, a therapeutic molecule used for the prevention or treatment of a condition not dependent on a metabolic pathway involving Syk. According to one preferred embodiment, the molecules or salts according to the invention are used in combination with a molecule used for the treatment of allergy or type I hypersensitivity or for the treatment of the symptoms associated therewith. More preferably, the molecules or salts according to the invention are used in combination with epinephrine (or adrenaline), an H1 antihistamine, for example diphenhydramine, meclizine, fluphenazine, perphenazine, prochlorperazine, trifluoperazine, acrivastine, astemizole, cetirizine, levocetirizine, fexofenadine, loratadine, desloratadine, mizolastine, azelastine, levocabastine, olopatadine, cromoglicate, nedocromil, a non-steroidal anti-inflammatory drug (NSAID) or a steroidal anti-inflammatory drug, for example cortisone, hydrocortisone (or cortisol), cortisone acetate, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone, deoxycorticosterone acetate or aldosterone. According to a further preferred embodiment, the molecules or salts according to the invention are used in combination with an allergic desensitisation (or anti-allergic vaccination), i.e. a treatment based on regular increasing doses of an allergen. According to one particular embodiment, the molecule according to the invention is not used in combination with a glucocorticoid receptor agonist.

The molecules or salts according to the invention may be administered by any administration route, particularly by the oral, sublingual, nasal, ocular, local, intravenous, intraperitoneal, subcutaneous routes, by aerosol or by inhalation.

The molecules or salts according to the invention may particularly be administered to adult, child or newborn human patients. The molecules or sales according to the invention may also be administered to animal patients, particularly mammals such as dogs, cats, rats, mice.

In particular, the molecules or salts according to the invention are administered to a human patient at doses determined particularly on the basis of the patient's condition, medical history and age, for example doses between 0.1 mg/kg and 200 mg/kg.

The present invention also relates to a therapeutic method for the treatment or prevention of a condition dependent on a metabolic pathway involving Syk in a human or animal patient comprising the administration of a molecule according to the invention to the patient at doses, intervals and periods determined particularly on the basis of the patients condition, medical history and age.

According to one particularly preferred embodiment, the molecules according to the invention are selected from all the molecules consisting of C-13, molecules No. 1 to 87 given in table No. 1 and the molecules having any of the following formulas (I), (II), (III) or (IV):

where

    • R1 is an optionally substituted aromatic group, or an optionally substituted heterocycle comprising at least one S, O or N atom;

R2 is an optionally substituted aromatic group, an optionally substituted heterocycle, an optionally saturated carbon chain, comprising an amine group, an optionally saturated carbon chain comprising an optionally substituted aromatic group or an optionally saturated carbon chain comprising an optionally substituted heterocycle comprising at least one S, O or N atom;

R3 is an optionally substituted phenyl, 2-pyridinyl, 3-pyridinyl or 4-pyridinyl group;

where

    • n=0 or 1; n′=0 or 1;
    • R4 is an optionally saturated carbon chain comprising 1 to 5 carbon atoms, optionally substituted with an aromatic group;
    • R5 is an optionally substituted aromatic group or an optionally substituted amine group;
    • R6 is a hydrogen atom, alkoxy group, alkyl group or halogen;
    • R7 is a hydrogen atom, alkoxy group, alkyl group or halogen;
    • R8 is a hydrogen atom, alkoxy group, alkyl group or halogen;

where

    • m=0, 1 or 2;
    • R9 is a hydrogen atom and R10 is an optionally substituted phenyl group, or R9 and R10 are part of the same optionally substituted heterocycle, or R9 and R10 are part of the same optionally substituted aromatic group;
    • R11 is a hydrogen atom, alkoxy group or alkyl group;
    • R12 is a hydrogen atom, alkoxy group or alkyl group;
    • R13 is a hydrogen atom or an alkyl or alkoxy group;
    • R14 is a hydrogen atom or an alkyl or alkoxy group;

where

    • A is an oxygen or sulphur atom;
    • R15 is an optionally saturated carbon chain comprising 1, 2 or 3 carbon atoms, optionally substituted by an optionally substituted aromatic group, an optionally substituted heterocycle or an amine group belonging to optionally substituted heterocycle;
    • R16 is a hydrogen atom, halogen or alkoxy group;
    • R17 is a hydrogen atom, alkoxy group or acetoxy group.

According to one particular embodiment, the group R1 of molecules having formula (I) is selected from the following groups:

    • a phenyl group, optionally substituted by an F or Cl atom, a methyl or ethyl group, an N,N-dimethyl-sulphonamide or two groups selected from the methyl, ethyl, hydroxy, methoxy or ethoxy groups,
    • a group

    • a furan group optionally substituted by a methyl, ethyl, hydroxyl, methoxy or ethoxy group,
    • a thiophene group optionally substituted by a methyl, ethyl, hydroxy, methoxy or ethoxy group;
      the group R2 of molecules having formula (I) is selected from the following groups:
    • a group

    • where R21 and R22 are carbon atoms each belonging to an alkyl chain comprising 1, 2 or 3 carbon atoms, or both belonging with the nitrogen atom with which they are bound to the same optionally saturated heterocycle also comprising an oxygen atom or a second nitrogen atom,
    • a or a group

    • where R23 and R24 are carbon atoms each belonging to an alkyl chain comprising 1, 2 or 3 carbon atoms, or both belonging with the nitrogen atom with which they are bound to the same optionally saturated heterocycle also comprising an oxygen atom or a second nitrogen atom,
    • or a group

and the group R3 of molecules having formulas (I) is selected from the following groups:

    • a non-substituted 2-pyridinyl, 3-pyridinyl or 4-pyridinyl group,
    • a phenyl group optionally substituted by a benzoxy group, and/or by a hydroxyl group, and/or by a methyl group, and/or by an ethyl group, and/or by a propyl group, and/or by one or two Br, F or Cl atoms, and/or by one to three hydroxyl, methoxy or ethoxy groups.

According to one particular embodiment, when the group R3 of molecules having formula (I) is a phenyl group, the group R2 of molecules having formula (I) is not an aromatic group or a heterocycle.

According to one particular embodiment, the group R4 of molecules having formula (II) is an optionally saturated carbon chain comprising 1, 2 or 3 carbon atoms; the group R5 is a phenyl group or a secondary amine grow substituted by an optionally substituted phenyl group, or by a group

the group R6 of molecules having formula (II) is a hydrogen or chlorine atom or a methyl, ethyl, hydroxyl, methoxy or ethoxy group; the group R7 of molecules having formula (II) is a hydrogen or chlorine atom or a methyl, ethyl, hydroxy, methoxy or ethoxy group; and the group R8 of molecules having formula (II) is a hydrogen or chlorine atom or a methyl, ethyl, hydroxy, methoxy or ethoxy group.

According to one particular embodiment,

    • the group R9 of molecules having formula (III) is a hydrogen atom and the group R10 is an optionally substituted phenyl group, or the groups R9 and R10 belong to the same optionally substituted heterocycle comprising 2 nitrogen atoms and 4 carbon atoms;
    • the group R11 of molecules having formula (III) is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group;
    • the group R12 of molecules having formula (III) is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group;
    • the group R13 of molecules having formula (III) is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group;
    • and the group R14 of molecules having formula (III) is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group.

According to a further particular embodiment, the group R15 of molecules having formula (IV) is a group

the group R16 of molecules having formula (IV) is a hydrogen or chlorine atom or a methyl, ethyl, hydroxy, methoxy or ethoxy group and the group R17 of molecules having formula (IV) is a methyl, ethyl, hydroxy, methoxy, ethoxy, acetoxy, methoxycarbonyl or ethoxycarbonyl group.

According to one particular embodiment, the molecule according to the invention is not

or if it is any of these molecules, it is not used in combination with a glucocorticoid agonist.

The term carbon chain refers to an organic chain having a linear or cyclic, optionally branched, chain formation of adjacent carbon atoms, connected by covalent bonds, as the network thereof. A carbon chain according to the present invention may for example be a linear chain formation of one to twenty, preferably 1 to 12, 1 to 10, 1 to 6, 1 to 5 or 1 to 4 carbon atoms. In particular, it may consist of an alkyl group, i.e. derived from an alkane (linear or branched saturated hydrocarbon molecule) due to the loss of a hydrogen atom, for example a methyl group, an ethyl group, a linear or branched propyl group or a linear or branched butyl group or a linear or branched unsaturated hydrocarbon chain, for example an ethenyl or ethynyl group. It is understood that the electrons of the outer layer (4 in number) of each carbon atom forming the carbon chain network are not involved in a covalent bond with a further carbon atom or with a heteroatom are involved in a covalent bond with a hydrogen atom.

The term heteroatom refers to a non-metallic atom other than carbon or hydrogen, for example oxygen, nitrogen, sulphur, phosphorus or halogens.

The term aromatic or aryl group refers to an unsaturated cycle system observing Hackers aromaticity rule. For example, it may consist of a phenyl group (group derived from a benzene nucleus).

The term heterocycle refers to a cycle system wherein one or a plurality of carbon atoms is replaced by a heteroatom such as, for example, oxygen, nitrogen or sulphur. It may in particular consist of aromatic heterocycles, such as pyrrole, thiophene, furan and pyridine or of saturated heterocycles, such as sugars, or uses. For example, a heterocycle according to the invention comprises 2 to 8 carbon atoms and 1 to 4 heteroatoms, preferably it comprises 2, 3, 4 or 5 carbon atoms and 1, 2, 3 or 4 heteroatoms.

Each atom belonging to a carbon chain, aromatic group, cycle or heterocycle according to the present invention may be substituted via a covalent bond by one or a plurality of halogens, for example Fluorine, Chlorine, Iodine or Bromine, and/or by one or a plurality of organic groups, for example one or a plurality of aromatic (such as a phenyl group), cyclic, heterocyclic (such as a furan or thiophene group), alkyl (such as a methyl group, ethyl group, linear or branched propyl group or a linear or branched butyl group), alkoxy (such as a methoxy (OCH3) or ethoxy (OCH2CH3) group), carboxyl (such as a carboxy (COON) group), carbonyl (such as an acetoxy (OCOCH3) or methoxycarbonyl (COOCH3) or ethoxycarbonyl (COOCH2CH3) group), primary, secondary or tertiary amine, amide (such as an acetamide group) or sulphonamide (such as an N,N-dimethyl-sulphonamide group) groups. It is understood that a saturated, unsaturated or aromatic cycle or heterocycle may be merged with a further cycle, for example by means of a single or double bond between two carbon atoms.

The present invention also relates to a pharmaceutical composition comprising a molecule according to the invention and a pharmacologically acceptable excipient.

According to one embodiment, the pharmaceutical composition according to the invention also comprises a further therapeutic molecule. For example, it may consist of a therapeutic molecule also used for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk or, on the other hand, a therapeutic molecule used for the prevention or treatment of a condition not dependent on a metabolic pathway involving Syk. According to one particular embodiment, the pharmaceutical composition according to the invention does not comprise a glucocorticoid receptor agonist.

According to a further particular embodiment, the pharmaceutical composition according to the invention may be used in combination with one or a plurality of further pharmaceutical compositions. For example, it may be pharmaceutical compositions also used for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk or, on the other hand, pharmaceutical compositions used for the prevention or treatment of a condition not dependent on a metabolic pathway involving Syk. According to this embodiment, the pharmaceutical composition according to the invention and the further pharmaceutical composition(s) may be administered simultaneously or in alternation, by the same administration route or by different routes. According to one particular embodiment, the pharmaceutical composition according to the invention is not used in combination with a glucocorticoid receptor agonist.

The present invention also relates to a method for identifying an organic molecule having a molecular weight between 50 and 2500 Dalton binding with Syk tyrosine kinase protein and capable of inhibiting by at least 5%, preferably at least 10%, for example at least 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80 or 85% in vitro the binding of (i) antibody fragment G4G11 (SEQ ID No. 2), or (ii) antibody fragment G4E4 (SEQ ID No. 3), or (iii) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein on an epitope comprising at least one of residues 65 to 74 of the amino acid sequence of human Syk tyrosine kinase protein represented by the sequence SEQ ID No. 1, or (iv) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein and inhibits by at least 10% the binding of antibody fragments G4G11 or G4E4 with human Syk tyrosine kinase protein (SEQ ID No. 1), to human Syk tyrosine kinase protein or to any of the variants thereof in animals, comprising at least the following steps:

    • a) screening, from a bank of candidate organic molecules having a molecular weight between 50 and 2500 Da, those liable to bind with Syk protein on the three-dimensional binding cavity on the Syk protein of a molecule selected from the molecules having formula C-13, I, II, III, IV or 1 to 87 as illustrated above;
    • b) selecting from the molecules identified in a) those capable of inhibiting by at least 5%, preferably at least 10%, for example at least 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80 or 85% in vitro the binding of the antibody or antibody fragment (i), (ii), (iii) or (iv) with Syk protein.

According to one particularly preferred embodiment, the molecule from step a) selected from the molecules having formula C-13, I, II, III, IV or 1 to 87 is the molecule C-13, molecule (59) or molecule (61).

According to one preferred embodiment, the method according to the invention comprises an additional step prior to step a) for identifying the three-dimensional binding cavity on the Syk protein of the molecule selected from the molecules having the formula C-13, I, II, III, IV or 1 to 87. This prior step may particularly be performed by means of “in silico docking”.

The term “in silico docking” or “molecular docking” or “virtual docking” refers to the use of a bioinformatics tool for predicting and modelling the position of a ligand in a macromolecule. In particular, in silico docking tools can be used to calculate the probability that a given chemical compound will be able to dock with an active target protein, for example on a previously identified three-dimensional binding cavity.

According to one preferred embodiment, the method according to the invention comprises an additional step c) for selecting from the molecules identified in b) those capable of inhibiting by 50% in vitro at mast cell degranulation to a concentration (IC50) between 1 ng/ml and 1 mg/ml, for example at a concentration between 1 ng/ml and 500 μg/ml, between 1 ng/ml and 250 μg/ml, between 1 ng/ml and 100 μg/ml, between 1 ng/ml and 50 μg/ml, between 1 ng/ml and 10 μg/ml, between 1 ng/ml and 5 μg/ml or between 1 ng/ml and 2 μg/ml.

According to one particularly preferred embodiment of the uses and methods described above, the molecule according to the invention is selected from the group consisting of the molecule C-13 and molecules No. 1 to 87 given in table 1.

These molecules were identified by the inventors within the scope of a project following a previous study (Dauvillier et al., 20027), during which they expressed scFv (“single chain variable domain”) (or “intracellular antibodies” or “intrabodies”), G4G11 (SEC) ID No. 2) and G4E4 (SEC) ID No. 3) antibody fragments in a mast cell line. This study demonstrated the inhibitory effects of these “intracellular antibodies” or “intrabodies” on the release of allergic mediators induced by FcεRI stimulation on the mast cell membrane. The scFv G4G11 and G4E4 antibody fragments were isolated from a combinatory bank screened against a recombinant protein containing the SH2 domains of Syk and inter-domain A region separating same, i.e. a portion of Syk protein not comprising the Syk kinase domain8.

The ADA (“antibody displacement assay”) method is a method developed by the inventors and described in WO2005106481, particularly for identifying a ligand capable of selectively modulating a functional cascade involving a target, comprising a first step for identifying an intracellular antibody capable of binding with the target and modulating the functional cascade in question, a second step for screening from a bank of small organic molecules, ligands modulating the binding between the target and the intracellular antibody potentially being performed in vitro in an extracellular test, and a third step for identifying from the modulating ligands obtained in step 2, those capable of modulating the functional cascade in the cell.

The inventors suggested the theory whereby the antibody fragments G4G11 and G4E4 bind on a Syk region interacting with one or more essential partners in the functional cascade giving rise to degranulation. Taking into consideration the limits of the use of intracellular antibodies in therapy, such as the effective transfer of the gene encoding the antibody in target cells9, the inventors sought to isolate organic molecules acting as functional mimics of the intrabody G4G11 and suitable for easier use in therapy. To this end, they used the ADA method for screening a bank of 3000 small organic molecules and identifying potential allergic response inhibitors.

Among the 3000 small organic molecules tested, the inventors identified the small molecule C-13 (methyl 2-{5-[(3-benzyl-4-oxo-2-thioxo-1,3-thiazolidin-5-ylidene)methyl]-2-furyl}benzoate) and demonstrated the ability thereof to modulate the interaction of the antibody fragment G4G11 or G4E4 with Syk in vitro and inhibit mast cell degranulation induced by FcεRI in vitro.

The inventors particularly demonstrated the fact that the compound C-13 inhibits anaphylactic shock when administered orally and has promising anti-allergic properties, illustrating the strong therapeutic potential of medicinal product candidates isolated using the approach described herein.

The inventors also demonstrated the fact that C-13 binds with Syk on a newly identified cavity situated between both SH2 domains and inter-domain A of Syk (FIG. 1). The binding cavity of C-13 forms a unique interaction zone which is specific to Syk, and does not correspond to a known binding side of physiological ligands of Syk such as doubly phosphorylated ITAM peptide on tyrosine residues (FIG. 1A). The results obtained suggest that C-13 inhibits the interaction of Syk with some of the macromolecular substrates thereof, either directly in that C-13 occupies a surface whereon a partner of Syk could establish direct contact, and/or by means of an allosteric effect.

The biochemical studies conducted on mast cells indeed demonstrated that C-13 inhibits FcεR1-dependent phosphorylation of SLP-76 on tyrosine residues contributing to the adapting function thereof for the binding and/or stabilisation of Btk, PLC-γ and Vav with the macromolecular signalling complex formed with LAT22, 28-30 (FIG. 2). This affects the phosphorylation and catalytic activity of Btk and PLC-γ renewal in the vicinity of Syk and/or Btk for the complete phosphorylation thereof which is required for maintaining calcium flow and exocytosis31-35. Indeed, C-13 inhibited early (β-hexosaminidase release) and delayed (TNF-α secretion) mast cell responses induced by aggregation on the FcεRI receptors with an estimated IC50 of 2 μM (FIG. 3).

Significantly, the oral administration of a single dose of C-13 inhibited IgE-induced passive systemic anaphylaxis (PSA) with an estimated IC50 of 110 mg/kg (FIG. 4), confirming the promising anti-allergic properties of this compound. On the contrary, a single oral administration of 100 mg/kg of C-13 did not affect Syk-dependent neutrophil recruitment induced by thioglycollate in the peritoneal cavity in the presence of Bordetella pertussis toxin (FIG. 5A). Furthermore, the inventors demonstrated that, despite the fact that BCR-dependent B lymphocyte in vitro proliferation was inhibited in a dose-dependent fashion by C-13 (FIG. 5B), the antibody responses of mice immunised with a thymus-dependent antigen were not affected by the oral administration of 150 mg/kg of C-13 (FIG. 5C). Therefore, the molecule C-13 does not affect some responses dependent on Syk but not dependent on mast cells in vivo at administration doses and periods at which it is liable to inhibit a severe allergic response.

Taking into consideration the lack of an apparent toxic effect following the oral or local administration of C-13 over a period ranging from one hour to 12 days, C-13 may be considered as the potential first member of a new family of Syk inhibitors suitable for oral administration and pharmacologically active molecules having an anti-inflammatory effect. The pharmaceutical molecule screening approach described herein represents a generic platform wherein the initial use of antibodies makes it possible to detect the domains of the target molecule having a therapeutic potential, thus facilitating the design of chemical molecules (via in silico and/or in vitro screening) capable acting as functional antibody mimics and as potential protein-protein interaction inhibitors. Furthermore, the inventors demonstrated that these small molecules can induce the desired response in cell and animal models, supporting the concept in favour of the replacement of large macromolecules that are difficult to administer by small organic molecules suitable for oral administration.

The possible binding site with Syk was predicted in silico, guided by the location of the epitope of G4G11. One candidate cavity situated next to the epitope of G4G11, on the interface situated between the two SH2 domains and the inter-domain binder of Syk and comprising the residues Ser 9, Gln 43, Phe 51, Ile 66, Glu 67, Arg 68, Glu 69, Leu 70, Asn 71, Gly 72, Thr 73, Tyr 74, Ala 75, Glu 121 and Glu 155 was thus identified (FIG. 10). Targeted mutagenesis experiments confirmed that the residues Arg 68, Glu 121 and Glu 155 of human Syk protein (SEQ ID No, 1) play a significant role in interaction with C-13, the mutation of said residues suppressing the inhibition caused by C-13, whereas the binding of scFv G4G11 is maintained (FIG. 1A, 10). These results tend to confirm the theory whereby the binding cavity of C-13 or Syk is located in the vicinity of the binding site of the intrabody G4G11.

The inventors then performed virtual docking on a molecule bank to identify candidate molecules having the best binding properties on said three-dimension cavity, and tested the ability of said candidate molecules to inhibit the binding of scFv G4G11 with Syk. These molecules are given in table 1 (see example 2). The ability of these molecules to inhibit mast cell degranulation in vitro was also tested. With molecules No. 59 and 61 in particular the concentration inhibiting mast cell degranulation by 50% in vitro is in the region of 5 μM (see FIG. 6).

The results given in the experimental part and particularly in table 1 demonstrate that the molecules or salts according to the invention inhibit type I hypersensitivity reactions, particularly IgE-dependent mast cell degranulation, and are also capable of interfering in vivo with passive cutaneous and systemic anaphylaxis in BALB/c mice.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1. Binding of C-13 with Syk in vitro. A. Chemical structure of C-13. 3D structure of cavity predicted and validated for C-13 (or “Compound 13”) comprising the residues Ser 9, Gln 43, Phe 51, Arg 68, Glu 121 and Glu 155 (mesh representation); the G4G11 epitope comprises the residues 65 to 74 of human Syk protein represented by SEQ ID No. 1. B. Binding of scFv G4G11 fragment with Syk measured using ADA method in the presence of C-13 (▪). Binding of C-13 with Syk measured using fluorescence spectroscopy (o) (Kd=4.8±0.2 μM). C. Binding of G4G11 with Syk mutants with ADA method. Significant inhibition with C-13 versus DMF: **P<0.01.

FIG. 2. C-13 inhibits FcεRI-induced mast cell activation. A. The immunoblots produced on RBL-2H3 cell lysates were analysed with immunoblots with the specified antibodies. The in vitro kinase activities of the Syk, Btk and Lyn immunoblots were examined. B. The RBL-2H3 cell lysates and C. BMMCs were subjected to electrophoresis and the proteins were analysed by means of immunoblot. These data are representative of at least two experiments.

FIG. 3. C-13 inhibits FcεRI-dependent calcium release and degranulation. A. FACS analysis of IgE-dependent calcium flow in RBL-2H3 cells. B. Release of β-hexosaminidase in RBL-2H3 cells sensitised with IgE/DNP. C. Release of β-hexosaminidase and, D. titration of TNF-α in BMMCs cells sensitised with IgE/DNP; C-13: 3 μM. E. FACS analysis of FcεRI surface expression in RBL-2H3 cells. (0=0.25% DMF), Significant inhibition with C-13 versus DMF: **P<0.01 and *P<0.05.

FIG. 4. In vivo studies on BALB/c mice. A-C. PSA response. A. Temperature progression, B. Evans blue extravasation quantification, C Photograph representing Evans blue extravasation according to an oral administration of 130 mg/kg of C-13, a non-relevant compound (IR) or the vector (T); Untreated animal (NT). The ears and tail of the mice received the non-relevant compound (IR) or the vector alone (T) turned a pronounced blue colour, those of the mice treated with C-13 turned a light blue colour and those of the untreated mice are normal in colour D. PCA response: Evans blue extravasation quantification. Significant inhibition with C-13 versus a non-relevant compound: **P<0.01; versus the vehicle+DNP-KLH: *P<0.05.

FIG. 5. In vivo and in vitro effects of C-13 on neutrophils and B lymphocytes. A. Neutrophil recruitment in the peritoneal cavity after injecting thioglycollate or the vehicle in the presence of Bordetella Pertussis toxin (where specified) (n=4). B. In vitro B lymphocyte proliferation induced by anti-IgM. C. Serum immunoglobulin concentration 12 days after immunisation with TNP-KLH (n=4).

FIG. 6. Effect of molecules No. 59 (ref. Chembridge 7501888) and 61 (ref. Chembridge 7722851) on RBL-2H3 cell degranulation. The level of β-hexosaminidase release in RBL-2H3 cells sensitised with IgE/DNP was measured in the presence A. of molecule No. 59 and B. of Molecule No. 61 at concentrations ranging from 0 to 40 μM.

FIG. 7. Amino acid sequences of intrabodies scFv A. G4G11 (SEQ ID No. 2) and B. G4E4 (SEQ ID No. 3).

FIG. 8. Amino acid sequence of human A. (SEQ ID No. 1) corresponding to accession No. NP003168 (NCBI) and B. murine Syk protein corresponding to accession No. NP035648 (NCBI). The residues identified as belonging to the three-dimensional binding cavity of C-13 on human Syk protein (A) and the equivalent thereof on murine Syk protein (B) are shown in bold type and underlined.

FIG. 9. Schematic representation of the metabolic pathways involving Syk tyrosine kinase in mast cells.

FIG. 10. Toxicity test of C-13 on BMMC cells. The BMMC cells were incubated at 37° C. in the presence of 2.5 μM or 5 μM of C-13, 0.25% DMF or Staurosporine. The percentages of live BMMC cells after 3 hours and after 5 days were detected by means of double Annexin-V and Propidium Iodide labelling. The viability of BMMC cells incubated in the presence of 2.5 μM or 5 μM of C-13 or 0.25% DMF was not affected to noteworthy degree after 3 hours (78%, 74% and 79% respectively) or five days (62, 56 and 57%, respectively). On the other hand, the viability of BMMC cells treated under the same conditions with Staurosporine was reduced to a level of 16% after only 3 hours.

EXAMPLES Example 1 Identification of Molecule C-13 Potentially Suitable for Oral Administration to Prevent Anaphylactic Shock

1) Identification of Compound 13 (C-13) and the Binding Cavity thereof on Syk

The inventors developed the ADA (Antigen Displacement Assay) method to identify small molecules capable of displacing the association with scFv G4G11 with Syk. Of the members of a bank of 3000 chemical molecules, 15 small molecules proved to be capable of competing with the binding of scFv G4G11 with Syk, and of these compounds, that hereinafter referred to as C-13 (FIG. 1A) displayed the best inhibition potential with an estimated IC50 of 4 μM (FIG. 1B). This result is in line with the dissociation constant value (or Kd) equal to 4.8 μM obtained by measuring by means of fluorescence spectroscopy (or spectrofluorometry) the in vitro affinity of C-13 for Syk (FIG. 1B). To understand the mode of action of C-13, the inventors firstly identified the binding site of G4G11 using the SPOT method16. An epitope located on the N-terminal SH2 domain of Syk, comprising amino acids 65-74 and 100% preserved in mouse, rat and human sequences, was identified.

On the basis of this information and the known 3D structure of the peptide complex formed by the SH2 domains of Syk and ITAM motifs17, the inventors used computing approaches to locate a putative binding site for C-13. A candidate cavity comprising the residues Ser 9, Gln 43, Phe 51, Ile 66, Glu 67, Arg 68, Glu 69, Leu 70, Asn 71, Gly 72, Thr 73, Tyr 74, Ala 75, Glu 121 and Glu 155 of human Syk protein (see FIG. 8A, SEQ ID No. 1) and situated in the vicinity of the G4G11 epitope was identified (FIG. 1C). A structural analysis specified that the residues Ser 9, Gln 43, Phe 51, Arg 68, Glu 121 and Glu 155 could be involved in the binding of the ligand, and could be mutated without impairing the 3D structure of the protein. To validate the cavity in more detail, these six amino acids were mutated individually and the Syk mutants were subjected to the ADA test. The residues Arg 68, Glu 121 and Glu 155 proved to have a significant role in the interaction with the small molecule, given that the mutation thereof cancelled the inhibition caused by C-13, whereas the binding of scFv G4G11 was maintained (FIG. 10). These data confirmed the fact that the binding cavity of C-13 on Syk is located in the vicinity of the binding site of G4G11.

2) FcεRI-Induced Mast Cell Activation

To examine the functional similarities with G4G11, the inventors explored the biological effects of C-13 on mast cell activation. The incubation of RBL-2H3 cells with 0-13 did not affect the phosphorylation and FcεRI-induced kinase activity of Syk (FIG. 2A) and, accordingly, the overall level of tyrosine phosphorylation of all the cell proteins known as being essentially Syk-dependent was normal (FIG. 2B, C). Similarly to the intrabody G4G11, C-13 inhibited the phosphorylation and FcεRI-induced kinase activity of Btk (FIG. 2A) and the phosphorylation of PLC-γ1 and PLC-γ2, the two PLC-γ isoforms expressed in mast cells (FIG. 2A, C). PTK Lyn phosphorylates both Syk and Btk giving rise to the complete activation thereof and the subsequent phosphorylation of PLC-γ18. Given that C-13 did not affect FcεRI-dependent Lyn activation (FIG. 2A), it can be concluded that the reduction of the level of Btk and PLC-γ phosphorylation could be due to a defect with respect to the correct location thereof in the vicinity of the upstream PTK.

3) Fyn- and Lyn-Dependent Signalling Cascade Analysis

In mast cells, the signalling cascade Fyn/Gab2/PI3K gives rise to the activation of PI3K and the generation of PI-3,4,5-P3 recruiting a number of proteins containing a pleckstrin homology domain (PH), including Btk and PLC-γ on the plasma membrane19. The analysis of the phosphorylation of Akt, a PI3K activity marker, indicated that C-13 did not affect the Fyn-dependent cascade (FIG. 2B, C), suggesting that the reduced level of phosphorylation of Btk and PLC-γ was not due to a defect on the membrane location thereof, known as being an essential factor in the increase in calcium flows20.

Btk and PLC-γ recruitment on the membrane also requires the canonical signalling cascade Lyn/Syk/LAT/SLP-76. The phosphorylation of LAT by Syk gives rise to the translocation of SLP-76 to the complex organised by LAT21, where SLP-76 is co-located with Syk22. This location enables Syk to phosphorylate N-terminal tyrosines of SLP-7623 which become binding sites for Vav, Nck and Btk. LAT and SLP-76 (via the proline-rich domain thereof recruiting PLC-γ) interact to locate PLC-γ on said membrane complex, enabling the phosphorylation and activation of PLC-γ by Btk24 and/or Syk25. The use of phospho-specific antibodies demonstrated that C-13 inhibits the phosphorylation of SLP-76, but increases the phosphorylation of LAT in a dose-dependent fashion (FIG. 2A). The inventors suggested the theory whereby the inhibition of SLP-76 phosphorylation could enable a larger quantity of LAT to interact with Syk, thus causing an increase in the phosphorylation level thereof. These results demonstrate that the reduction in SLP-76 phosphorylation was not due to a defect in terms of the recruitment thereof to LAT, and resulted in a co-location defect of Btk and, to a lesser extent, that of Vav with SLP-76 (FIG. 2A). Nevertheless, Vav phosphorylation known to be independent from the recruitment thereof to SLP-7626 was not inhibited (FIG. 2A).

4) MAPK Activation

The association of SLP-76 with Vav and/or Nck plays a role in optimal MAP kinase activation in mast cells27. The inventors demonstrated that C-13 affects MAP kinase activation slightly (evaluated via the phosphorylation level thereof): a high C-13 concentration reduces the phosphorylation level ERK1/2, whereas the phosphorylation levels of p38 and JNK remain normal (FIG. 2B, C).

5) Calcium Flow and Degranulation

Binding of Btk and Vav with SLP-76 is critical for regulating PLC-γ activity on the membrane, calcium mobilisation and granule exocytosis27, 28. The inventors demonstrated that the association of PLC-γ with LAT was inhibited by C-13 in a dose-dependent fashion (FIG. 2A). Consistently with the defect in terms of PLC-γ1 and PLC-γ2 phosphorylation, the mast cells showed a reduced calcium flow range in response to FcεRI binding (FIG. 3A), and early and delayed FcεRI-induced allergic responses in BMMC (“bone marrow derived mast cell”) cells and in the RBL-2H3 cell line are also weakened in a dose-dependent fashion, based on the measurement of β-hexosaminidase release and TNF-α secretion (FIG. 3B, C, D). The results also demonstrated that C-13 had no toxic effect on mast cells. Indeed, ionomycin-induced degranulation (FIG. 3B), or BMMC cell viability (see FIG. 10) were not detectably affected by treatment with C-13. Furthermore, the defects observed in terms of mast cell activation are not due to a reduced level of FcεRI surface expression, flow cytometry analysis indicating that the cells incubated with C-13 express similar levels of FcεRI to those of control cells (FIG. 3E).

6) Passive Systemic (PSA) and Cutaneous Anaphylaxis (PCA)

Finally, to extend these observations to mast cell functions in vivo, the inventors tested the effects of C-13 on passive systemic (PSA) and cutaneous anaphylaxis (PCA) induced in BALB/c mice by administering DNP-specific IgE molecules followed by intravenous stimulation with DNP-KLH hapten. This mimics systemic anaphylaxis as demonstrated by the immediate cardiopulmonary changes and the increase in vascular permeability. The intensity of systemic anaphylaxis was determined by measuring both the drop in body temperature and the increase in vascular permeability following antigen administration. After the oral administration of C-13 (and prior to antigen stimulation), the animals appeared to be healthy with no obvious sign of toxicity. The administration of a single oral dose of 100 mg/kg of C-13 inhibited hypothermia and accelerated the recovery of the animals (FIG. 4A). On the basis of the Evans blue extravasation quantification, it was determined that C-13 inhibits the increase in vascular permeability with an estimated IC50 of 110 mg/kg (FIGS. 4B and 4C). C-13 also demonstrated an inhibitory effect on PCA with an estimated IC50 of 25 μM (FIG. 4D).

Example 2 Identification Using Binding Cavity of C-13 of Further Potential Mast Cell Degranulation Inhibitors

Using the binding cavity identified in example 1-1), virtual docking screening was conducted on a set of 350,000 molecules contained in the ChemBridge Corporation (San Diego, USA) chemical bank to identify the 1000 molecules displaying the best binding properties in said cavity.

The ADA method was then applied to each of these 1000 molecules to measure the ability thereof to inhibit the binding of scFv G4G11 with Syk. The 87 molecules having the best inhibition rate (between 11 and 86.5%) are given in table 1.

These 87 molecules were also tested in vitro to assess the ability thereof to inhibit RBL-2H3 cell degranulation (see table 1). The two molecules exhibiting the best potential (molecules No. 59 and 61) were tested at various concentrations on RBL-2H3 cells to assess the concentration inhibiting degranulation by 50% in more detail (see FIG. 6).

Finally, the in vitro affinity of the molecule C-13 and some of the 87 molecules mentioned above for Syk protein was measured by means of fluorescence spectroscopy (or spectrofluorometry) and is expressed by the dissociation constant (or Kd) in μmole/litre (μM).

TABLE 1 C-13 and the 87 molecules identified using C-13 and the antibody fragment G4G11 CB In vit. Degran. Structure ref. Name Rank inhib. IC50 Kd No. Gp 6197026 methyl 2-{5- [(3-benzyl-4- oxo-2-thioxo- 1,3- thiazolidin-5- ylidene) methyl]-2- furyl} benzoate 81% 1 μg/ml 4.8 μM C-13 6752784 4-(4-chloro benzoyl)-3- hydroxy-5-(3- phenoxy phenyl)-1-(3- pyridinyl methyl)-1,5- dihydro-2H- pyrrol-2-one 611 86.5 >10 μg/ml 5 μM  1 I 6670340 5-(2,4- dimethoxy phenyl)-3- hydroxy-1-[3- (1H-imidazol- 1-yl)propyl]-4- (2-thienyl carbonyl)-1,5- dihydro-2H- pyrrol-2-one 792 81 >10 μg/ml 16.3 μM  2 I 6422575 {4-bromo-2- [3-(ethoxy carbonyl)-2- méthyl-5-oxo- 4,5-dihydro- 1H- indeno[1,2- b]pyridin-4- yl]phenoxy} acetic acid 706 81 >10 μg/ml 6.2 μM  3 6882059 3-hydroxy-5- (3-methoxy phenyl)-4-(4- méthyl benzoyl)-1-[3- (4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 243 79.5 >10 μM 9.4 μM  4 I 7111786 4-benzoyl-5- (2,5- dimethoxy phenyl)-3- hydroxy-1-[3- (1H-imidazol- 1-yl)propyl]- 1,5-dihydro- 2H-pyrrol-2- one 557 77.5 >10 μg/ml  5 I 6203863 ethyl 4-[3- benzoyl-2- (2,4- dimethoxy phenyl)-4- hydroxy-5- oxo-2,5- dihydro-1H- pyrrol-1-yl] benzoate 423 73.5 >10 μg/ml 6.1 μM  6 I 7347627 4-(2,5- dimethyl benzoyl)-3- hydroxy-5-(2- methoxy phenyl)-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 775 72 >10 μg/ml  7 I 7489416 5-(3-bromo-4- hydroxy-5- methoxy phenyl)-3- hydroxy-1-(2- phenylethyl)- 4-(2-thienyl carbonyl)-1,5- dihydro-2H- pyrrol-2-one 648 71 >10 μg/ml  8 I 6719738 4-(4-fluoro benzoyl)-3- hydroxy-5-(3- phenoxy phenyl)-1-(3- pyridinyl methyl)-1,5- dihydro-2H- pyrrol-2-one 977 71 >10 μg/ml  9 I 6650234 ethyl 2-[3-(4- fluoro- benzoyl)- 4-hydroxy-2- (4-methyl phenyl)-5-oxo- 2,5-dihydro- 1H-pyrrol-1- yl]-4-methyl- 1,3-thiazole-5- carboxylate 946 71 ~10 μg/ml 6.3 μM 10 I 6652639 5-(2,5- dimethoxy phenyl)-3- hydroxy-4-(4- methyl benzoyl)-1-(3- pyridinyl methyl)-1,5- dihydro-2H- pyrrol-2-one 829 70.5 >10 μg/ml 11 I 6673225 ethyl 2-[3- benzoyl-4- hydroxy-2-(4- methoxy phenyl)-5-oxo- 2,5-dihydro- 1H-pyrrol-1- yl]-4-methyl- 1,3-thiazole-5- carboxylate 301 69 >10 μg/ml 12 I 6800873 3-[2-(2,4- dimethoxy phenyl)-2- oxoethyl]-3- hydroxy-1-(1- naphthyl methyl)-1,3- dihydro-2H- indol-2-one 250 67.5 >10 μg/ml 4.6 μM 13 6879058 3-hydroxy-4- (4-methoxy-2- methyl benzoyl)-1-[2- (4- morpholinyl) ethyl]-5-(3- pyridinyl)-1,5- dihydro-2H- pyrrol-2-one 758 67.5 >10 μg/ml 14 I 6282824 2-methoxy-N- (4-{4-methyl- 5-[(2-oxo-2- phenylethyl) thio]-4H-1,2, 4-triazol-3- yl}phenyl) benzamide 905 67.5 ~2.5 μg/ml 5.6 μM 15 II 6750319 methyl 2-[3- benzoyl-4- hydroxy-2-(4- méthyl phenyl)-5-oxo- 2,5-dihydro- 1H-pyrrol-1- yl]-4-methyl- 1,3-thiazole-5- carboxyiate 850 67 >10 μg/ml 16 I 6474819 4-[(4-benzyl- 1-piperidinyl) methyl]-N-(2- methoxy-5- methylphenyl) benzamide 954 63 >10 μg/ml 17.9 μM 17 6498669 4-{[N-[(4- methoxy phenyl) sulphonyl]-N- (2-phenyl ethyl)glycyl] amino} benzamide 808 63 >10 μg/ml 0.8 μM 18 III 6651552 4-(4-fluoro benzoyl)-3- hydroxy-5-(4- isopropyl phenyl)-1-[3- (4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 100 60 >10 μg/ml 19 I 6453860 N,N′-1,5- naphthalene- diylbis[2-(3- methyl- phenoxy) acetamide] 145 60 >10 μg/ml 20 6853966 N-[4-({[4- (acetyl amino) phenyl] sulphonyl} amino)-2,5- dimethoxy phenyl] benzamide 11 58 >10 μg/ml 21 6866968 7,7-dimethyl- 1-(4-methyl phenyl)-2,5- dioxo-N- (2,2,6,6- tetramethyl-4- piperidinyl)- 1,2,5,6,7,8- hexahydro-3- quinoline carboxamide 255 57.5 >10 μg/ml 22 7938324 4-(benzyl{[1- phenyl-3-(2- thienyl)-1H- pyrazol-4- yl]methyl} amino)-4-oxo butanoic acid 795 57 >10 μg/ml 23 _— 6905988 4-(4-fluoro benzoyl)-3- hydroxy-5-(3- methoxy phenyl)-1-[3- (4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 843 57 >10 μg/ml 24 I 6885782 4-(4-chloro benzoyl)-3- hydroxy-1-[3- (4- morpholinyl) propyl]-5-(3, 4,5-trimethoxy phenyl)-1,5- dihydro-2H- pyrrol-2-one 249 56.5 >10 μg/ml 25 I 6663684 4-benzoyl-3- hydroxy-5-(4- isopropyl phenyl)-1 -[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 530 56.5 >10 μg/ml 26 I 6672500 4-(4-chloro benzoyl)-5- (3,4- dimethoxy phenyl)-3- hydroxy-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 194 55 >10 μg/ml 27 I 7721949 1-[2-(diethyl amino)ethyl]- 5-(2,5- dimethoxy phenyl)-3- hydroxy-4-(4- methyl benzoyl)-1,5- dihydro-2H- pyrrol-2-one 139 54 >10 μg/ml 28 I 7966545 2-fluoro-N- [(5- {[(4-oxo-3,4- dihydro-2- quinazolinyl) methyl]thio}- 4-phényl-4H- 1,2,4-triazol- 3-yl)methyl] benzamide 773 53.5 ~10 μg/ml 6.7 μM 29 7437580 2-{[4-(1,3- dioxo-1,3- dihydro-2H- isoindol-2- yl)butanoyl] amino}-N- (tetrahydro-2- furanyl methyl)-5,6- dihydro-4H- cyclopenta[b] thiophene-3- carboxamide 222 51 >10 μg/ml 30 6654239 5-(3,4- dimethoxy phenyl)-4-(4- fluoro benzoyl)-3- hydroxy-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 343 50 >10 μg/ml 31 I 6670570 4-({4- hydroxy- 1-[2-(4- morpholinyl) ethyl]-5-oxo- 2-phenyl-2,5- dihydro-1H- pyrrol-3-yl} carbonyl)- N,N-dimethyl benzene sulphonamide 926 50 >10 μg/ml 32 I 6670673 5-(2,4- dimethoxy phenyl)-4-(4- fluoro- benzoyl)- 3-hydroxy-1- [2-(4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 742 50 >10 μg/ml 33 I 6670747 3-hydroxy-5- (4-methoxy phenyl)-4-(4- methyl benzoyl)-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 413 50 >10 μg/ml 34 I 6671401 5-(3,4- diméthoxy phenyl)-4-(2- furoyl)-3- hydroxy-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 954 50 >10 μg/ml 35 I 6672500 4-(4-chloro benzoyl)-5- (3,4- dimethoxy phenyl)-3- hydroxy-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 194 50 >10 μg/ml 36 I 6673225 ethyl 2-[3- benzoyl-4- hydroxy-2-(4- methoxy phenyl)-5-oxo- 2,5-dihydro- 1H-pyrrol-1- yl]-4-methyl- 1,3-thiazole-5- carboxylate 124 50 >10 μg/ml 37 I 6677533 5-(3,4- dimethoxy phenyl)-3- hydroxy-1-[2- (4- morpholinyl) ethyl]-4-(2- thienyl carbonyl)-1,5- dihydro-2H- pyrrol-2-one 409 50 >10 μg/ml 38 I 6683618 4-(4-chloro benzoyl)-3- hydroxy-5-(4- methoxy phenyl)-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 808 50 >10 μg/ml 39 I 6417902 N-[2-(4- benzyl-1- piperazinyl)-2- oxoethyl]-N- (3,5-dimethyl phenyl) benzene sulphonamide 897 49.5 >10 μg/ml 40 III 6437157 1-methyl-2-(4- methyl phenyl)-2- oxoethyl 2-(3- chloro-4- methyl phenyl)-1,3- dioxo-5- isoindoline carboxylate 871 49.5 >10 μg/ml 41 6465972 N~2~-[(3,4- dimethoxy phenyl) sulphonyl]- N~1~-(2- methoxy-5- methyl- phenyl)- N~2~-(4- methylphenyl) glycinamide 926 49 >10 μg/ml 42 III 7723671 5-(2,5- dimethoxy phenyl)-4-(4- fluoro benzoyl)-3- hydroxy-1-[3- (4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 34 48.5 >10 μg/ml 43 I 6648368 4-(4- chloro- benzoyl)- 5-(2- fluorophenyl)- 3-hydroxy-1- [3-(4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 465 48.5 >10 μg/ml 44 I 6656195 4-(4-fluoro benzoyl)-3- hydroxy-5-(4- isopropyl phenyl)-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 613 48.5 >10 μg/ml 45 I 7778331 4-{[2-(4- morpholinyl) ethyl]amino}- 3-(4- morpholinyl sulphonyl)-N- phenyl- benzamide 998 48 >10 μg/ml 3 μM 46 6994060 2-chloro-N- {4-[4-methyl- 5-({2-oxo-2- [(tetrahydro- 2-furanyl methyl) amino] ethyl}thio)- 4H-1,2,4- triazol-3-yl] phenyl} benzamide 623 47.5 >10 μg/ml 47 II 6458830 N-[2-(4- benzyl-1- piperazinyl)-2- oxoethyl]-N- (3,4-dimethyl phenyl)-4- methyl benzene sulphonamide 837 47 >10 μg/ml 48 III 7524107 2-[(4-{[(4- isopropyl phenoxy) acetyl]amino}- 3-methyl benzoyl) amino] benzoic acid 789 46.5 ~10 μg/ml 5.5 μM 49 6661524 4-({2-(3,4- dichloro phenyl)-1-[3- (dimethyl amino)propyl]- 4-hydroxy-5- oxo-2,5- dihydro-1H- pyrrol-3-yl} carbonyl)- N,N-dimethyl benzene sulphonamide 428 46.5 >10 μg/ml 50 I 7739436 4-(1,3- benzodioxol- 5-yl carbonyl)- 1-[3-(diethyl amino)propyl]- 3-hydroxy-5- (3-pyridinyl)- 1,5-dihydro- 2H-pyrrol-2- one 976 46 >10 μg/ml 51 I 6881804 1-[2-(dimethyl amino)ethyl]- 3-hydroxy-4- (5-methyl-2- furoyl)-5- (3,4,5- trimethoxy phenyl)-1,5- dihydro-2H- pyrrol-2-one 230 46 >10 μg/ml 52 I 6907921 3-hydroxy-5- (3-methoxy phenyl)-4-(5- methyl-2- furoyl)-1-[2-(4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 609 45.5 >10 μg/ml 53 I 7495334 ethyl 4-({[(5,6- di-2-furyl- 1,2,4-triazin-3- yl)thio]acetyl} amino) benzoate 380 43 ~5 μg/ml 1.6 μM 54 7509862 ethyl 5-cyano- 4-(2-furyl)-6- ({2-[(3- methoxy phenyl) amino]-2- oxoethyl} thio)-2-phenyl- 1,4-dihydro-3- pyridine carboxylate 170 42.5 ~5 μg/ml 55 7325385 5-(2,3- dimethoxy phenyl)-4- (2,5-dimethyl benzoyl)-3- hydroxy-1-[3- (4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 65 42 ~10 μg/ml 56 I 6669449 3-(6-amino-5- cyano-3- phenyl-1,4- dihydro pyrano[2,3- c]pyrazol-4- yl)phenyl 2- furoate 140 42 >10 μg/ml 57 7348779 1,4-bis [(mesityloxy) acetyl] piperazine 209 40 ~10 μg/ml 58 7501888 N-(4-chloro pheny)-2-{[4- (2-phenyl ethyl)-5- (3,4,5- trimethoxy phenyl)-4H- 1,2,4-triazol- 3-yl]thio} acetamide 544 40 ~2 μg/ml 8.2 μM 59 6946138 {[3-(ethoxy carbonyl)-2- phenyl-1- benzofuran-5- yl]oxy} (phenyl) acetic acid 298 40 >10 μg/ml 60 7722851 ethyl 4-({[1- (4-chloro phenyl)-5- oxo-3-(3- pyridinyl methyl)-2- thioxo-4- imidazolidinyl] acetyl} amino) benzoate 638 39.5 ~2 μg/ml 21.8 μM 61 IV 7517583 5,5′-oxybis [2- (tetra hydro-2- furanyl methyl)-1H- isoindole- 1,3(2H)-dione] 912 38.5 >10 μg/ml 62 6634701 7-acetyl-6-[3- (benzyloxy) phenyl]-3- (methylthio)- 6,7-dihydro [1,2,4] triazino[5,6- d][3,1] benzoxazepine 934 38.5 ~5 μg/ml 63 7726450 3-hydroxy-1- [3-(1H- imidazol-1- yl)propyl]-4- [(7-methoxy- 1-benzofuran- 2-yl) carbonyl]-5- (2-pyridinyl)- 1,5-dihydro- 2H-pyrrol-2- one 472 37.5 >10 μg/ml 64 I 6662088 4-{[4-hydroxy- 1-[3-(4- morpholinyl) propyl]-5-oxo- 2- (3-pyridinyl)- 2,5-dihydro- 1H-pyrrol-3- yl]carbonyl}- N,N-dimethyl benzene sulphonamide 156 37 >10 μg/ml 65 I 7752193 N-{1-[4-allyl- 5-({2-[(3- methoxy phenyl) amino]-2- oxoethyl} thio)-4H-1,2,4- triazol-3- yl]ethyl} benzamide 248 35 ~5 μg/ml 8 μM 66 7238569 N-(2-hydroxy- 1,1-dimethyl- ethyl)-5-{4-[(3- hydroxyphenyl) amino]-1- phthalazinyl}- 2-methyl benzene sulphonamide 749 32 >10 μg/ml 67 7724000 4-(1,3- benzodioxol- 5-ylcarbonyl)- 5-(2- fluorophenyl)- 3-hydroxy-1- [3-(4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 303 30 ~10 μg/ml 68 I 7443270 N-(2,4- dimethoxy- phenyl)- 2-{[3-(2- furylmethyl)-4- oxo-3,4- dihydro-2- quinazolinyl] thio} acetamide 895 30 ~5 μg/ml 69 7245019 N-[(2-hydroxy- 7-méthyl-3- quinolinyl) methyl]-3- methoxy-N-(2- methoxy phenyl) benzamide 608 30 >10 μg/ml 70 6909597 4-(4- chlorobenzoyl)- 3-hydroxy-5- (3-methoxy phenyl)-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 790 29 >10 μg/ml 71 I 7661882 2-(4-methoxy phenoxy)-N- [2-methyl-5- (3-methyl-4- oxo-3,4- dihydro-1- phthalazinyl) benzyl] acetamide 941 28 ~10 μg/ml 72 7667791 N-{4-[({[4-(4- methoxy phenyl) tetrahydro-2H- pyran-4-yl] methyl} amino) carbonyl] phenyl}-2- furamide 797 28 >10 μg/ml 73 6891745 4-benzoyl-5- (2,3- dimethoxy phenyl)-3- hydroxy-1 -[3- (4- morpholinyl) propyl]-1,5- dihydro-2H- pyrrol-2-one 115 27.5 >10 μg/ml 74 I 7783660 isopropyl 3- ({[(4-allyl-5- {[(3-methyl benzoyl) amino] methyl}-4H- 1,2,4-triazol-3- yl)thio]acetyl} amino) benzoate 266 27 ~5 μg/ml 4.8 μM 75 II 7653478 2-{5-[1-(4- morpholinyl) cyclohexyl]- 1H-tetrazol-1- yl}ethyl 1- naphthyl carbamate 365 27 >10 μg/ml 76 7723330 methyl 4-({[3- (1,3- benzodioxol- 5-ylmethyl)- 2,5-dioxo-1- phenyl-4- imidazol idinyl]acetyl} amino) benzoate 965 26 >10 μg/ml 77 IV 7199725 4-(3,4- dihydro-2(1H)- isoquinolinyl- methyl)-N-[2- (1-pyrrolidinyl carbonyl) phenyl] benzamide 987 25 >10 μg/ml 78 6987235 9-{3-chloro-4- [(4-methyl benzyl)oxy] phenyl}-10- ethyl- 3,4,6,7,9,10- hexahydro- 1,8(2H,5H)- acridine dione 680 25 >10 μg/ml 79 _— 7140931 ethyl 1-(4- {[(3,4-dimethyl phenyl) (methyl sulphonyl) amino] methyl} benzoyl)-4- piperidine carboxylate 14 24.5 >10 μg/ml 80 7787455 methyl 4-{[N- (3-methoxy phenyl)-N- (phenyl sulphonyl) glycyl] amino} bonzoate 407 20 >10 μg/ml 81 III 7660465 N-{[5-({2-[(4- bromo-2,3- dimethyl phenyl) amino]-2- oxoethyl} thio)-4-ethyl- 4H-1,2,4- triazol-3- yl]methyl}-4- chloro benzamide 881 20 ~7 μg/ml 3.2 μM 82 II 7661751 4-(2,3- dihydro-1,4- benzodioxin- 6-yl carbonyl)- 3-hydroxy-1- [3-(4- morpholinyl) propyl]-5-(4- pyridinyl)-1,5- dihydro-2H- pyrrol-2-one 951 19 ~10 μg/ml 83 I 7722914 N-(4-ethoxy phenyl)-2-{1- (4-methoxy phenyl)-3-[2- (4- morpholinyl) ethyl]-5-oxo-2- thioxo-4- imidazolidinyl} acetamide 806 19 ~7 μg/ml 15.6 μM 84 IV 7745040 5-(3-bromo phenyl)-3- hydroxy-4-[(7- methoxy-1- benzo furan- 2-yl) carbonyl]-1-[2- (4- morpholinyl) ethyl]-1,5- dihydro-2H- pyrrol-2-one 313 18 ~10 μg/ml 85 I 7735385 1-[3-(diethyl amino)propyl]- 3-hydroxy-4- [(7-methoxy- 1-benzofuran- 2-yl) carbonyl]-5- (2-pyridinyl)- 1,5-dihydro- 2H-pyrrol-2- one 21 17 ~10 μg/ml 86 I 7756003 1-(4-{[(4,6- dimethyl-2- pyrimidinyl) thio]acetyl}-1- piperazinyl)-4- (4-methyl phenyl) phthalazine 361 11 ~5 μg/ml 87 CB ref: ChemBridge reference; Rank: rank of each molecule in the list of the 1000 best molecules after in silico docking; In vit. inhib.: mean inhibition % obtained with each molecule for the displacement of the binding of scFv G4G11 with Syk in vitro in the ADA method; Degran. IC50: concentration inhibiting mast cell degranulation by 50%; Kd: dissociation constant with respect to Syk measured in vitro by spectrofluorometry No.: number assigned by the inventors Gp: groups to which the molecules belong.

Example 3 Materials and Methods

1) Chemical products and antibodies. A chemical bank of 3000 molecules (a varied subset) was acquired from ChemBridge, Inc. (San Diego, Calif.). Stocks of solutions of small molecules were prepared at a rate of 10 mM in DMSO (dimethylsulphoxide), except for C-13 (methyl 2-{5-[(3-benzyl-4-oxo-2-thioxo-1,3-thiazolidin-5-ylidene)methyl]-2-furyl}benzoate, Chem Bridge ID No. 6197026) prepared in DMF (dimethylformamide). Unless specified otherwise, all the reagents were supplied by Sigma. Dinitrophenyl (DNP) hapten was acquired from Calbiochem. Sepharose GammaBind G and all the secondary antibodies were supplied by GE Health Amersham Biosciences. The anti-Syk, anti-Lyn, anti-Btk, anti-PLC-γ1, anti-PLC-γ2, anti-LAT, anti-SLP-76, anti-p38, anti-JNK, anti-Vav, anti-Akt1 and 9E10 antibodies conjugated with HRP were acquired from Santa Cruz Biotechnology. The anti-phospho-p44/42 MAP Kinase, anti-p44/42 MAP Kinase, anti-phospho-p38, anti-phospho-JNK, anti-phospho-Akt1 antibodies were acquired from Cell Signaling. The anti-phospho-LAT and anti-phospho-PLC-γ1 antibodies were supplied by Biosource and the anti-phospho-SLP-76 antibodies by BD Pharmingen. The 4G10 anti-phosphotyrosine monoclonal antibody was acquired from Upstate Biotechnology.

2) ADA method: ELISA type high-speed molecule screening test, based on antibody displacement (WO 2005106481). The recombinant fusion protein GST:Syk 6-2428 comprising the residues 6 to 242 of murine Syk tyrosine kinase protein illustrated in FIG. 8B (SEQ ID No. 4) was immobilised on an ELISA plate at a final concentration of 10 μg ml−1. For the screening of the chemical molecule bank, the small molecules, diluted in PBS at a final concentration of 10 μM were added to the wells for one hour at ambient temperature, before adding the fragment scFv G4G11 at a final concentration of 100 nM for an additional hour. The binding of G4G11 with Syk was assessed by adding the 9E10 monoclonal antibody conjugated with HRP detecting the amino acid sequence EQKLISEEDLN of human c-myc protein located at the C-terminal end of the scFv fragment. To generate Syk mutants, targeted mutagenesis was used on GST:Syk 6-242 protein and the binding of G4G11 with the mutants was assessed in the presence of 5 μM of C-13.

3) Cells, culture conditions and functional tests. Anti-DNP 2682-I mouse monoclonal antibody was used as the culture supernatant of hybridomas containing 1 μg/ml of IgE. Femoral bone marrow cells were sampled and cultured in Opti-MEM medium (Gibco) supplemented with 10% foetal calf serum and 4% X63 transfectant supernatant secreting murine IL-3. RBL-2H3 (ATCC) leukaemic rat basophil cells were maintained in a single-layer culture in RPMI 1640 medium supplemented with 10% foetal calf serum (Gibco). Measurements of β-hexosaminidase released by the RBL-2H3 cells were performed as described previously7, except that, after 12-16 hours of incubation with anti-DNP IgE (0.5 μg/ml), the cells were incubated for 90 min at 37° C. in RPMI medium supplemented with the specified concentrations of C-13 or DMF (0.25%). The cells were stimulated for 45 min with DNP-BSA (50 ng ml−1) or ionomycin (1.5 μM). The BMMC cells were incubated for one hour at 37° C. with anti-DNP IgE (100 ng/ml). They were then incubated with C-13 (3 μM) or DMF (0.3%) for 3 hours at 37° C., and stimulated with varied concentrations of DNP-BSA. The level of β-hexosaminidase released was measured 10 min later and the TNF-α titration was performed by means of a cytotoxicity test on L929 cells as described previously10, 3 hours after stimulation. The results illustrated in FIG. 3 are representative of three independent experiments.

4) Immunoprecipitations, in vitro kinase assays and immunodetection. All the experiments were conducted as described previously7, except that, before stimulation with DNP-BSA (50 ng ml−1, 3 min), the RBL-2H3 cells were incubated for 90 min at 37° C. in RPMI medium supplemented with the specified concentrations of C-13 or DMF. The cells were solubilised in DOC modified lysis buffer (1% NP-40, 0.25% sodium deoxycholate, 0.1% SDS in PBS buffer supplemented with protease and phosphatase inhibitors) and the protein concentration was determined (BCA Protein Assay, PIERCE). For immunoprecipitations, cell lysates, non-stimulated and stimulated with IgE/DNP were incubated with preformed complexes of antibodies and Sepharose GammaBind G, and the in vitro kinase activity of Syk, Btk and Lyn immunoprecipitates were examined. Before SDS-PAGE gel separation the lysates or immunoprecipitates were prepared by adding SDS sample buffer (60 mM Tris, pH 6.8, 2.3% SDS, 10% glycerol, 0.01% bromophenol blue). The proteins were transferred onto a nitrocellulose membrane (Schleicher & Schuell), and detected using suitable antibodies and the chemoluminescence system improved (ExactaCruz, Santa Cruz Biotechnology).

5) Flow cytometry analysis of level and calcium mobilisation and FcεRI membrane expression. The intracellular free calcium concentration was determined by previously charging 1×106 cells with 5 mM of Fluo-3 AM (Molecular Probes, Invitrogen) in the presence of 0.2% Pluronic F-127 for 30 min at ambient temperature. Prior to stimulation with DNP-BSA or ionomycin, the cells were treated for 90 min at 37° C. in RPMI medium supplemented with C-13 or DMF (0.25%), and the intracellular free calcium concentration was measured with a flow cytometer (Beckton Dickinson). For the FcεRI surface expression evaluation, the cells were incubated for 2 hours at 37° C. with anti-DNP IgE. Membrane-bound IgE was detected using biotinylated anti-mouse Ig, and streptavidine conjugated with Fitc.

6) Anaphylaxis induction. Female BALB/c mice (aged 6-8 weeks) were acquired from Charles River and kept at the IRCM animal house under pathogen-free conditions. The Ig-dependent passive systemic anaphylaxis (PSA) and passive cutaneous anaphylaxis (PCA) protocols were conducted as described previously11. Briefly, the mice received, by intravenous injection, 100 μg of IgE (SPE-7, Sigma) in 200 μl of PBS for PSA, or, by intradermal injection, 25 ng of IgE in 10 μl of PBS for PCA, and were stimulated 24 hours later by means of an intravenous injection of 1 mg of DNP-KLH in 2% of Evans blue. C-13, a non-relevant chemical molecule or the vehicle were administered 1 hours before stimulation, either orally (PSA) in 200 μl of 1% carboxymethylcellulose, or locally in the ear by means of intradermal injection (PCA) in an acetone/olive oil mixture (4:1). The animals were sacrificed 20 min after stimulation. The ears were removed, ground and Evans blue was extracted after overnight incubation in formamide at 80° C. For the temperature measurements in PSA, C-13 (100 mg/kg) or the vehicle were administered orally, 3 hours prior to stimulation performed in the absence of Evans blue. The temperature was measured using an electronic thermometer with a rectal probe (YSI, Yellow Springs, Ohio) before stimulation and for 60 minutes afterwards, prior to sacrifice. The absorbance was measured at 610 nm. The experiments were conducted with 4-5 mice per condition. The data illustrated in FIG. 4 are representative of three different experiments.

7) Structural studies. The three-dimensional cavities liable to be pharmaceutical targets were predicted using Q-SiteFinder12 and ICM13. The molecule C-13 was docked using LigandFit14 and Surflex15. The first 20 positions were analysed and a consensus position is given in FIG. 1A. The images were generated with PyMol.

8) Peritonitis (FIG. 5). Syk-dependent peritoneal neutrophil recruitment was induced in 8-week old female BALB/c mice as described in36 by intravenous injection of 4 μg of Bordetella pertussis toxin (donated by Dr D. Raze, Inserm, Lille, France) and 2 hours later by intraperitoneal injection of 4% thioglycollate in sterile water. A peritoneal lavage was performed with 5 ml of PBS 4 hours later and the total number of neutrophils was determined after labelling with anti-Gr1 conjugated with APC (Becton-Dickinson) and flow cytometry analysis. C-13 (100 mg/kg) in CMC or the vehicle alone was administered orally one hour prior to injecting Bordetella pertussis toxin.

9) In vitro B Lymphocyte purification and proliferation. Spleen B lymphocyte cells were purified from 8-week old female BALB/c mice on magnetic beads by negative selection using micro-beads coated with CD43 and LS columns (Miltenyi Biotec) as described previously37. After two hours of incubation with variable concentrations of C-13 or the vehicle, 50,000 cells were cultured for 48 hours in 96-well plates in the presence of absence of 10 μg/ml of donkey anti-mouse IgM F(ab′)2 fragment (Jacson Immunoresearch). Alamar blue (Serotec) was added to the cultures 24 prior to measuring the reduced versus oxidised forms of the reagent at 570 and 620 nm, in accordance with the manufacturers instructions.

10) Antibody production (FIG. 5C). This experiment was conducted as described in38. Eight-week old female BALB/c mice received an oral dose of C-13 (150 mg/kg) in 1% CMC or the vehicle alone, were immunised 3 hours later by an intraperitoneal injection of 10 μg of trinitophenyl-keyhole limpet haemocyanin (TNP-KLH) in Rehydragel alum (Reheiss). The serum was collected before immunisation and on day 12. The antigen-specific immunoglobulin levels were measured by means of ELISA with 10 μg/ml of plate-bound TNP-OVA (Biosearch technologies) as the capture agent. The IgM, IgG1, IgG2a, IgG2b, IgG3 and IgA levels were measured using samples diluted to 1:5000 using goat antibodies conjugated with peroxidase and isotype-specific (Southern Biotechnology), whereas the IgE levels were measured using samples diluted to 1:50 using biotinylated anti-mouse IgE rat antibodies (Becton Dickinson) and streptavidine conjugated with peroxidase (R&D Systems). After incubation with TMB substrate, the optical density (OD) was measured at 450 nm.

11) Statistical analyses. The mean numeric data are expressed as means±standard deviations (SD). Students t test was used to determine the statistical significance of the differences between groups.

12) C-13 toxicity test on BMMC cells. To assess the potential toxic effect of C-13 on mast cells, BMMC cells were incubated for 5 days at 37° C. in the presence of 2.5 μM or 5 μM of C-13, or 0.25% DMF (corresponding to the DMF concentration used with 5 μM of C-13) under the same conditions as for functional tests. Double labelling with Annexin-V and Propidium Iodide demonstrated the level of BMMC cell viability after 3 hours and after 5 days. The viability of the BMMC cells treated under the same conditions with Staurosporine was measured under the same conditions.

REFERENCES

  • 1. Kambayashi T, Koretzky G A. Proximal signaling events in Fc epsilon R1-mediated mast cell activation. J Allergy Clin Immunol 2007; 119:544-52; quiz 53-4.
  • 2. Rivera J, Gilfillan A M, Molecular regulation of mast cell activation. J Allergy Clin Immunol 2006; 117:1214-25; quiz 26.
  • 3. Costello P S, Turner M, Walters A E, Cunningham C N, Bauer P H, Downward J, et al. Critical role for the tyrosine kinase Syk in signalling through the high affinity IgE receptor of mast cells. Oncogene 1996; 13:2595-605.
  • 4. Wong W S, Leong K P. Tyrosine kinase inhibitors: a new approach for asthma. Biochim Biophys Acta 2004; 1697:53-69.
  • 5. Turner M, Schweighoffer E, Colucci F, Di Santo J P, Tybulewicz V L. Tyrosine kinase Syk: essential functions for immunoreceptor signalling. Immunol Today 2000; 21:148-54.
  • 6. Coopman P J, Mueller S C. The Syk tyrosine kinase: a new negative regulator in tumor growth and progression. Cancer Lett 2006; 241:159-73.
  • 7. Dauvillier S, Merida P, Visintin M, Cattaneo A, Bonnerot C, Dariavach P. Intracellular single-chain variable fragments directed to the Src homology 2 domains of Syk partially inhibit Fc epsilon R1 signaling in the RBL-2H3 cell line. J Immunol 2002; 169:2274-83.
  • 8. Peneff C, Lefranc M P, Dariavach P. Characterisation and specificity of two single-chain Fv antibodies directed to the protein tyrosine kinase Syk. J Immunol Methods 2000; 236:105-15.
  • 9. Lobato M N, Rabbitts T H. Intracellular antibodies and challenges facing their use as therapeutic agents. Trends Mol Med 2003; 9:390-6.
  • 10. Latour S, Bonnerot C, Fridman W H, Daeron M. Induction of tumor necrosis factor-alpha production by mast cells via Fc gamma R. Role of the Fc gamma RIII gamma subunit. J Immunol 1992; 149:2155-62.
  • 11. Dombrowicz D, Flamand V, Miyajima I, Ravetch J V, Galli S J, Kinet J P. Absence of Fc epsilonRI alpha chain results in upregulation of Fc gamnnaRIII-dependent mast cell degranulation and anaphylaxis. Evidence of competition between Fc epsilonRI and Fc gammaRIII for limiting amounts of FcR beta and gamma chains. J Clin Invest 1997;
  • 12. Laurie A T, Jackson R M. Q-SiteFinder: an energy-based method for the prediction of protein-ligand binding sites. Bioinformatics 2005; 21:1908-16.
  • 13. An J, Totrov M, Abagyan R. Comprehensive identification of “druggable” protein ligand binding sites. Genome Inform 2004; 15:31-41.
  • 14. Venkatachalam C M, Jiang X, Oldfield T, Waldman M. LigandFit: a novel method for the shape-directed rapid docking of ligands to protein active sites. J Mol Graph Model 2003; 21:289-307.
  • 15. Jain A N. Surflex: fully automatic flexible molecular docking using a molecular similarity-based search engine. J Med Chem 2003; 46:499-511.
  • 16. Frank R, Overwin H. SPOT synthesis. Epitope analysis with arrays of synthetic peptides prepared on cellulose membranes. Methods Mol Biol 1996; 66:149-69.
  • 17. Futterer K, Wong J, Grucza R A, Chan A C, Waksman G. Structural basis for Syk tyrosine kinase ubiquity in signal transduction pathways revealed by the crystal structure of its regulatory SH2 domains bound to a dually phosphorylated ITAM peptide. J Mol Biol 1998; 281:523-37.
  • 18. Nadler M J, Matthews S A, Turner H, Kinet J P. Signal transduction by the high-affinity immunoglobulin E receptor Fc epsilon R1: coupling form to function. Adv Immunol 2000; 76:325-55.
  • 19. Parravicini V, Gadina M, Kovarova M, Odom S, Gonzalez-Espinosa C, Furumoto Y, et al. Fyn kinase initiates complementary signals required for IgE-dependent mast cell degranulation. Nat Immunol 2002; 3:741-8.
  • 20. Rameh L E, Rhee S G, Spokes K, Kazlauskas A, Cantley L C, Cantley L G. Phosphoinositide 3-kinase regulates phospholipase Cgamma-mediated calcium signaling. J Biol Chem 1998; 273:23750-7.
  • 21. Saitoh S, Odom S, Gomez G, Sommers C L, Young H A, Rivera J, et al. The four distal tyrosines are required for LAT-dependent signaling in FcepsilonRI-mediated mast cell activation. J Exp Med 2003; 198:831-43.
  • 22. Silverman M A, Shoag J, Wu J, Koretzky G A. Disruption of SLP-76 interaction with Gads inhibits dynamic clustering of SLP-76 and FcepsilonRI signaling in mast cells. Mol Cell Biol 2006; 26:1826-38.
  • 23. Hendricks-Taylor L R, Motto D G, Zhang J, Siraganian R P, Koretzky G A. SLP-76 is a substrate of the high affinity IgE receptor-stimulated protein tyrosine kinases in rat basophilic leukemia cells. J Biol Chem 1997; 272:1363-7.
  • 24. Takata M, Kurosaki T. A role for Bruton's tyrosine kinase in B cell antigen receptor-mediated activation of phospholipase C-gamma 2. J Exp Med 1996; 184:31-40.
  • 25. Law C L, Chandran K A, Sidorenko S P, Clark E A. Phospholipase C-gammal interacts with conserved phosphotyrosyl residues in the linker region of Syk and is a substrate for Syk. Mol Cell Biol 1996; 16:1305-15.
  • 26. Pivniouk V I, Martin T R, Lu-Kuo J M, Katz H R, Oettgen H C, Geha R S. SLP-76 deficiency impairs signaling via the high-affinity IgE receptor in mast cells. J Clin Invest 1999; 103:1737-43.
  • 27. Kettner A, Pivniouk V, Kumar L, Falet H, Lee J S, Mulligan R, et al. Structural requirements of SLP-76 in signaling via the high-affinity immunoglobulin E receptor (Fc epsilon R1) in mast cells. Mol Cell Biol 2003; 23:2395-406.
  • 28. Manetz T S, Gonzalez-Espinosa C, Arudchandran R, Xirasagar S, Tybulewicz V, Rivera J. Vav1 regulates phospholipase cgamma activation and calcium responses in mast cells. Mol Cell Biol 2001; 21:3763-74.
  • 29. Wilson B S, Pfeiffer J R, Oliver J M. FcepsilonRI signaling observed from the inside of the mast cell membrane. Mol Immunol 2002; 38:1259-68.
  • 30. Wu J N, Jordan M S, Silverman M A, Peterson E J, Koretzky G A. Differential requirement for adapter proteins Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa and adhesion- and degranulation-promoting adapter protein in FcepsilonRI signaling and mast cell function. J Immunol 2004; 172:6768-74.
  • 31. Barker S A, Caldwell K K, Pfeiffer J R, Wilson B S. Wortmannin-sensitive phosphorylation, translocation, and activation of PLCgamma1, but not PLCgamma2, in antigen-stimulated RBL-2H3 mast cells. Mol Biol Cell 1998; 9:483-96.
  • 32. Fluckiger A C, Li Z, Kato R M, Wahl M I, Ochs H D, Longnecker R, et al. Btk/Tec kinases regulate sustained increases in intracellular Ca2+ following B-cell receptor activation. Embo J 1998; 17:1973-85.
  • 33. Scharenberg A M, El-Hillal O, Fruman D A, Beitz L O, Li Z, Lin S, et al. Phosphatidylinositol-3,4,5-trisphosphate (PtdIns-3,4,5-P3)/Tec kinase-dependent calcium signaling pathway: a target for SHIP-mediated inhibitory signals. Embo J 1998; 17:1961-72.
  • 34. Wen R, Jou S T, Chen Y, Hoffmeyer A, Wang D. Phospholipase C gamma 2 is essential for specific functions of Fc epsilon R and Fc gamma R. J Immunol 2002; 169:6743-52.
  • 35. Qi Q, August A. Keeping the (Kinase) Party Going: SLP-76 and ITK Dance to the Beat. Sci STKE 2007; 2007: pe39.
  • 36. Mocsai A, Zhou M, Meng F, Tybulewicz V L, Lowell C A. Syk is required for integrin signaling in neutrophils. Immunity 2002; 16: 547-558.
  • 37. Matsumoto T, Guo Y J, Ikejima T, Yamada H. Induction of cell cycle regulatory proteins by murine B cell proliferating pectic polysaccharide from the roots of Bupleurum falcatum L. Immunology letters 2003; 89: 111-118.
  • 38. Okkenhaug K, Bilancio A, Farjot G, Priddle H, Sancho S, Peskett E, et al. Impaired B and T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice. Science 2002; 297: 1031-1034.
  • 39. Gell PGH, Coombs RRA, eds. Clinical Aspects of Immunology. 1st ed. Oxford, England: Blackwell; 1963
  • 40. S. M. Berge et al. Pharmaceutical Salts. J. Pharm. Sci, 1977, 66: p. 1-19

Claims

1. A compound which binds with Syk tyrosine kinase protein on a three-dimensional cavity comprising the Arginine residue situated in position 68 and the two glutamic acid residues situated in positions 121 and 155 of human Syk protein, the sequence of which is set forth in SEQ ID No. 1, and capable of inhibiting by at least 10% in vitro binding of with human Syk tyrosine kinase protein or with any of the variants thereof in animals, for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk in humans or animals, where said compound (C-13) has the formula or an organic compound functionally equivalent to molecule C-13 having a molecular weight between 50 and 2500 Dalton, selected from compounds having any of the following formulae: where or where or where or where or or a stereo-isomer, racemate or pharmacologically acceptable salt of C-13 or said equivalent compound.

(i) antibody fragment G4G11 (SEQ ID No. 2), or
(ii) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein on an epitope comprising at least one of residues 65 to 74 of the amino acid sequence of human Syk tyrosine kinase protein represented by the sequence SEQ ID No. 1, or
(iii) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein and inhibiting by at least 10% the binding of antibody fragments G4G11 with human Syk tyrosine kinase protein (SEQ ID No. 1),
R1 is an unsubstituted or substituted aromatic group, or an unsubstituted or substituted heterocycle comprising at least one S, O or N atom;
R2 is an unsubstituted or substituted aromatic group, an unsubstituted or substituted heterocycle, an unsaturated or saturated carbon chain comprising an amine group, an unsaturated or saturated carbon chain comprising an unsubstituted or substituted aromatic group or an unsaturated or saturated carbon chain comprising an unsubstituted or substituted heterocycle comprising at least one S, O or N atom;
R3 is an unsubstituted or substituted phenyl, 2-pyridinyl, 3-pyridinyl or 4-pyridinyl group;
n=0 or 1; n′=0 or 1;
R4 is an unsaturated or saturated carbon chain comprising 1 to 5 carbon atoms, unsubstituted or substituted with an aromatic group;
R5 is an unsubstituted or substituted aromatic group or an unsubstituted or substituted amine group;
R6 is a hydrogen atom, an alkoxy group, an alkyl group or a halogen;
R7 is a hydrogen atom, an alkoxy group, an alkyl group or a halogen;
R8 is a hydrogen atom, an alkoxy group, an alkyl group or a halogen;
m=0, 1 or 2;
R9 is a hydrogen atom and R10 is an unsubstituted or substituted phenyl group, or R9 and R10 are part of the same unsubstituted or substituted heterocycle, or
R9 and R10 are part of the same unsubstituted or substituted aromatic group;
R11 is a hydrogen atom, an alkoxy group or an alkyl group;
R12 is a hydrogen atom, an alkoxy group or an alkyl group;
R13 is a hydrogen atom or an alkyl or alkoxy group;
R14 is a hydrogen atom or an alkyl or alkoxy group;
A is an oxygen or sulphur atom;
R15 is an unsaturated or saturated carbon chain comprising 1, 2 or 3 carbon atoms, unsubstituted or substituted by an unsubstituted or substituted aromatic group, an unsubstituted or substituted heterocycle or an amine group belonging to unsubstituted or substituted heterocycle;
R16 is a hydrogen atom, a halogen or an alkoxy group;
R17 is a hydrogen atom, an alkoxy group or an acetoxy group;

2. A compound according to claim 1, wherein said compound binds with Syk tyrosine kinase protein at a site located outside the catalytic domain thereof.

3. A compound according to claim 1, wherein said antibody or antibody fragment binds with human Syk tyrosine kinase protein on an epitope comprising at least 5 of residues 65 to 74 of the amino acid sequence of human Syk tyrosine kinase protein illustrated by SEQ ID No. 1.

4. A compound according to claim 1, wherein said three-dimensional cavity further comprises the Serine residue situated in position 9, the Glutamine residue situated in position 43, the Phenylalanine residue situated in position 51, the Isoleucine residue situated in position 66, the Glutamate residues situated in position 67 and 69, the Leucine residue situated in position 70, the Asparagine residue situated in position 71, the Glycine residue situated in position 72, the Threonine residue situated in position 73, the Tyrosine residue situated in position 74 and the Alanine residue situated in position 75 of human Syk protein, the sequence of which is illustrated by SEQ ID No. 1.

5. A compound according to claim 1 wherein said compound is used for the prevention or treatment of type I hypersensitivity reactions.

6. A compound according to claim 5, wherein said compound inhibits IgE-dependent mast cell degranulation.

7. A compound according to claim 6, wherein said compound is capable of inhibiting, by 50% in vitro, mast cell degranulation at a concentration (IC50) between 1 ng/ml and 1 mg/ml.

8. A compound according to claim 5, wherein said the metabolic pathway involving Syk is a mast cell or basophil activation pathway.

9. Molecule A compound according to claim 5, wherein said condition is allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia or an allergy to an antibiotic.

10. A compound according to claim 5, wherein said compound has no effect on the metabolic pathways involving human Syk protein (SEQ ID No. 1) other than those giving rise to mast cell degranulation and/or type I hypersensitivity reactions.

11. A compound according to claim 10, wherein said compound has no effect on the antibody response which follows immunisation by a thymus-dependent antigen or on neutrophil-dependent Syk recruitment.

12. A compound according to claim 1, wherein said metabolic pathway involving Syk is a B lymphocyte, T lymphocyte, neutrophil, eosinophil, NK cell, platelet, erythrocyte, osteoclast, epithelial cell or cancer cell activation pathway.

13. A compound according to claim 12, wherein said condition is rheumatoid arthritis, an autoimmune disease, inflammation or cancer.

14. A compound according to claim 1, wherein said compound is used in combination with a further therapeutic molecule.

15. (canceled)

16. (canceled)

17. A compound according to claim 1, wherein said compound is intended to be administered by the oral, sublingual, nasal, ocular, local, intravenous, intraperitoneal, subcutaneous route, by aerosol or by inhalation.

18. A compound according to claim 1, wherein said compound is intended to be administered to adult, child or newborn human patients.

19. A compound according to claim 1, wherein said compound is intended to be administered at doses between 0.01 mg/kg and 200 mg/kg.

20. A compound according to claim 19, wherein said equivalent compound is selected from compounds having any of the following formulae: or or or

formula (I) where R1 is a phenyl group, unsubstituted or substituted by an F or Cl atom, a methyl or ethyl group, an N,N-dimethyl-sulphonamide or two groups selected from the methyl, ethyl, hydroxy, methoxy or ethoxy groups, or a group
a furan group unsubstituted or substituted by a methyl, ethyl, hydroxyl, methoxy or ethoxy group, a thiophene group unsubstituted or substituted by a methyl, ethyl, hydroxy, methoxy or ethoxy group; R2 a group
where R21 and R22 are carbon atoms each belonging to an alkyl chain comprising 1, 2 or 3 carbon atoms, or both belonging with the nitrogen atom with which they are bound to the same unsaturated or saturated heterocycle also comprising an oxygen atom or a second nitrogen atom, or a group
where R23 and R24 are carbon atoms each belonging to an alkyl chain comprising 1, 2 or 3 carbon atoms, or both belonging with the nitrogen atom with which they are bound to the same unsaturated or saturated heterocycle also comprising an oxygen atom or a second nitrogen atom, or a group
and R3 is a non-substituted 2-pyridinyl, 3-pyridinyl or 4-pyridinyl group, a phenyl group unsubstituted or substituted by a benzoxy group, and/or by a hydroxyl group, and/or by a methyl group, and/or by an ethyl group, and/or by a propyl group, and/or by one or two Br, F or Cl atoms, and/or by one to three hydroxyl, methoxy or ethoxy groups.
formula (II) where R4 is an unsaturated or saturated carbon chain comprising 1, 2 or 3 carbon atoms; R5 is a phenyl group or a secondary amine group unsubstitued or substituted by an unsubstituted or substituted phenyl group, or by a group
R6 is a hydrogen or chlorine atom or a methyl, ethyl, hydroxyl, methoxy or ethoxy group; R7 is a hydrogen or chlorine atom or a methyl, ethyl, hydroxy, methoxy or ethoxy group; R8 is a hydrogen or chlorine atom or a methyl, ethyl, hydroxy, methoxy or ethoxy group.
the formula (III) where R9 is a hydrogen atom and the group R10 is an unsubstituted or substituted phenyl group, or the groups R9 and R10 belong to the same unsubstituted or substituted heterocycle comprising 2 nitrogen atoms and 4 carbon atoms; R11 is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group; R12 is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group; R13 is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group;
R14 is a hydrogen atom or methyl, ethyl, hydroxy, methoxy or ethoxy group;
formula (IV) where A is an Oxygen or Sulphur atom; R15 is a group
R16 is a hydrogen or chlorine atom or a methyl, ethyl, hydroxy, methoxy or ethoxy group; R17 is a methyl, ethyl, hydroxy, methoxy, ethoxy, acetoxy, methoxycarhonyl or ethoxycarbonyl group.

21. A compound according to claim 20, wherein said equivalent compound is selected from the following:

22. A compound according to claim 1, wherein the in vitro affinity of said compound for Syk protein is less than 25 μM.

23. A composition comprising a compound according to claim 1 and a pharmacologically acceptable excipient for the prevention or treatment of a condition dependent on a metabolic pathway involving Syk in humans or animals.

24. (canceled)

25. A method for identifying an organic compound having a molecular weight between 50 and 2500 Dalton binding with Syk tyrosine kinase protein and capable of inhibiting by at least 10% in vitro binding of with human Syk tyrosine kinase protein or with any of the variants thereof in animals, comprising at least the following steps:

(i) antibody fragment G4G11 (SEQ ID No. 2), or
(ii) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein on an epitope comprising at least one of residues 65 to 74 of the amino acid sequence of human Syk tyrosine kinase protein represented by the sequence SEQ ID No. 1, or
(iii) an antibody or antibody fragment which binds with human Syk tyrosine kinase protein and inhibiting by at least 10% the binding of antibody fragment G4G11 with human Syk tyrosine
kinase protein (SEQ ID No. 1),
a) screening, from a bank of candidate organic compounds having a molecular weight between 50 and 2500 Da, those liable to bind with Syk protein on the three-dimensional binding cavity on the Syk protein of a compound selected from the molecules having formula C-13, I, II, III, IV or 1 to 87 as illustrated above;
b) selecting from the compounds identified in a) those capable of inhibiting by at least 10% in vitro the binding of the antibody or antibody fragment (i) or (ii) with Syk protein.

26. (canceled)

27. (canceled)

28. (canceled)

29. (canceled)

30. (canceled)

Patent History
Publication number: 20110112098
Type: Application
Filed: Apr 8, 2009
Publication Date: May 12, 2011
Inventors: Piona Dariavach (Montpellier), Pierre Emile Ulysse Martineau (Saint Gely Du Fesc), Bruno Villoutreix (Paris)
Application Number: 12/937,189
Classifications
Current U.S. Class: The Additional Hetero Ring Is Attached Indirectly To The Morpholine Ring By An Acyclic Chain Having A Hetero Atom As A Chain Member (514/237.2); Plural Chalcogens Bonded Directly To Ring Carbons Of The Thiazole Ring (548/183); By Measuring The Ability To Specifically Bind A Target Molecule (e.g., Antibody-antigen Binding, Receptor-ligand Binding, Etc.) (506/9); 1,3-thiazole Ring Bonded Directly To The Six-membered Hetero Ring (546/270.4); The Tricyclo Ring System Consists Of A Five-membered Carbocyclic Ring Ortho Fused To Both A Six-membered Carbocyclic Ring And The Six-membered Hetero Ring (e.g., Indenopyridines, Etc.) (546/111); Five-membered Hetero Ring Consisting Of One Nitrogen And Four Carbons (544/141); Having -c(=x)-, Wherein X Is Chalcogen, Bonded Directly To The Six-membered Hetero Ring (546/156); Carbocyclic Ring Containing (544/391); Chalcogen Bonded Directly To Ring Carbon Of The Thiazole Ring (514/369); Ring Sulfur In The Additional Hetero Ring (514/342); Tricyclo Ring System Having The Six-membered Hetero Ring As One Of The Cyclos (514/290); Chalcogen Attached Directly To The Six-membered Hetero Ring By Nonionic Bonding (514/312); Nitrogen Or -c(=x)-, Wherein X Is Chalcogen, Bonded Directly To The Piperazine Ring (514/255.01)
International Classification: A61K 31/5377 (20060101); C07D 277/36 (20060101); C07D 417/14 (20060101); C07D 221/16 (20060101); C07D 413/06 (20060101); C07D 215/227 (20060101); C07D 241/04 (20060101); A61K 31/427 (20060101); A61K 31/4439 (20060101); A61K 31/435 (20060101); A61K 31/4709 (20060101); A61K 31/4965 (20060101); A61P 37/06 (20060101);