WO2004016270A1 - Utilisation d'inhibiteurs de l'itk pour le traitement de maladies induites par les mastocytes ou par les basophiles - Google Patents

Utilisation d'inhibiteurs de l'itk pour le traitement de maladies induites par les mastocytes ou par les basophiles Download PDF

Info

Publication number
WO2004016270A1
WO2004016270A1 PCT/SE2003/001274 SE0301274W WO2004016270A1 WO 2004016270 A1 WO2004016270 A1 WO 2004016270A1 SE 0301274 W SE0301274 W SE 0301274W WO 2004016270 A1 WO2004016270 A1 WO 2004016270A1
Authority
WO
WIPO (PCT)
Prior art keywords
itk
itk inhibitor
cells
ester
pharmaceutically acceptable
Prior art date
Application number
PCT/SE2003/001274
Other languages
English (en)
Inventor
Per-Olof Ericsson
Karin Kristensson
Paschalis Sideras
Peter Sjö
Original Assignee
Astrazeneca Ab
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Astrazeneca Ab filed Critical Astrazeneca Ab
Priority to AU2003251271A priority Critical patent/AU2003251271A1/en
Publication of WO2004016270A1 publication Critical patent/WO2004016270A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics

Definitions

  • the present invention relates to the treatment of mast cell- or basophil-driven conditions or diseases by inhibiting the activity of Inducible T cell kinase (hereinafter referred to as Itk).
  • Itk Inducible T cell kinase
  • Mast cells and basophils each have a complex role in acquired and innate immunity. These roles include both effector cell and, potentially, immunoregulatory activities. Mast cells and basophils display a number of similarities but they also differ in many aspects of natural history and function, as explained below.
  • Both mast cells and basophils are derived from CD34 + hematopoietic progenitor cells, such as those present in adult bone marrow.
  • Basophils like other granulocytes
  • Basophils typically mature in the bone marrow and then circulate in the peripheral blood, from where they can then be recruited into the tissues.
  • mature mast cells typically do not circulate in the blood but complete their differentiation in vascularized tissues (Galli et al. 1999).
  • basophils Under physiological conditions, basophils have a short life-span of a few days.
  • IL-3 can promote the production and survival of human basophils in vitro and can induce basophilia in vivo (Galli 2000; Valent et al. 1989; Lantz et al. 1998).
  • mast cells can be very long-lived and mast cells that are apparently mature can proliferate under certain conditions (Galli et al. 1999; Schwartz and Huff 1998).
  • Both mast cells and basophils express the ⁇ 2 form of the high-affinity receptor for IgE (the Fc ⁇ RI receptor) on their surface.
  • Both cell types can be activated to secrete diverse preformed mediators, lipid mediators (synthesized de novo), and cytokines, after cross-linking of Fc ⁇ RI-bound IgE with bivalent or multivalent antigen.
  • mast cells and basophils Upon appropriate stimulation (for example, via the Fc ⁇ RI receptor), mast cells and basophils can secrete mediators that are either preformed and granule- associated (for example, histamine, proteoglycans and neutral proteases) or are synthesized de novo (for example, leukotriene C 4 [LTC 4 ], platelet activating factor [PAF] and [in mast cells only] prostaglandin D 2 [PGD 2 ]).
  • mediators that are either preformed and granule- associated (for example, histamine, proteoglycans and neutral proteases) or are synthesized de novo (for example, leukotriene C 4 [LTC 4 ], platelet activating factor [PAF] and [in
  • mouse or human mast cells represent potential sources of many cytokines with effects in inflammation, immunity, haematopoiesis, tissue remodelling and diverse other biological processes (for example, IL-1 , IL-3, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-16, TNF- ⁇ , bFGF, VPF/VEGF, TGF- ⁇ and several C-C chemokines, including MIP-1 ⁇ and MCP-1 ).
  • the spectrum of basophil-derived cytokines appears to be more limited but includes IL-4 and IL-13 (Brunner et al. 1993; MacGlashan et al. 1994).
  • Mast cells and basophils are not only important effector cells in acute IgE-associated allergic reactions but may also contribute significantly to the expression of aspects of acquired immune responses that develop over hours (for example, late-phase reactions [LPRs]) or days to weeks (for example, chronic allergic inflammation) (Galli et al. 1999; Schwartz and Huff 1998; Metcalfe et al. 1997; Galli 2000). Mast cells and basophils may also mediate immunoregulatory functions, both through their ability to produce certain cytokines and by other mechanisms (Galli et al. 1999; Schwartz and Huff 1998; Metcalfe et al. 1997; Galli 2000; Mecheri and David 1997).
  • mast cells can contribute to leukocyte infiltration, tissue remodelling and long-term functional changes in the context of IgE-associated acquired immunity.
  • Basophils can be prominent in the leukocytic infiltrates elicited at sites of IgE-associated late-phase responses in the skin, nose and airways of humans, and may contribute to the expression of acquired immunity to certain nematodes or to the feeding of ectoparasites such as ticks.
  • mast cells and/or basophils can function as antigen-presenting cells (Mecheri and David 1997) and represent sources of co-stimulatory activity.
  • mast cells are essential for virtually all of the augmented vascular permeability and tissue swelling that are associated with IgE- dependent passive cutaneous anaphylaxis reactions (Galli et al. 1999).
  • Several findings indicate that mast cells are critical for the immediate phase of IgE-associated 'Type I' reactions also in humans (Galli et al. 1999; Schwartz and Huff 1998; Metcalfe et al. 1997; Galli 2000; Schwartz 1994).
  • the immediate reaction to cutaneous antigenic challenge is followed 4-8 hours later by persistent swelling and leukocyte infiltration (i.e. the late phase response) (Galli et al. 1999; Schwartz and Huff 1998; Metcalfe et al.
  • mast cell activation can contribute to the leukocyte infiltration associated with late phase responses.
  • the leukocytes recruited to sites of LPRs can include basophils, eosinophils, neutrophils, lymphocytes and monocytes/macrophages. All of these cells may influence the reactions by providing additional proinflammatory mediators and cytokines, with the relative importance of the various potential effector cells varying depending on the individual circumstances (Galli et al. 1999; Schwartz and Huff 1998; Metcalfe et al. 1997; Galli 2000).
  • cytokines and other mediators from mast cells and basophils has the potential to influence many aspects of the pathophysiology at sites of allergic diseases, including some of the chronic changes (for example, airway hyper-reactivity, connective tissue changes and enhanced mucus production) that are associated with these disorders (Galli et al. 1999; Schwartz and Huff 1998; Metcalfe et al. 1997; Galli 2000; Gordon and Galli 1994).
  • Studies with adoptively reconstituted mast-cell-deficient mice having genetically manipulated mast cell lines have shown that mast cells can contribute significantly to the development of at least three of the long-term features of asthma in mouse models of the disorder: allergen-induced bronchial hyper-reactivity to cholinergic stimulation (Kobayashi et al.
  • IL-4 which can be released from either T cells or basophils recruited to sites of allergic inflammation, as well as from at least some mast cells, may contribute to the local enhancement of mast cell effector function and proliferation (Bischoff et al. 1999; Lorentz et al. 2000). It has been demonstrated that not only mast cells but also basophils accumulate in nasal epithelium after allergen challenge (Klein 2000). In addition to the Th2 cells, mast cells also play an important role in initiating and maintaining the allergic response in asthma and allergic rhinitis (Holgate 2000).
  • Exposure to allergens trigger a range of cellular events in the mast cell including release of pre- formed granule-held mediators such as histamine and tryptase, synthesis and release of newly formed mediators such as PGD2 and LTC4, and the stimulation of a wide range of pro-inflammatory cytokines and chemokines such as TNF ⁇ , IL-4, IL-5, IL-6, IL-8, IL-13 and GM-CSF (Bingham and Austen, 2000).
  • the mast cell cannot only initiate an immediate reaction but can also initiate and co-ordinate the later inflammatory response, This process has also been shown to occur in seasonal allergic rhinitis (Bentley et al 2000).
  • IgE and mast cells contribute to the allergic response.
  • IgE and mast cells have been implicated both in the acute and late phases of an allergic response.
  • the T cell and in particular the Th2 cell, is thought to orchestrate the allergic responses through the production of cytokines such as IL-4, IL-5, IL-9 and IL-13.
  • cytokines such as IL-4, IL-5, IL-9 and IL-13.
  • IL-4 allergen-specific IgE
  • IL-3 regulates the eosinophil component
  • IL-3 and IL-4 will also be involved in the differentiation of basophils and mast cells.
  • Recent data implicate IL-13, and potentially IL-9, to be key players in regulating the hyper-reactivity and mucus production seen in asthmatic patients.
  • the acute-phase classical hypersensitivity reaction mediated by cross-linking of allergen specific IgE bound to the Fc ⁇ RI on mast cells and basophils.
  • This induces an immediate release of granule-associated mediators such as histamine and leukotrienes, followed by subsequent release of chemokines and cytokines involved in the recruitment of cells responsible for the late phase reactions.
  • the late phase reaction is characterized by the influx of eosinophils and activated Th2 cells, and is manifested by oedema and mucus production.
  • the late phase also triggers the influx of other inflammatory cells such as monocytes and neutrophils, which contribute to a sustained general inflammation.
  • Preventing activation of mast cells and/or basophils has for a long time been considered an attractive strategy for treatment of allergic symptoms. Such activation may occur either by IgE-dependent or IgE-independent stimuli that leads to the secretion of several mediators including histamine, heparin, pro-inflammatory cytokines and proteolytic enzymes.
  • IgE-dependent or IgE-independent stimuli that leads to the secretion of several mediators including histamine, heparin, pro-inflammatory cytokines and proteolytic enzymes.
  • a variety of different mechanisms and targets have been suggested as suitable to intervene in the Fc ⁇ RI mediated mast cell / basophil activation and degranulation process (Oliver ef a/2000). These include, among others, inhibition of the two kinases Syk and Lyn located proximally to the Fc ⁇ RI receptor.
  • Lyn mediates the phosphorylation of the immunoreceptor tyrosine-based activation motifs (ITAMs) of the ⁇ and ⁇ subunits of the tetrameric ( ⁇ 2 ) Fc ⁇ RI.
  • ITAMs immunoreceptor tyrosine-based activation motifs
  • Syk when activated, is responsible for the downstream propagation of the signal (Jouvin 1994). Data from Syk knock-out mice shows that mast cells from these mice fail to degranulate (Costello era/, 1996). However, Syk is also expressed in platelets and studies of Syk-/- mice showed that this enzyme is critical for effective blood coagulation and vascular integrity (Law et al, 1999) and hence may not provide an attractive drug target.
  • Lyn-/- mice showed accumulation of plasma cells in spleen as they age, resulting in elevated levels of serum IgM and glomerulonephritis due to the presence of immune complexes containing auto-reactive antibodies (Wang et al, 1996). Therefore, inhibiting Lyn for therapeutic purposes may present a significant challenge.
  • Mast cell stabilizers are currently being used to reduce the release of histamine and other inflammatory mediators by stabilizing mast cells, the mechanism of action has yet to be determined (Spector 1999).
  • Itk is a 72 kDa kinase expressed in the cytoplasm of T cells, NK cells, mast cells and basophils, but not in other immune cells or outside the haematopoietic system. Itk is also known as lnterleukin-2-inducible T cell kinase, Emt (expressed mainly in T cells) or Tsk (J_cell specific tyrosine kinase). Itk is a member of the Tec-family of cytosolic protein tyrosine kinases.
  • this family also includes Btk (Bruton's tyrosine kinase), Tec, Bmx, and Txk.
  • Btk Brunauer's tyrosine kinase
  • Tec Bmx
  • Txk Txk
  • Tec-family kinases have the following general structure: a N-terminal pleckstrin-homology (PH) domain, a Tec-homology domain that includes a Btk motif and one or two proline-rich (PR) motifs, a SH3 domain, a SH2 domain and a c-terminal catalytic (SH1) domain. These kinases are expressed almost exclusively in hematopoietic tissues. Tec and Bmx have also been detected in endothelial cells. The cellular distribution differs for each Tec-family member. For example, Itk is expressed by T cells, NK cells, mast cells and basophils, whereas Btk is expressed by all hematopoietic cells except T cells. Thus hematopoietic cells may express one or several Tec-family kinases. For example T cells express Itk, Tec and Txk, and mast cells express Btk, Itk and Tec.
  • the gene for Itk (the itk gene) is located on human chromosome 5q in a cluster of genes associated with a Th2 type immune response (for example, IL-4 and IL-5). No disease association has been described for the itk gene.
  • the gene for Btk has been very thoroughly studied due to its association with X-linked agammaglobulinemia (XLA). XLA patients are virtually devoid of mature B cells and their Ig levels are strongly reduced. The B cell deficiency is due to a defective expansion of the pre-B cell pool.
  • XLA X-linked agammaglobulinemia
  • Btk-/- mice, and x/ ' d mice carrying a mutation which results in the substitution of Arg with Cys at residue 28 in the Btk protein, have about half the number of B cells as compared to normal mice (Smith et al 2001 ).
  • Mast cell development is normal in Btk-/- mice, but bone- marrow derived mast cells (BMMCs) exhibit a reduction in antigen-induced histamine release.
  • BMMCs bone- marrow derived mast cells
  • T and B cells signalling through T cell receptors and B cell receptors leads to activation of Itk and Btk, respectively.
  • Downstream of Itk and Btk a number of different messengers are engaged, including scaffolding proteins (SLP-76, LAT, SLP-65), Src kinases, MAP kinases, and PI3-K. These events are followed by PLC- ⁇ activation that leads to IP3 generation and sustained Ca z+ flux, and subsequently activation of transcription factors.
  • PLC- ⁇ 1 has been suggested as a direct substrate for Itk (Miller and Berg 2002).
  • Itk (and Tec) may also mediate signalling through the CD28 co-receptor.
  • Btk is the Tec-family kinase mediating signalling from the Fc ⁇ RI.
  • No other Tec family kinase has yet been shown to have a critical role in the signalling pathways downstrem of Fc ⁇ RI. .
  • Tec-family kinases can also be regulated by PH domain-mediated plasma membrane localization, and by Src-family-mediated phosphorylation of critical tyrosine residues.
  • T cells In T cells, it is known that Itk is involved in the signalling cascade downstream of the T cell receptor (TcR), and is also associated with the co-stimulatory molecule CD28.
  • TcR T cell receptor
  • Most of current knowledge on the role of Itk in T cell signalling has been generated in Itk-deficient knockout mice (Itk-/-). Such animals have a decreased number of mature thymocytes and a reduced response to anti-CD3 stimulation, but no general T cell defect, as demonstrated by their intact response to PMA/ionomycin stimulation (Liao and Liftman 1995). From studies using Itk-/- mice, it has been proposed that Itk is required for Th2 but not Th1 cell development. This was demonstrated in the N. brasiliensis and L.
  • WO 02/50071 discloses particular thiazolyl compounds that are inhibitors of the Tec-family kinases and may be used as immunosuppressive, anti-inflammatory, anti-allergic and anti-cancer agents.
  • the compounds are described as inhibitors of Itk and other Tec-family kinases including Btk, Txk, Tec and Bmx.
  • WO 01/25220 discloses particular triazine compounds that are inhibitors of a wide range of phosphoryl transferases or kinases, including the Tec-family kinases (Bmx, Btk, Itk, Tec and Txk). The compounds are described as useful for treating a wide range of diseases.
  • Tec-family kinase inhibitors preferably inhibitors of Btk, Tec, Bmx or Txk and most preferably of Btk.
  • Itk inhibitors may be used as pharmaceutical agents for the treatment or prevention of mast cell-driven or basophil-driven conditions or diseases.
  • Itk as a target for inhibiting several key events in both acute and late phase allergic reactions common to allergic rhinitis and asthma.
  • Itk is the critical, rate-limiting enzyme that controls signalling from the Fc ⁇ RI and drives the downstream responses.
  • an Itk inhibitor should reduce both the acute and late phase responses of an allergic reaction, as seen in both rhinitis and asthma and other conditions or diseases.
  • an Itk inhibitor may be used as a pharmaceutical agent to treat either the acute phase or the late phase responses of an allergic reaction. It is also possible to treat both the acute phase and the late phase responses with a single pharmaceutical agent (the Itk inhibitor), avoiding the need for polytherapy .
  • a single drug may replace the use of two separate drugs (such as an anti-histamine drug plus a glucocorticosteroid drug).
  • a method of treating or preventing a mast cell- driven or a basophil-driven condition or disease which comprises administering a therapeutically effective amount of an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof to a human or mammal suffering from or susceptible to the condition or disease.
  • an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof for the treatment or prevention of a mast cell-driven or a basophil-driven condition or disease.
  • the Itk inhibitor (or its salt or ester) is used to inhibit mast cell responses and/or basophil responses, including activation and degranulation.
  • an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof for the treatment or prevention of a mast cell-driven condition or disease.
  • an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof in the manufacture of a medicament for the treatment or prevention of a mast cell-driven condition or disease.
  • the treatment or prevention of a condition or disease covers all forms of medical therapy, including prophylactic, diagnostic and therapeutic regimens carried out in vivo or ex vivo on humans or other mammals.
  • Prevention of a condition or disease includes reducing the risk of that condition or disease.
  • Humans or mammals susceptible to a condition or disease are those at risk of suffering from the condition or disease.
  • Prophylaxis is expected to be particularly relevant to the treatment of persons who have suffered a previous episode of, or are otherwise considered to be at increased risk of, the condition or disease in question.
  • Persons at risk of developing a particular condition or disease generally include those having a family history of the disease or condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the condition or disease.
  • Mast cell-driven and basophil-driven conditions or diseases include allergic, inflammatory, autoimmune, proliferative and hyper-proliferative diseases. Particular types and specific examples of conditions and disease are discussed below.
  • Allergic, inflammatory or auto-immune conditions or diseases of the respiratory tract include: reversible obstructive airway diseases such as asthma (for example, bronchial, allergic, intrinsic asthma, extrinsic and chronic asthma), and associated manifestations of the disease (late responses, hyper-responsiveness); farmer's lung and related diseases; fibrosis; idiopathic interstitial pneumonia; chronic obstructive airway disease (also known as chronic obstructive pulmonary disease or COPD); bronchoiectasis; cystic fibrosis; eosinophilic pneumonias; adult respiratory distress syndrome (ARDS); emphysema; and alveolitis (for example cryptogenic fibrosing alveolitis).
  • asthma for example, bronchial, allergic, intrinsic asthma, extrinsic and chronic asthma
  • associated manifestations of the disease late responses, hyper-responsiveness
  • farmer's lung and related diseases fibrosis; idiopathic interstitial pneumonia; chronic ob
  • Allergic, inflammatory or auto-immune conditions or diseases in the nose include all conditions or diseases characterised by inflammation of the nasal mucosal membrane such as acute rhinitis, allergic rhinitis, chronic rhinitis including caseosa, hypertrophic rhinitis, rhinitis purulenta, and rhinitis siccia, rhinitis medicamentosa, membranous rhinitis including croupous, fibrinous and pseudomembranous rhinitis, scrofulous rhinitis, seasonal rhinitis including rhinitis nervosa (hay fever) and vasomotor rhinitis.
  • allergic rhinitis and seasonal rhinitis including rhinitis nervosa hay fever
  • Suitable conditions or diseases in the nose also include nasal polyps and allergic manifestations of nasopharynx.
  • Allergic, inflammatory or auto-immune conditions or diseases of the eye include conjunctivitis (allergic, acute, vernal, of hay fever, chronic) and inflammation disorders of the eyelids, cornea, uveal tract and retina.
  • Allergic, inflammatory and auto-immune conditions or diseases of the gastrointestinal tract include food allergy and food intolerance, ulcerative colitis, Crohn ' s disease, irritable bowel disease, gastric ulcers, and food related allergic diseases which have symptomatic manifestations remote from the gastrointestinal tract (for example migraine, rhinitis and eczema).
  • Allergic, inflammatory or auto-immune conditions or diseases of the skin include psoriasis, atopical dermatitis, contact dermatitis/dermatitis herpetiformis, erythema nodosum, urticaria, cutaneous eosinophilias, acne, Alopecia areata, eosinophilic fasciitis dermatomyositis, photoallergic sensitivity and periodontal disease.
  • Allergic, inflammatory or auto-immune conditions or diseases of the joints and connective tissue include osteoarthritis, systemic lupus erythematosus, vasculitis, Wegener ' s granulomatosis, polyarthritis nodosa, bursitis, tendonitis, gout, Behcet's syndrome, ankylosing sponditis, Reiter ' s syndrome and psoriatic arthritis.
  • Allergic, inflammatory, and auto-immune conditions or diseases of the circulatory system include artheroma, reperfusion injury (such as angioplasty), myocardial infarction, thrombosis, and vascular and tissue damage caused by ischaemia or injury.
  • Allergic, inflammatory, and auto-immune conditions or diseases of the central nervous system include Parkinsons ' s disease, Alzheimers and other dementias, stroke and subarachnoid heamorrage.
  • Inflammatory conditions or diseases of the liver include hepatitis, cirrhosis and glomerulonenephritis.
  • Allergic, inflammatory, and auto-immune conditions or diseases of the bladder and uro- genital tract include cystitis.
  • a method for treating or preventing a reversible obstructive airway disease which comprises administering a therapeutically effective amount of an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof to a human or mammal suffering from or susceptible to the disease.
  • a reversible obstructive airway disease especially asthma
  • a method for treating or preventing rhinitis which comprises administering a therapeutically effective amount of an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof to a human or mammal suffering from or susceptible to rhinitis.
  • rhinitis includes all the various types of rhinitis discussed above. Of particular interest are allergic rhinitis and seasonal rhinitis including rhinitis nervosa or hay fever).
  • a method for treating or preventing chronic obstructive pulmonary disease which comprises administering a therapeutically effective amount of an Itk inhibitor or a pharmaceutically acceptable salt or ester thereof to a human or mammal suffering from or susceptible to chronic obstructive airway disease.
  • An Itk inhibitor for use in the invention is any compound having Itk inhibitory activity.
  • the Itk inhibitor is a direct inhibitor of the Itk enzyme.
  • the level of Itk inhibitory activity must be sufficient for the Itk inhibitor to possess useful therapeutic properties.
  • the Itk inhibitor preferably gives an IC 50 value for inhibition of Itk activity of less than 5 ⁇ M (most preferably less than 1 ⁇ M).
  • the Itk inhibitor may be a compound of any chemical structure that possesses the necessary Itk inhibitory activity.
  • suitable Itk inhibitors include the compounds described in the Examples.
  • the Itk inhibitor may be a racemate or may exist in enantiomeric or diastereoisomeric forms or mixtures thereof.
  • the Itk inhibitor is a selective Itk inhibitor (that is, an inhibitor that is more active against Itk than it is against Btk).
  • a selective Itk inhibitor for use in the invention is any compound which has Itk inhibitory activity that is significantly greater than its Btk inhibitory activity, preferably selectivity is over 50-fold against Btk (ie the Itk inhibitor gives an IC 50 value for inhibition of Btk activity in vitro which is more than 50-fold greater than its IC 50 value for inhibition of Itk activity in vitro).
  • selectivity of the Itk inhibitor is also over 10-fold (preferably over 20-fold) against Syk and/or over 10-fold (preferably over 50-fold) against Lyn. (The kinases Syk and Lyn are believed to have a regulatory role in the signalling pathway directly downstream of the Fc ⁇ RI receptor).
  • the selective Itk inhibitor gives an IC 50 value for inhibition of Itk activity of less than 5 ⁇ M (most preferably less than 1 ⁇ M) when tested in the assay described in the Examples.
  • the dose of the Itk inhibitor to be administered will depend on the compound employed, the disease being treated, the mode of administration, the age, weight and sex of the patient. Such factors may be determined by the attending physician. However, in general, satisfactory results are obtained when the compounds are administered to a human at a daily dosage of between 0.1 mg/kg to 100 mg/kg (measured as the active ingredient).
  • the Itk inhibitor may be used on its own, or in the form of appropriate pharmaceutical formulations comprising the Itk inhibitor in combination with a pharmaceutically acceptable diluent, adjuvant or carrier.
  • Particularly preferred are compositions not containing material capable of causing an adverse reaction, for example, an allergic reaction.
  • Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, "Pharmaceuticals - The Science of Dosage Form Designs", M. E. Aulton, Churchill Livingstone, 1988.
  • the Itk inhibitors may be administered topically, for example, to the lungs and/or the airways, in the form of solutions, suspensions, HFA aerosols or dry powder formulations, for example, formulations in the inhaler device known as the Turbuhaler ® ; or systemically, for example, by oral administration in the form of tablets, pills, capsules, syrups, powders or granules; or by parenteral administration, for example, in the form of sterile parenteral solutions or suspensions; or by rectal administration, for example, in the form of suppositories.
  • Dry powder formulations and pressurized HFA aerosols of the Itk inhibitor may be administered by oral or nasal inhalation.
  • the compound is desirably finely divided.
  • the finely divided compound preferably has a mass median diameter of less than 10 ⁇ m, and may be suspended in a propellant mixture with the assistance of a dispersant, such as a C 8 -C 20 fatty acid or salt thereof, (for example, oleic acid), a bile salt, a phospholipid, an alkyl saccharide, a perfluorinated or polyethoxylated surfactant, or other pharmaceutically acceptable dispersant.
  • a dispersant such as a C 8 -C 20 fatty acid or salt thereof, (for example, oleic acid), a bile salt, a phospholipid, an alkyl saccharide, a perfluorinated or polyethoxylated surfactant, or other pharmaceutically acceptable dispersant.
  • the Itk inhibitor may also be administered by means of a dry powder inhaler.
  • the inhaler may be a single or a multi dose inhaler, and may be a breath actuated dry powder inhaler.
  • a carrier substance for example, a mono-, di- or polysaccharide, a sugar alcohol, or an other polyol.
  • Suitable carriers are sugars, for example, lactose, glucose, raffinose, melezitose, lactitol, maltitol, trehalose, sucrose, mannitol; and starch.
  • the finely divided Itk inhibitor may be coated by another substance.
  • the powder mixture may also be dispensed into hard gelatine capsules, each containing the desired dose of the active compound.
  • This spheronized powder may be filled into the drug reservoir of a multidose inhaler, for example, that known as the Turbuhaler ® in which a dosing unit meters the desired dose which is then inhaled by the patient.
  • a multidose inhaler for example, that known as the Turbuhaler ® in which a dosing unit meters the desired dose which is then inhaled by the patient.
  • the active Itk inhibitor with or without a carrier substance, is delivered to the patient.
  • the Itk inhibitor may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone, and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets.
  • a carrier for example, lactose, saccharose, sorbitol, mannitol
  • a starch for example, potato starch, corn starch or amylopectin
  • a cellulose derivative for example, gelatine or polyvinylpyrrolidone
  • a lubricant for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the
  • the cores may be coated with a concentrated sugar solution which may contain for example, gum arabic, gelatine, talcum, titanium dioxide, and the like.
  • the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
  • the Itk inhibitor may be admixed with for example, a vegetable oil or polyethylene glycol.
  • Hard gelatine capsules may contain granules of the compound using either the above mentioned excipients for tablets. Also liquid or semisolid formulations of the drug may be filled into hard gelatine capsules.
  • Liquid preparations for oral application may be in the form of syrups or suspensions, for example solutions containing the Itk inhibitor, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol.
  • Such liquid preparations may contain colouring agents, flavouring agents, saccharine and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art.
  • the Itk inhibitor may also be administered in conjunction with other compounds used for the treatment of the above conditions or diseases.
  • Itk inhibitors that may be useful pharmaceutical agents, it is advantageous to screen for and select compounds that inhibit Itk activity in mast cells or basophils using appropriate in vitro or in vivo assays. In particular, it is advantageous to screen for and select compounds that have the potential to inhibit Itk activity in these cells in vivo (for example, because they are targeted to these cells).
  • Itk inhibitors may also be advantageous to develop Itk inhibitors by improving their ability to inhibit Itk activity in mast cells or basophils (for example, by developing compound features or other mechanisms that help to target the inhibitors to these cells).
  • Appropriate assays that may be useful in identifying and/or developing Itk inhibitors (particularly selective Itk inhibitors) include one or more assays selected from those described in the Examples: Itk LANCE TRF assay, Btk LANCE TRF assay, Lyn LANCE TRF assay, Syk assay, Basophil Histamine Release assay (algE induced), Basophil Leukotriene C4 (LTC4) Release assay (algE induced), Basophil lnterleukin-4 (IL-4)
  • Basophil Histamine Release assay (algE induced), Basophil Leukotriene C4 (LTC4) Release assay (algE induced), Basophil lnterleukin-4 (IL-4) Release assay (algE induced), Lung Mast Cell Histamine Release assay (algE induced).
  • Reactions were monitored at 254 nm by analytical HPLC, using a Kromasil C-18 column (150 x 4.6 mm) and a gradient (containing 0.1% trifluoroacetic acid) of 5 to 100% of acetonitrile in water at a flow rate of 1 ml/min. Evaporations of solvents were performed under reduced pressure using a rotary evaporator at a maximum temperature of 60°C. Products were dried under reduced pressure at 40 °C.
  • Compound B3 was synthesized following the protocol for the preparation of 2- arylbenzothiazole-6-carboxamides. From 2-(4-chloro-2-hydroxyphenyl)benzothiazole-6- carboxylic acid (0.059 g, 0.19 mmol) and 2-pyrrolidin-1 -ylethanamine (0.050 ml, 0.40 mmol), the title compound was isolated (0.018 g, 23%).
  • the 2-arylbenzothiazole-6-(amine-1 -ylcarbonyl) (0.2 mmol) was dissolved in 5 ml of THF. After cooling at 0°C, a 1 M solution of LiAIH 4 in THF (0.4 mmol) was added dropwise. The ice-bath was removed and the reaction mixture stirred at rt until completion (LC-MS).
  • the Itk kinase assay utilized recombinant human Itk kinase domain fused with GST (Glutathione S-Transferase).
  • the protein was expressed in High five insect cells, purified in one step on an affinity chromatography glutathione column and stored in 50 mM Tris/HCI (pH 7.6), 150 mM NaCI, 5% (w/v) mannitol, 1 mM DTT, 30% glycerol at -70°C.
  • the kinase substrate used in the assay was a biotinylated peptide derived from the Src- optimal substrate (Nair et al, J. Med.
  • test compounds or controls; 1 ⁇ l in 100% DMSO
  • Test compounds were added to black 96-well flat-bottomed plates (Greiner 655076) followed by 20 ⁇ l Itk in assay buffer and the reaction was started by adding 20 ⁇ l ATP and peptide substrate in assay buffer.
  • the assay buffer constitution during phosphorylation was: 50 mM HEPES (pH 6.8), 10 mM MgCI 2 , 0.015% Brij 35, 1 mM DTT, 10% glycerol, 160 ng/well Itk, 2 ⁇ M peptide substrate and 50 ⁇ M ATP.
  • the assay was stopped after 50 minutes (RT) by adding 150 ⁇ l ice-cold Stop solution (50 mM Tris/HCI, pH 7.5, 10 mM EDTA, 0.9% NaCI and 0.1 % BSA) together with LANCE reagents (2 nM PT66-Eu 3+ , Wallac AD0069 and 5 ⁇ g/mL Streptavidin-APC, Wallac AD0059. Both concentrations were final in stopped assay solution).
  • the plates were measured on a Wallac 1420 Victor 2 instrument with TRF settings after 1 h incubation, and the ratio (665 signal/615 signal) * 10000 was used to calculate the inhibition values.
  • IC50 values were determined using XLfit.
  • Btk LANCE TRF assay The Btk kinase assay utilized recombinant human full length Btk fused with GST
  • Glutathione S-Transferase The protein was expressed in baculo virus transfected Sf9 insect cells, purified in one step on an affinity chromatography glutathione-sepharose column and stored in 10 mM T s-HCI (pH 7.4), 150 mM NaCI, 1 mM DTT and 30 % glycerol and stored at -70°C.
  • the kinase substrate used in the assay was Btk217-229 (biotm-KKVVALYDYMPMN) comprising the Btk autophosphorylation site.
  • the assay additions were as follows Test compounds (or controls, 1 ⁇ l in 100% DMSO) were added to black 96-well flat-bottomed plates (Greiner 655076) followed by 20 ⁇ l Btk and substrate in assay buffer and the reaction was started by adding 20 ⁇ l ATP.
  • the assay buffer constitution during phosphorylation was 50 ⁇ M Tns/HCI (pH 7 4), 7.5 mM MnCI 2 , 0.002% Brij 35, 10 mM DTT, 5% glycerol, 40 ng/well Btk, 2 ⁇ M peptide substrate and 20 ⁇ M ATP.
  • the assay was stopped after 60 minutes (RT) by adding 120 ⁇ l ice-cold Stop solution (50 mM Tris/HCI, pH 7.5, 10 mM EDTA, 0 9% NaCI and 0.1% BSA) together with LANCE reagents (1 nM PT66-Eu 3+ , Wallac AD0069 and 10 nM Streptavidin-APC, Wallac AD0059. Both concentrations were final in stopped assay solution).
  • the plates were measured on a Wallac 1420 Victor 2 instrument with TRF settings after 1h incubation, and the ratio (665 s ⁇ gnal/615 s ⁇ gnal) * 10000 was used to calculate the inhibition values IC50 values were determined using XLfit
  • the Lyn kinase assay utilized recombinant N-terminal 6H ⁇ s-tagged human Lyn
  • the protein was expressed in baculo virus infected Sf9 insect cells and used as a crude cell lysate, stored at -70°C.
  • the autophosphorylation site Lyn 363-369 (biotin-EDNEYTA) from human p56 Lyn was used as kinase substrate in the asay.
  • the assay additions were as follows Test compounds (or controls, 1 ⁇ l in 100% DMSO) were added to black 96- well flat-bottomed plates (Greiner 655076) followed by 20 ⁇ l Lyn in assay buffer and the reaction was started by adding 20 ⁇ l ATP/substrate
  • the assay buffer constitution during phosphorylation was 90 mM Hepes (pH 7 4), 1.5 mM MnCI 2 , 0.09% Triton X-100, 80 ⁇ M DTT, 80 ⁇ M Na 3 VO 4 , Lyn lysate diluted 1/20000, 7 ⁇ M peptide substrate and 7.5 ⁇ M ATP.
  • the assay was stopped after 100 minutes (RT) by adding 20 ⁇ l ice-cold Stop solution (100 mM Hepes, pH 7 4, 0 1 % Triton X-100 and 10 mM EDTA) together with LANCE reagents (2 nM PT66-Eu 3+ , Wallac AD0069 and 1 16 nM Phycolink Streptavidin-APC, Prozyme PJ25S Both concentrations were final in stopped assay solution).
  • the plates were measured on a Wallac 1420 Victor 2 instrument with TRF settings after 1 h incubation, and the ratio (665 s ⁇ gnal/615 s ⁇ gnal) * 10000 was used to calculate the inhibition values IC50 values were determined using XLfit Syk assay
  • Syk kinase was cloned in baculovirus and expressed as a full-length protein in Sf9 cells (AZAP).
  • a crude enzyme extract was used as enzyme source.
  • SLP76(107- 120) Biotin-6-aminohexanoate-Ser-Phe-Glu-Glu-Asp-Asp-Tyr-Glu-Ser-Pro-Asn-Asp-Asp- s Gln-OH
  • the reaction was performed in flat bottom 384- well solid black plates (Costar) in a final volume of 20 ⁇ l, containing 50 mM Hepes pH 7.4, 0.05% Triton X-100, 0.2 mM DTT, 1 ⁇ M ATP, 3 mM MnCI 2 , 0.2 mM peptide substrate and a 5000-fold dilution of SYK insect cell extract. After 30 min incubation at room temperature the reaction was terminated by addition of 15 ⁇ l EDTA to a final concentration of 20 mM diluted in detection buffer (50 mM Hepes pH 7.4, 0.17% BSA, 0.033% Tx-100, 250 mM NaCI).
  • Basophils were obtained from human blood by density centrifugation on polymorphprep (AXIS-SHIELD). Mononuclear cells were collected from the top layer and washed in THG " buffer (0.8% NaCI, 0.24% HEPES, 0.1% D-glucose anhydrous, 0.1% Gelatine Type B, 0.1 % 1.8M NaH 2 PO 4 • 2 H 2 O, 0.1 % 2.7M KCI/ 1 L H 2 O). Basophils were purified by negative selection using MACS Basophil Isolation Kit (Miltenyi Biotech). Briefly, cells were labelled with magnetic beads in cold MACS buffer (PBS, 2 mM EDTA, and 0.5% BSA) and separated using the AutoMACS separation instrument.
  • MACS Basophil Isolation Kit MACS Basophil Isolation Kit
  • the cells were subsequently washed in RPMI 1640 supplemented with 25 mM HEPES (GIBCO-BRL) and 5% heat inactivated Fetal Calf Serum (GIBCO-BRL), and incubated with 5 ⁇ g IgE (Serotec
  • Histamine content was measured in the supernatants by addition of 20 ⁇ l HMT/ 3 H- SAM [1 ml Histamine Methyl Transferase (HMT), purified from male rat kidneys (for description see below) and 1 Mbq S-Adenosyl-L-[methyl- 3 H] methionine (Amersham Pharmacia Biotech) in 5 ml THG ++ buffer] to 100 ⁇ l sample (volume adjusted with THG ++ buffer). For total histamine release 1.5 x 10 4 boiled basophils were used.
  • HMT Histamine Methyl Transferase
  • Samples were incubated at 37°C for 60 minutes and the reaction stopped by addition of 50 ⁇ l NaOH (10 mM) followed by 400 ⁇ l toluene/isoamyl alcohol (4:1 ). Tubes were capped and vigorously shaken for 40 sec. to extract methyl histamine and subsequently centrifuged to separate the phases. To measure 3 H-methyl histamine content in the top (organic) phase, samples were transferred to LumaPlates (Packard), dried and read in 1450 MicroBeta instrument (Wallac).
  • HMT Histamine Methyl Transferase preparation
  • HMT was obtained from male rat kidneys. Briefly, the membrane was peeled off and kidneys (80 g) homogenized with 10 volumes of 0.25 M sucrose. The homogenate was centrifuged at 15000xg for 90 minutes at 0°C. The supernatant was collected and saturated to 45% with ammonium sulphate (277 mg/ ml) whilst stirring in the cold and subsequently centrifuged at 10000g for 20 min. The supernatant was collected, adjusted to 70% saturation with ammonium sulphate (171 mg/ ml) and centrifuged at 10000g for 15 min. Supernatants were discarded and pellets dissolved in 0.1 M sodium phosphate buffer pH 7.4, and dialysed against 2 x 4 L of cold 1 mM sodium phosphate buffer pH 7.4.
  • Human basophils (purified according to the protocol described for basophil histamine release assay above) were resuspended to a density of 600,000/ ml in THG " buffer. Cells (50 ⁇ l) were added to 96 well plates containing 25 ⁇ l compound (4x final concentration i.e. 4 x 10 "9 to 4 x 10 "6 M). Release was started by the addition of 10 ⁇ l of 10 times concentrated anti-human IgE to give a final dilution of 1 :10000 (Sigma code I-0632) at 37°C for 25 minutes . Cells were spun at 300xg for 5 minutes at 4°C and 50 ⁇ l supernatant sampled for the measurement of LTC4.
  • LTC4 was measured by a commercial ELISA kit ( BioTRAKTM Leukotriene C4/D4/E4 Enzyme immunoassay system RPN224). Assays were carried out as per kit instructions Briefly, 50 ⁇ l supernatant from basophil release experiments were sampled into the biotrak ELISA plate, 50 ⁇ l of anti-serum was added and incubated for 2 hours at room temperature. Subsequently 50 ⁇ l of peroxidase conjugate was then added and incubated for a further hour at room temperature. Wells were then emptied and washed four times with 300 ⁇ l wash buffer. Substrate (150 ⁇ l) was then added to each well and incubated for 30 minutes on a plate shaker at room temperature.
  • Human basophils (purified according to the protocol described for basophil histamine release assay above) were resuspended to a density of 2x10 6 / ml in HEPES-buffered RPMI 1640 with 10 % FCS and 2 mM L-glutamine.
  • Cells 50 ⁇ l were added to 96 well plates containing 25 ⁇ l compound (4x final concentration i.e. 4 x 10 "9 to 4 x 10 '6 M). Release was started by the addition of 10 ⁇ l of 10 times concentrated anti-human IgE to give a final dilution of 1.10,000 (Sigma code I-0632) at 37°C for 4 hours.
  • IL-4 was measured by a commercial ELISA kit ( BioTRAKTM, lnterleukin-4 [(h)IL-4] Human, Elisa System, code RPN2753). Assays were carried out as per kit instructions. Briefly, 50 ⁇ l of biotinylated antibody reagent was added to the BioTRAKTM plate. Supernatant from basophil release experiments were sampled into the plate. This was then incubated at room temperature for two hours at room temperature. Wells were then emptied and washed three times with 300 ⁇ l wash buffer.
  • Streptavidin-HRP conjugate 100 ⁇ L was then added and incubated at room temperature for 30 minutes. Wells were then emptied and washed three times with wash buffer. Substrate (100 ⁇ l) was then added to each well and incubated for 30 minutes in the dark at room temperature. Stop solution (100 ⁇ l 0.18 M sulphuric acid) was then added and the OD determined at 450nM. IL-4 released was calculated by comparison to a standard curve set up as in the kit, but using release buffer (RPMI 1640).
  • Human lung tissue was obtained from from lung resection patients The tissue (usually 5- 20 g) was chopped on arrival with scissors into strips of approximately 5 mm by 20 mm, and stored overnight in 50ml RPMI 1640 with 5% FCS and Penicillin streptomycin solution (Gibco, 10,000 ⁇ g /mL of Streptomycin sulphate and 10,000 Units/mL of penicillin G ) 1 ml/100 ml RPMI The following day the tissue was finely chopped using a mechanical tissue chopper (Mcllwain tissue chopperTM) into small cubes ( ⁇ 1 mm 3 ) and washed through nylon gauze with 1 L THG (Tyrodes-HEPES-gelatin pH 7.4).
  • Mcllwain tissue chopperTM mechanical tissue chopper
  • the washed tissue was then suspended in 10X volume (w/v) of digestion buffer (THG containing 1 mM MgCI 2 , 5 mg/ml bovine serum albumin, 0 01 mg/ml DNase 1 type IV (Sigma), and 1 mg/ml collagenase type 1 A (Sigma)) and incubated for 30 minutes at 37°C Tissue was further disrupted by passing through a 50 ml syringe (with tip removed) 10 times, and incubated for a further 30 minutes in the digestion buffer
  • the digestion buffer was then diluted 1 :1 with THG Isolated cells were harvested by passing through 120 ⁇ m nylon gauze and centrifugation at 300xg for 5 minutes at 20°C The cells were washed 3 x in THG After the final wash the cells were resuspended in 50 ml RPMI-1640 containing 5% FCS 1 % glutamine and 1 % Penicillin /streptomycin and stored overnight at 4°C Mast cells were purified
  • Beads (300 ⁇ l) were incubated overnight at 4°C with 50 ⁇ l of anti-human CD117 (Pharmingen) The beads were then washed 4 X in THG and collected using a Dynal magnet The prepared cells were then centrifuged at 300g for 5 minutes at 4°C and resuspended in 2 mL of THG and pre-treated Dynal beads added (10 beads/mast cell). The cells and beads were mixed and left for 1 hour on a roller mixer at 4°C At the end of this incubation the cells were diluted with a further 8 ml of THG and cells attached to beads were isolated using the Dynal magnet, and washed 5 x in THG (10 mis) .
  • the beads were then removed from the cells by incubation with 200 ⁇ l THG containing 20 ⁇ l of releasing buffer (supplied with Dynal CELLectionTM kit) for 15 minutes at 37°C. Cells were aspirated in a pipette 5 times and detached beads removed using the Dynal magnet Purified mast cells were collected in the supernatant The beads were washed in THG (200 ul) a further 3 times to maximise recovery of mast cells The supernatants were pooled and centrifuged at 300xg 5 minutes at room temperature. The cells were then washed in 10 ml THG and resuspended into 1 ml RPMI-1640 with 5%FCS.
  • mast cells were counted in a haemocytometer (20 ⁇ l cells + 180 ⁇ l Kimura's stain). Pure mast cells (1 ml in RPMI 1640 containing 5% FCS) were sensitised by incubating with 5 ⁇ g/ml human IgE (Serotec) for 1 hour at 37°C. Cells were then diluted to 10 ml with THG and centrifuged, 300xg for 5 minutes at room temperature. The pellet was washed in 10 ml THG " and cells recovered by centrifugation, 300xg for 5 minutes at room temperature. The pellet was resuspended in 1 ml THG ++ .
  • Total histamine was determined by freeze-thawing 50 ⁇ l of mast cells with 150 ⁇ l distilled water and microwaving for 30 seconds. 25 ⁇ l of this is sampled for Histamine analysis. Histamine content was determined according to the same protocol as for the basophil histamine release assay described above.
  • the structurally diverse compounds A5, A14, B1 , B3, C1 , C7, D5 and D6 were tested for their ability to inhibit histamine release from primary human basophils isolated from blood obtained from healthy volunteers. The basophils were stimulated with anti-lgE. The compounds were also profiled to determine their capability to inhibit the catalytic activity of the protein tyrosine kinases Btk, Syk, and Lyn which are all present in mast cells and important in the signalling downstream of the Fc ⁇ RI receptor.
  • Results show that inhibition of anti-lgE induced histamine release was dependent on Itk inhibition.
  • Compound C7 is a selective Btk inhibitor which does not have the preferred Itk inhibitory activity (Itk IC 50 is not less than 5 ⁇ M). Compound C7 does not inhibit histamine release. Each of the other compounds is a good Itk inhibitor (Itk IC 50 less than 5 ⁇ M) and each inhibits histamine release.
  • Compounds A5, B1 , B3, D5 and D6 are selective Itk inhibitors (Btk IC 50 >50 ⁇ M compared to Itk IC 50 ⁇ 5 ⁇ M).
  • Compounds A5, A14, B1 , B3, D5 and D6 are more than 10-fold selective over the kinases Syk and Lyn.
  • Compound C1 is a non- selective Itk inhibitor which is more active against Btk.
  • Example 2 Two of the compounds tested in Example 1 were tested for their ability to inhibit IL-4 and/or LTC4 release from anti-lgE stimulated human primary basophils. Results are shown in the table below. Note that the IL-4 release was measured in the presence of 10% serum, which reduces the effect of the compound due to compound binding to the serum proteins.
  • Example 2 Two of the compounds tested in Example 1 were tested for their ability to inhibit histamine release from human lung mast cells prepared from human lung tissue. These compounds were found to inhibit histamine release from primary human lung mast cells (results shown in the table below).

Abstract

La présente invention concerne le traitement de maladies ou d'états pathologiques induits par les mastocytes ou par les basophiles par inhibition de l'activité de la kinase des lymphocytes T inductible (Itk). Les maladies ou états pathologiques particuliers pouvant être traités incluent l'asthme, la rhinite et la BPCO.
PCT/SE2003/001274 2002-08-14 2003-08-13 Utilisation d'inhibiteurs de l'itk pour le traitement de maladies induites par les mastocytes ou par les basophiles WO2004016270A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003251271A AU2003251271A1 (en) 2002-08-14 2003-08-13 USE OF Itk INHIBITORS FOR THE TREATMENT OF MAST CELL-DRIVEN OR BASOPHIL-DRIVEN DISEASES

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SE0202464A SE0202464D0 (sv) 2002-08-14 2002-08-14 Use of compounds
SE0202464-4 2002-08-14

Publications (1)

Publication Number Publication Date
WO2004016270A1 true WO2004016270A1 (fr) 2004-02-26

Family

ID=20288752

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2003/001274 WO2004016270A1 (fr) 2002-08-14 2003-08-13 Utilisation d'inhibiteurs de l'itk pour le traitement de maladies induites par les mastocytes ou par les basophiles

Country Status (3)

Country Link
AU (1) AU2003251271A1 (fr)
SE (1) SE0202464D0 (fr)
WO (1) WO2004016270A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005102325A1 (fr) * 2004-04-20 2005-11-03 Ab Science Utilisation d'inhibiteurs de c-kit pour le traitement de troubles musculaires inflammatoires comprenant la myosite et la dystrophie musculaire
WO2013153539A1 (fr) 2012-04-13 2013-10-17 Glenmark Pharmaceuticals S.A. Composés tricycliques à titre d'inhibiteurs de kinases tec
CN103804381A (zh) * 2012-11-06 2014-05-21 韩冰 一类治疗缺血性脑损伤的化合物及其用途
WO2018011138A1 (fr) 2016-07-11 2018-01-18 Kancera Ab Dérivés de 2-phenylimidazo[4,5-b]pyridin-7-amine utilisés comme inhibiteurs de l'activité de la tyrosine kinase mammifère ror1
US10550113B2 (en) 2015-02-02 2020-02-04 Kancera Ab 2-phenyl-3H-imidazo[4,5-B]pyridine derivates useful as inhibitors of mammalian tyrosine kinase ROR1 activity
US11660303B2 (en) 2016-07-11 2023-05-30 Kancera Ab 2-phenylimidazo[4,5-b]pyridin-7-amine derivates useful as inhibitors of mammalian tyrosine kinase ROR1 activity

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999024035A1 (fr) * 1997-11-10 1999-05-20 Bristol-Myers Squibb Company Composes de benzothiazole utilises comme inhibiteurs de la proteine tyrosine-kinase
WO2001025220A1 (fr) * 1999-10-07 2001-04-12 Amgen Inc. Inhibiteurs de triazine kinase
WO2001047922A2 (fr) * 1999-12-24 2001-07-05 Aventis Pharma Limited Azaindoles
WO2002050071A1 (fr) * 2000-12-21 2002-06-27 Bristol-Myers Squibb Company Inhibiteurs thiazolyl des tyrosine kinases de la famille tec

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999024035A1 (fr) * 1997-11-10 1999-05-20 Bristol-Myers Squibb Company Composes de benzothiazole utilises comme inhibiteurs de la proteine tyrosine-kinase
WO2001025220A1 (fr) * 1999-10-07 2001-04-12 Amgen Inc. Inhibiteurs de triazine kinase
WO2001047922A2 (fr) * 1999-12-24 2001-07-05 Aventis Pharma Limited Azaindoles
WO2002050071A1 (fr) * 2000-12-21 2002-06-27 Bristol-Myers Squibb Company Inhibiteurs thiazolyl des tyrosine kinases de la famille tec

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005102325A1 (fr) * 2004-04-20 2005-11-03 Ab Science Utilisation d'inhibiteurs de c-kit pour le traitement de troubles musculaires inflammatoires comprenant la myosite et la dystrophie musculaire
WO2013153539A1 (fr) 2012-04-13 2013-10-17 Glenmark Pharmaceuticals S.A. Composés tricycliques à titre d'inhibiteurs de kinases tec
CN103804381A (zh) * 2012-11-06 2014-05-21 韩冰 一类治疗缺血性脑损伤的化合物及其用途
US10550113B2 (en) 2015-02-02 2020-02-04 Kancera Ab 2-phenyl-3H-imidazo[4,5-B]pyridine derivates useful as inhibitors of mammalian tyrosine kinase ROR1 activity
WO2018011138A1 (fr) 2016-07-11 2018-01-18 Kancera Ab Dérivés de 2-phenylimidazo[4,5-b]pyridin-7-amine utilisés comme inhibiteurs de l'activité de la tyrosine kinase mammifère ror1
US11008318B2 (en) 2016-07-11 2021-05-18 Kancera Ab 2-phenylimidazo[4,5-b]pyridin-7-amine derivates useful as inhibitors of mammalian tyrosine kinase ROR1 activity
US11660303B2 (en) 2016-07-11 2023-05-30 Kancera Ab 2-phenylimidazo[4,5-b]pyridin-7-amine derivates useful as inhibitors of mammalian tyrosine kinase ROR1 activity

Also Published As

Publication number Publication date
SE0202464D0 (sv) 2002-08-14
AU2003251271A1 (en) 2004-03-03

Similar Documents

Publication Publication Date Title
US11649224B2 (en) IRE1 small molecule inhibitors
CN110522731B (zh) 作为抗炎化合物的芳族杂环化合物
EP3119765B1 (fr) Inhibiteurs de syk de type hétéroaryle
CN101932571B (zh) 具有crth2拮抗活性的化合物
RU2743170C2 (ru) Замещенные индазолы, пригодные для лечения и предупреждения аллергических и/или воспалительных заболеваний у животных
JP5765866B1 (ja) p38MAPキナーゼ阻害剤としての1−ピラゾリル−3−(4−((2−アニリノピリミジン−4−イル)オキシ)ナフタレン−1−イル)尿素
EP2885284B1 (fr) Urees pyrazolyles en tant qu'inhibiteurs de kinase
KR101959194B1 (ko) 신규한 피페리디노-디하이드로티에노피리미딘 설폭사이드 및 copd 및 천식을 치료하기 위한 이들의 용도
EP2981534B1 (fr) Inhibiteurs de kinase à base de n-alkyl pyrazoles
TWI808083B (zh) Jak抑制劑化合物之結晶型式
JP2011526295A (ja) 5員および6員複素環化合物
EP2643321A1 (fr) Dérivés d'imidazo[1,2-b]pyridazine et d'imidazo[4,5-b]pyridine en tant qu'inhibiteurs des jak
JP2014528403A (ja) p3iMAPキナーゼ阻害剤としてのl−ピラゾリル−3−(4−((2−アニリノピリミジン−4−イル)オキシ)ナフタレン−1−イル)尿素
TW201014841A (en) Tri-substituted pyrimidine compounds and their use as PDE10 inhibitors
CN103502239B (zh) 可用作脾酪氨酸激酶(syk)抑制剂的吡啶基-和吡嗪基-甲基氧基-芳基衍生物
US11446301B2 (en) Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
KR20110022611A (ko) 베타2-아드레날린수용체 활성의 조절을 위한, 4-히드록시-2-옥소-2,3-디히드로-1,3-벤조티아졸-7-일 화합물을 포함하는 제약 조성물
TW201808945A (zh) 磷脂酸肌醇3-激酶γ的新穎抑制劑
JP2008529995A (ja) Atm阻害剤
WO2004016270A1 (fr) Utilisation d'inhibiteurs de l'itk pour le traitement de maladies induites par les mastocytes ou par les basophiles
US11434239B2 (en) Aza-dihydro-acridone derivatives
ES2845230T3 (es) Derivados de pirazol[1,5-a]pirimidina como inhibidores de quinasa JAK
EP3837251B1 (fr) Agents inhibiteurs d'ask1

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP