CA2569406A1 - Methods for treating mast cell disorders - Google Patents

Methods for treating mast cell disorders Download PDF

Info

Publication number
CA2569406A1
CA2569406A1 CA002569406A CA2569406A CA2569406A1 CA 2569406 A1 CA2569406 A1 CA 2569406A1 CA 002569406 A CA002569406 A CA 002569406A CA 2569406 A CA2569406 A CA 2569406A CA 2569406 A1 CA2569406 A1 CA 2569406A1
Authority
CA
Canada
Prior art keywords
quinazolin
methyl
purin
ylamino
phenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002569406A
Other languages
French (fr)
Inventor
Joel S. Hayflick
Noah Pefaur
Kamal D. Puri
William Tino
Toshi Kawakami
Yuko Kawakami
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Icos Corp
La Jolla Institute for Allergy and Immunology
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2569406A1 publication Critical patent/CA2569406A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

The present invention provides methods of inhibiting mast cell activity by administering a selective inhibitor of phosphoinositide 3-kinase delta (P13K.delta.). The invention also provides methods for treating or preventing a condition associated with undesirable mast cell activity in an individual comprising administering an effective amount of a selective P13K.delta.
inhibitor.

Description

METHODS FOR TREATING MAST CELL DISORDERS

This application claims the priority benefit of U.S. Provisional Patent Application No. 60/576,947, filed June 4, 2004, incorporated herein by reference.

FIELD OF THE INVENTION

The present invention relates to methods and compounds for modulating mast cell activity by inhibiting PI3K5. Such mast cell activity includes, but is not limited to, mast cell degranulation, mast cell migration, mast cell proliferation, and the expression and secretion of cytokines, chemokines, and growth factors by mast cells. As such, the methods and compounds of the invention may be used to treat or prevent conditions associated with such mast cell activity that is undesirable.

BACKGROUND OF THE INVENTION

Phosphoinositide 3-kinase (P13K) is a signaling enzyme that plays key roles in many cellular activities, including cellular growth, remodeling, and apoptosis [Wymann and Pirola, Biochem Biophys Acta. 1998;1436:127-150; Anderson et al., J
Biol Chem. 1999;274:9907-9910; Rameh et al., JBiol C12em. 1999;274:8347-8350;
Cantrell, JCell Sci. 2001;114:1439-1445; Coelho and Leevers, JCell Sci.
2000;113:2927-2934; Vanhaesebroeck et al., Ann. Rev Biochem. 2001;70:535-602;
Northcott et al., Circ Res.91:360-369 (2002); Yang et al., Ain JPhysiol Heart Circ Physiol. 280:H2144-H2152 (2001); Komalavilas, et al., JAppl Physiol. 91:1819-(2001)], P13K also plays roles in many other cellular processes, such as malignant transformation, growth factor signaling, inflammation, and immunity. See Rameh et al., J. Biol Chetn, 274:8347-8350 (1999) for a review. Such diverse activities may be attributed at least in part to PI3K's lipid and protein kinase activity.
Cloning of the catalytic subunits of PI3-kinase led to organizing the multi-gene family into three main classes based on their substrate specificity, sequence homology and regulation.
Class I P13-kinases are the most extensively investigated class and contain two subunits, one of which plays primarily a regulatory/adaptor role (p85a, (3, p55y or p101 isoform) and the other that maintains the catalytic role of the enzyme (pl 10a, 0, 8, or y isoform) [Wymann and Pirola, supra; Anderson et al., supra; Rameh et al., supra; Cantrell, supra; Coelho and Leevers, supra; Vanhaesebroeck et al., supra.]
Identification of the p1108 isoform of P13-kinase is described in Chantry et al. [JBiol Chem, 272:19236-41 (1997)]. It was observed that the human p 1105 isoform is expressed in a tissue-restricted fashion. It is expressed at high levels in lymphocytes and lymphoid tissues. Details concerning the pl lOb isoform also can be found in U.S. Patent Nos. 5,858,753; 5,822,910; and 5,985,589, each incorporated herein by reference. See also, Vanhaesebroeck et al., Proc Natl Acad Sci USA, 94:4330-5 (1997), and International Publication No WO 97/46688.

PI3Ks achieve intracellular signaling at least in part by catalyzing the addition of a phosphate group to the inositol ring of phosphoinositides [Wymann, et al., supra]. One target of these phosphorylated products is the serine/threonine protein kinase B(PKB or Akt). Akt subsequently phosphorylates several downstream targets, including the Bcl-2 family member Bad and caspase-9, thereby inhibiting their pro-apoptotic functions [Datta et al:, Cell 91:231-41 (1997); Cardone et al., Science 282:1318-21 (1998)]. Akt has also been shown to phosphorylate the forkhead transcription factor FKHR [Tang et al., J. Biol. Chem., 274:16741-6 (1999)].
In addition, many other members of the apoptotic machinery as well as transcription factors contain the Akt consensus phosphorylation site [Datta et al., supra].

Phosphorylation of Akt has been widely used as an indirect measure of Class I P13-kinase activity in multiple cell types, including endothelial cells, [Shiojima, et al., Circ. Res., 90:1243-1250 (2002); Kandel et al., Exp. Cell Res., 253:210-229 (1999); Cantley et al., Science 296:1655-1657 (2002)]. P13K
activity is required for growth factor mediated survival of various cell types [Fantl et al., Ann.
Rev. Biochem. 62:453-81 (1993); Datta et al., Genes & Dev. 13:2905-27 (1999)].
The nonselective phosphoinositide 3-kinase (P13K) inhibitors, LY294002 and wortmannin, have been shown to enhance destruction of tumor vasculature in irradiated endothelial cells [Edwards et al., Cancer Res.
62:4671-77 (2002)] and partially inhibit mast cell degranulation [Tkaczyk et al., JBiol Chena.
278:48474-84 (2003)]. LY294002 and wortmannin do not distinguish among the four members of class I PI3Ks, however. For example, the IC50 values of wortmannin against each of the various class 1 PI3Ks are in the range of 1-10 nM.
Similarly, the IC50 values for LY294002 against each of these PI3Ks is about 1 M [Fruman, et al., Ann. Rev. Biochem. 67:481-507 (1998)]. These inhibitors are not only nonselective with respect to class I PI3Ks, but are also potent inhibitors of DNA dependent protein kinase, FRAP-mTOR, smooth muscle myosin light chain kinase, and casein kinase [Hartley, et al., Cell 82:849-56 (1995); Davies, et al., Biochem. J. 351:95-105 (2000);
Brunn, et al., EMBO J. 15:5256-67 (1996)].

Because p110a, p110(3, p110y, and p110S isoforms are expressed differentially by a wide variety of cell types, the administration of nonselective P13K
inhibitors such as LY294002 and wortmannin often affect cell types that may not be targeted for treatment. Therefore, the effective therapeutic dose of such nonselective inhibitors would be expected to clinically unusable because otherwise non-targeted cell types will likely be affected, especially when such nonselective inhibitors are combined with other immunomodulatory therapies.

Mast cells play'diverse and significant roles. For example, mast cells are involved in mediating first line immune responses of the innate immune system seen in response to allergens or parasitic or bacterial infections. Mast cells also contribute to activation and recruitment of other inflammatory cells, such as neutrophils and T cells, to bring about second line immune responses required for an adaptive immune response. CD34+ mast cells circulate in the blood as committed precursor cells and fully mature in specific tissue sites. Mast cell development and maturation requires mast-cell growth factor, also known as stem cell factor (SCF), steel, or KIT ligand [Gurish et al., J. Exp. Med. 194:F1-F5 (2001)]. The interaction of KIT receptor with its ligand drives mast cell proliferation and differentiation (Feger et al., Ibat. Arch. Allergy Immunol. 127:110-14 (2002)].

Mast cells are activated through crosslinking of the high affinity FcERI
IgE receptor on the cell surface by antigen-bound IgE, and to a lesser extent through crosslinking of the FcyRI receptor by IgG [Tkaczyk et al., Int Arch Allergy Imnaunol.
133:305-15 (2004)]. Activation through FcsRI is typically seen in acute allergic reactions and other types of hypersensitivity reactions, leading to the stimulation of additional immune cells and a full blown immune response.

Mast cells contain metachromatic granules which store a variety of inflammatory mediators that are released upon mast cell activation. These mediators include: histamine and serotonin; prostaglandin D2; proteolytic enzymes, such as tryptase that can destroy tissue or cleave complement components or coagulation components; heparin or chondroitin sulfate, which are anticoagulants;
chemotactic factors, such as eosinophil chemotactic factor of anaphylaxis (an important regulator of eosinophil function) and neutrophil chemotactic factor. During mast cell activation, these mediators are released into the cellular environment causing acute and immediate immune responses such as vascular permeability and recruitment of lymphocytes. For example, tryptase levels rise within 1 hour and remain elevated for 4-6 hours, while histamine levels peak at approximately 5 minutes and decline rapidly within fifteen minutes. Histamine release causes dilation of blood vessels leading to fluid leak into the surrounding tissues, causing many initial symptoms of allergic reactions. Release of histamine and other mediators also leads to airway constriction, edema, vascular congestion and inflammatory cell recruitment characteristic of allergic reactions and asthma [Djukanovic et al., Clin Exp Allergy. 26 Supp13:44-51 (1996)].

Mast cell activity is necessary and desirable in healthy individuals.
Unwanted mast cell activity, or excessive proliferation of otherwise normal or abnormal mast cells, may be a component of a wide variety of disease states and/or their symptoms, however. In such instances, it is often desirable, from a therapeutic or preventative standpoint, to reduce or eliminate mast cell activity and/or proliferation. For example, numerous immune mediated diseases involve the release by mast cells of cytokines, chemokines and other factors. Cytokines, chemokines, and other factors recruit additional immune cells such as lymphocytes, including neutrophils and T cells, to sites of inflammation. This may lead to numerous immune mediated diseases. For example, mast cell activity such as degranulation and tryptase protein have recently been localized to cerebrospinal fluid of patients with multiple sclerosis, an autoimmune disease typically thought to be mediated by T cell activity [Rozniecki et al., Ann Neurol. 37:63-66 (1995)]. Additionally, mast cell deficient mice (W/W ) induced to develop an experimental model of multiple sclerosis demonstrate delayed development of MS-like symptoms [Secor et al., J. Exp.
Med.
191:813-22 (2000)].

Rheumatoid arthritis is an autoimmune disease characterized by chronic inflammation of the joints and the presence of inflammatory cells in the synovial fluid of the joints, leading to a painful and debilitating disease.
Mice lacking mast cells show resistance to induction of arthritis-like symptoms after infusion of antibodies "to a cytoplasmic enzyme [Lee et al., Science 297:1689-1693 (2002)]. Mast cells accumulate in the extremities of mice affected by collagen-induced arthritis and 5 degranulate during the disease [Woolley et al., Artlaritis Res. 2:65-74 (2000)], indicating that mast cells may play a role in mediating inflammation and recruiting additional cells to the joints of patients suffering from rheumatoid arthritis.

A significant population of mast cells resides in the skin. Bullous pemphigoid, an autoimmune disease of the skin exhibiting autoantibodies to cell junction proteins has also been shown to depend on mast cell activation. [Chen et al., J Clin Invest. 108:1151-58 (2001)]. W/W' mice deficient in mast cells did not develop bullous pemphigoid, although autoantibodies and complement proteins were present in the skin similar to normal mice, W/W' mice lacked neutrophil recruitment to the skin. Also, evidence of mast cell mediators have been detected in patients with bullous pemphigoid [Wintroub et al., New Eng. J. Med. 298:417-21 (2001)].

Other immune disorders in which rriast cells are thought to play a role include Sjogren's syndrome [Konttinen et al., Rheumatollnt. 19:141-7 (2000)], chronic urticaria [Napoli et al., Curr Allergy Asthma Rep. 1:329-36 (2001)], thyroid eye disease [Ludgate et al., Clin Exp Irnmunol. 2002 127:193-8. (2002)], vasculitis [Kiely et al., Jlmmunol. 159:5100-6 (1997)] and peritonitis [Malavyn et al., Nature 381:77-80 (1996)].

Another broad category of mast cell associated condition or disorder is termed mast cell disease, or mastocytosis. Mastocytosis encompasses a heterogeneous group of clinical disorders characterized by the proliferation and accumulation of mast cells in a variety of tissues, most often the skin, but also in the skeletal, hematopoietic, gastrointestinal, cardiopulmonary, and central nervous systems. Mastocytosis is characterized by excess proliferation of mast cells, distributed in a predictable pattern throughout the skin (e.g., cutaneous mastocytosis and urticaria pigmentosa), bone marrow, gastrointestinal tract, lymph nodes, liver and spleen [Brockow et al., Curr. Opin. Allergy Clin. Inamunol. 1:449-54 (2001)].
Mastocytosis is classified as either familial or sporadic, the latter being further subdivided into either cutaneous or systemic. Systemic mastocytosis is still further classified into indolent (chronic) mastocytosis and aggressive mastocytosis, as well as mast cell leukemia. Types of mastocytosis also emerge which have an associated hematologic disorder (AHD) (Brockow et al., supra). Cutaneous mastocytosis (CM) demonstrates typical clinical and histological skin lesions and absence of definitive signs (criteria) of systemic involvement. Most patients with CM
are children and present with maculopapular cutaneous mastocytosis (for example, urticaria pigmentosa, UP). Other less frequent forms of CM are diffuse cutaneous mastocytosis (DCM) and mastocytoma of skin.

Systemic mastocytosis (SM) is commonly seen in adults and defined by multifocal histological lesions in the bone marrow (almost ubiquitously expressed, a major criteria of diagnosis) or other extracutaneous organs together with cytological and biochemical signs (minor criteria) of systemic disease (SM-criteria).
Aggressive systemic mastocytosis is characterized by impaired organ-function due to infiltration of the bone marrow, liver, spleen, GI-tract, or skeletal system, by pathologic mast cells (MC). Mast cell leukemia is a'high-grade' leukemic disease defined by increased numbers of MC in bone marrow smears (greater than or equal to 20%) and peripheral blood, absence of skin lesions, multiorgan failure, and a short survival. In typical cases, circulating MC amount to greater than or equal to 10% of leukocytes (classical form of MCL). Mast cell sarcoma is a unifocal tumor that consists of atypical MC and shows a destructive growth without (primary) systemic involvement.
This high-grade malignant MC disease has to be distinguished from a localized benign mastocytoma in either extracutaneous organs (extracutaneous mastocytoma) or skin.

Additionally, mutations in the KIT receptor leading to mast cell hyper-proliferation have been found in patients with acute myeloid leukemia (AML) [Beghini et al., Cancen Genet Cytogenet. 119:26-31 (2000)], chronic myelogenous leukemia [Cairoli et al, Hematol J. 5:273-5 (2004)], chronic myelomonocytic leukemia [Sotlar et al., Leuk Res. 26:979-84 (2002)], germ cell tumors [Sakuma et al., Cancef- Sci. 94:486-91 (2003)], and gastrointestinal stromal tumors (GISTs) [Heinrich et al., J. Clin. Ottcol. 20:1692-1703. (2002)].

While some specific treatments for conditions associated with undesirable mast cell activity, for example allergy and asthma, have been developed, treatment regimens for mast cell related conditions typically employ non-specific treatment regimens developed for other proliferative or immune disorders (e.g., histamine receptor blockers, prostaglandin blockers, steroids), resulting in incomplete treatment, treatments which are not effective, or treatments that cause numerous immunosuppressive side-effects.

Further, a significant drawback to many therapies that may be used to treat conditions associated with undesirable mast cell activity is the non-specific inhibition of many cellular tyrosine kinases in mast cells and other cell types that are targeted by the treatment. For instance, two kinase inhibitor therapeutics for treating mast cell proliferative disorders were originally developed to inhibit kinases such as platelet-derived growth factor receptor (PDGF-R), vascular endothelial growth factor receptor (VEGFR), or the Bcr/Abl mutation. These potential therapeutics proved ineffective at treating all forms of mastocytosis. Thus, there remains a need in the art to develop effective therapeutics that more specifically target kinases involved in mediating mast cell activity.

Therefore, an important and significant goal is to develop and make available safer and more effective methods of treating and preventing disorders associated with allergy and mast cell related disorders, and to provide therapies which facilitate clinical management and continued patient compliance. The present invention addresses this and other needs.

SUMMARY OF THE INVENTION

The present invention provides methods for effectively treating or preventing a condition, and/or a symptom of a condition, associated with or caused, at least in part, by undesirable mast cell activity. The methods of the invention are particularly useful in treating or preventing conditions (or symptoms associated with conditions) mediated by immunoglobulin receptor cross-linking on mast cells.

In one embodiment, the invention provides a method for inhibiting an activity of mast cells, comprising administering to an individual a selective inhibitor of phosphoinositide 3-kinase delta (PI3K5) in an amount effective to inhibit mast cell activity. In one aspect, the mast cell activity being inhibited is mast cell migration, mast cell proliferation, mast cell degranulation, or expression of or secretion of cytokines, chemokines, or growth factors from mast cells. In a further aspect, the cytokine is TNF-a. In a related aspect, the cytokine is IL-6. In a related aspect, the chemokine being inhibited is eotaxin, MIP1-a, MIP1-0, MDC-1, MCP-1, or lymphotactin.

In a related embodiment, the invention provides methods of reducing or preventing lymphocyte infiltration to a site of inflammation in a condition associated with undesirable mast cell activity comprising the step of administering to an individual a selective inhibitor of phosphoinositide 3-kinase delta (PI3K8) in an amount effective to reduce or prevent lymphocyte infiltration to said site of inflammation in an amount effective to reduce lymphocyte recruitment signaling by 1& mast cells -in said individual. A condition associated with undesirable mast cell activity is any condition caused by or involving the underlying effects of any ,undesirable mast cell activity.

In another embodiment, the invention provides methods for treating or preventing a condition associated with undesirable mast cell activity in an individual, comprising the step of administering a selective inhibitor of phosphoinositide 3-kinase delta (PI3K6) in an amount effective to treat or prevent a condition associated with undesirable mast cell activity. In one aspect, in order to treat or prevent a condition associated with undesirable mast cell activity, the selective PI3K5 inhibitor inhibits mast cell activity. Examples of such mast cell activity include mast cell migration, mast cell proliferation, mast cell degranulation, or expression or secretion of cytokines, chemokines, or growth factors from mast cells.

The methods of the invention encompass treating or preventing conditions (or symptoms associated with conditions) mediated by immunoglobulin receptor cross-linking on mast cells "Mediated by immunoglobulin crosslinking"
or "Ig-mediated" refers to the ability of Ig bound to receptors on mast cells to initiate, or facilitate, a condition associated with undesirable mast cell activity.
Immunoglobulins which activate mast cells include IgG and IgE. In one embodiment, the condition associated with undesirable mast cell activity is an IgE-mediated condition. In an alternative embodiment, the condition is an IgG-mediated condition.
In a further embodiment, the condition is mediated by other stimuli such cytokines, chemokines or other growth factors.
Conditions, and symptoms of conditions amenable to treatment or prevention by methods according to the invention include, but are not limited to, asthma, allergic reactions, or autoimmune diseases.

In one aspect, the allergic reaction is type I hypersensitivity, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, or allergic asthma. Type I
hypersensitivity reactions are reactions in which antigens (allergens) combine with specific IgE antibodies that are bound to membrane receptors on tissue mast cells and blood basophils. The antigen-antibody reaction causes the rapid release of potent vasoactive and inflammatory mediators, (e.g., histamine, tryptase, leukotrienes and prostaglandins) and later release of proinflammatory cytokines (e.g., interleukin-4 and -interleukin-13). The mediators produce vasodilation, increased capillary permeability, glandular hypersecretion, smooth muscle spasm, and tissue infiltration of other inflammatory cells. Exemplary type I hypersensitivity disorders include allergic rhinitis, allergic conjunctivitis, atopic dermatitis, allergic asthma, some cases of urticaria and GI food reactions, and systemic anaphylaxis.

In another aspect, the condition is an autoimmune disease. The autoimmune disease contemplated by the invention may be multiple sclerosis, rheumatoid arthritis, bullous pemphigoid, Sjogren's syndrome, chronic urticaria, thyroid eye disease, vasculitis, and peritonitis.

The invention provides for a method of the invention wherein the PI3K8 selective inhibitor is administered in an amount effective to inhibit Akt phosphorylation in said mast cells.

The terms "selective P13K6 inhibitor," and variants thereof such as "PI3KS selective inhibitor" and "selective inhibitor of PI3KS" as used herein refer to a compound that inhibits the PI3K8 isozyme more effectively than other isozymes of the P13K family. A "selective PI3K6 inhibitor" compound is understood to be more selective for PI3K6 than compounds conventionally and generically designated inhibitors, e.g., wortmannin or LY294002. Concomitantly, wortmannin and LY294002 are deemed "nonselective P13K inhibitors."

The relative efficacies of compounds as inhibitors of an enzyme activity (or other biological activity) can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results. Typically, the preferred determination is the concentration that inllibits 50% of the activity in a biochemical assay, i.
e., the 50%
inhibitory concentration or "IC50." IC50 determinations can be accomplished using conventional techniques known in the art. In general, an IC50 can be determined by 5 measuring the activity of a given enzyme in the presence of a range of concentrations of the inhibitor under study. The experimentally obtained values of enzyme activity then are plotted against the inhibitor concentrations used. The concentration of the inhibitor that shows 50% enzyme activity (as compared to the activity in the absence of any inhibitor) is taken as the IC50 value. Analogously, other inhibitory 10 concentrations can be defined through appropriate determinations of activity. For example, in some settings it can be desirable to establish a 90% inhibitory concentration, i.e., IC90, etc.

Accordingly, a "selective PI3K8 inhibitor" alternatively can be understood to refer to a compound that exhibits a 50% inhibitory concentration (IC50) with respect to P13K5 that is at least 10-fold, in another aspect at least 20-fold, and in another aspect at least 30-fold, lower than the IC50 value with respect to any or all of the other Class I P13K family members. In an alternative embodiment of the invention, the term selective PI3K8 inhibitor can be understood to refer to a compound that exhibits an IC50 with respect to PI3K8 that is at least 50-fold, in another aspect at least 100-fold, in an additional aspect at least 200-fold, and in yet another aspect at least 500-fold, lower than the IC50 with respect to any or all of the other P13K Class I family members. In yet a further embodiment, the term selective PI3K6 inhibitor refers to an oligonucleotide that negatively regulates pl 108 expression at least 10-fold, in another aspect at least 20-fold, and in a further aspect at least 30-fold, lower than any or all of the other Class I P13K family catalytic subunits (i.e., p110oc, pl 10(3, and p110y). A PI3K8 selective inhibitor is administered to an individual in an amount such that the inhibitor retains its P13K6 selectivity, as described above.

Ranges may be expressed herein as from "about" or "approximately"
one particular value and/or to "about" or "approximately" another particular value.
When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedents such as "about" or "at least about,"
it will be understood that the particular value forms another embodiment.

Any selective inhibitor of PI3K8 activity, including but not limited to small molecule inhibitors, peptide inhibitors, non-peptide inhibitors, naturally occurring inhibitors, and synthetic inhibitors, may be used. Suitable PI3K8 selective inhibitors have been described in U.S. Patent Publication 2002/161(}14 to Sadhu et al., the, entire disclosure of which is hereby incorporated herein by reference.

In another aspect, compounds of any type that selectively negatively regulate p 1105 expression (i.e., more effectively than other isozymes of the farriily) and that possess acceptable pharmacological properties can be used as selective PI3K8 inhibitors in the methods of the invention. Accordingly, in one embodiment the invention provides for the use of antisense oligonucleotides which negatively regulate p110S expression via hybridization to messenger RNA (mRNA) encoding p110S. In one aspect, oligonucleotides that decrease pI lOS
expression may be used in the methods of the invention.

The methods of the invention may be applied to cell populations in vivo or ex vivo. "In vivo" means within a living individual, as within an animal or human. In this context, the methods of the invention may be used therapeutically in an individual, as described infra. The methods may also be used prophylactically.

"Ex vivo" means outside of a living individual. Examples of ex vivo cell populations include in vitro cell cultures and biological samples including but not-limited to fluid or tissue samples obtained from individuals. Such samples may be obtained by methods well known in the art. Exemplary biological fluid samples include blood, cerebrospinal fluid, and saliva. Exemplary tissue samples include tumors samples and biopsies of tissue. In this context, the invention may be used for a variety of purposes, including therapeutic and experimental purposes. For example, the invention may be used ex vivo to determine the optimal schedule and/or dosing of administration of a PI3K8 selective inhibitor for a given indication, cell type, individual, and other parameters. Information gleaned from such use may be used for experimental purposes or in the clinic to set protocols for in vivo treatment.
Other ex vivo uses for which the invention may be suited are described below or will become apparent to those skilled in the art.
"Mast cell activity" as used herein refers to those biological activities carried out by mast cells which may be modulated by the compounds useful in the methods of the invention. Examples of these activities include cell migration, proliferation, activation, degranulation, expression of or secretion of chemokines, cytokines or other growth factors, and modulation of cell signaling pathways, for example, modulation of AKT phosphorylation.

Alternatively, "modulation of mast cell activity" may be used herein.
"Modulation" of mast cell activity as used herein refers to the reduction, inhibition, prevention, promotion or increase of one of the above listed activities of mast cells upon administration of a selective PI3K6 inhibitor. A selective PI3K8 inhibitor may inhibit the enzyme itself, may inhibit any downstream signaling effect of the enzyme, or inhibit any further downstream activity of a mast cell. It may alternatively act prophylactically to prevent a particular activity of a mast cell as described above, such as degranulation or cellular migration. An inhibitor may promote or increase one mast cell activity in the course of reducing, inhibiting or preventing another mast cell activity. For example, production of a first cytokine, chemokine or growth factor may be increased upon reduction or inhibition of another second cytokine, chemokine or growth factor.

"Undesirable mast cell activity" means mast cell activity that deviates from the normal, proper, or expected course. For example, undesirable mast cell degranulation may include degranulation during allergic reaction causing hypersensitivity of the individual, while undesirable migration may include movement of mast cells into or out of tissue sites having unfavorable biological effects.
Undesirable mast cell proliferation may include cell proliferation mediated by, or resulting in inappropriately high levels of cell division, inappropriately low levels of apoptosis, or both. .

"hihibiting undesirable mast cell activity" means to slow or stop the rate at which undesirable mast cell activity takes place. This may result either from a decreased rate of mast cell receptor activation, decreased inflammatory mediator or growth factor release, decreased cellular replication, or an increased rate of cell death.
Cell death may occur by any mechanism, including apoptosis and mitotic catastrophe.
Use of the methods in accordance with the present invention may result in partial or complete inhibition of undesirable mast cell activity,.
"Preventing undesirable mast cell activity" means that the methods of the present invention may be used prophylactically to prevent or inhibit undesirable mast cell activity before it occurs, or to prevent or inhibit the recurrence thereof.
Thus, in all embodiments, the invention may be used ira vivo or ex vivo where no undesirable cell activity has been identified or where no undesirable cell activity is ongoing, but where undesirable cell activity is suspected or expected, respectively.
Moreover, the invention may also be used in all its embodiments wherever undesirable cell activity has been previously treated to prevent or inhibit recurrence of the same.

As used herein, a "therapeutically effective amount" or "amount effective" means an amount effective to inhibit or reverse development of, to alleviate the existing symptoms of, to prolong survival of, or to cure the individual being treated.

As used herein, the "therapeutic index" is the dose ratio between toxic ' or undesired effect and therapeutic, or desired, effects, and is expressed as the ratio of LD50 to ED50, which are defined below. An increase in the therapeutic index, as used herein, refers to a reduction in the amount of therapeutic necessary to reach a desired effect or to increase the effectiveness of the therapeutic administered.

It will be appreciated that the treatment methods of the invention are - useful in the fields of human medicine and veterinary medicine. Thus, the individual to be treated may be a mammal, preferably human, or another animal. For veterinary purposes, individuals include but are not limited to farm animals including cows, sheep, pigs, horses, and goats; companion animals such as dogs and cats;
exotic and/or zoo animals; laboratory animals including mice, rats, rabbits, guinea pigs, and hamsters; and poultry such as chickens, turkeys, ducks, and geese.

The invention fixrther provides methods wherein the selective inhibitor of phosphoinositide 3-kinase delta (PI3K8) s is administered in a plurality of doses. A
plurality of doses includes administration of the inhibitor or other agent in more than one dose. The invention fiirther provides that the selective PI3K8 inhibitor is administered in a regimen which includes administering one or more additional therapeutic compounds commonly utilized in treatment of a condition associated with undesirable mast cell activity, including at least one immunomodulatory agent or other agent as appropriate to the condition or symptom being treated or prevented.
In one embodiment, the invention provides a method of reducing or preventing mast cell activity in an individual having a condition associated with undesirable mast cell activity, comprising administering to said individual a therapeutically effective amount of a combination therapy comprising a selective inhibitor of phosphoinositide 3-kinase delta (PI3K8) and an immunomodulatory agent.' It is contemplated that the combination therapy may be administered in a single composition or each agent, such as the inhibitor and immunomodulatory agent,.
10; may be administered as a separate composition. It is further contemplated that each agent may be administered in a plurality of doses as necessary.

In a related embodiment, the invention provides a method of reducing or preventing lymphocyte infiltration to a site of inflamniation in an individual having a condition associated with undesirable mast cell activity, comprising administering to said individual a therapeutically effective amount of a combination therapy comprising a selective inhibitor of phosphoinositide 3-kinase delta (PI3K8) and a immunomodulatory agent.

Immunomodulatory agents contemplated by the invention include glucocorticoids or corticosteroids, immunosuppressants, antihistamines, aminosalicylates, steroid hormones, non-steroidal anti-inflammatory drugs (NSAIDs), .
sympathomimetics, and analgesics. Exemplary glucocorticoids are chosen from the group consisting of cortisone, dexamethosone, hydrocortisone, methylprednisolone, prednisolone, prednisone, and budesonide. Exemplary NSAIDs are chosen from the group consisting of ibuprofen, naproxen, naproxen sodium, Cox-2 inhibitors such as Vioxx (rofecoxib) and Celebrex (celecoxib), and salicylate. Suitable analgesics include acetaminophen, oxycodone, tramadol of proporxyphene hygrochloride.
Exemplary immunosuppressants include azathioprine (6-mercaptopurine (6-MP)), cyclophosphamide, cyclosporine, methotrexate, or penicillamine. Also contemplated are Xolair (oinalizumab), leukotriene antagonists, or other drugs commonly used for allergy or asthma.

Methods of the invention may include administering formulations comprising an inhibitor of the invention with a particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent.

More specifically and without limitation, methods of the invention may comprise administering an inhibitor with one or more of TNF, IL-1, IL-2, IL-3, IL-4, 5 IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IFN, G-CSF, Meg-CSF, GM-CSF, thrombopoietin, stem cell factor, and erythropoietin. Pharmaceutical compositions in accordance with the invention may also include other known angiopoietiiis, for example Ang- 1, Ang-2, Ang-4, Ang-Y, and/or the human angiopoietin-like polypeptide, and/or vascular endothelial growth 10 factor (VEGF). Growth factors for use in pharmaceutical compositions of the invention include angiogenin, bone morphogenic protein-1, bone morphogenic protein-2, bone morphogenic protein-3, bone morphogenic protein-4, bone morphogenic protein-5, bone morphogenic protein-6, bone morphogenic protein-7, bone morphogenic protein-8, bone morphogenic protein-9, bone morphogenic 15 protein-10, bone morphogenic protein-11, bone morphogenic protein-12, bone morphogenic protein- 13, bone morphogenic.protein-14, bone morphogenic protein-15; bone morphogenic protein receptor IA, bone morphogenic protein receptor IB, brain derived neurotrophic factor, ciliary neutrophic factor, ciliary neutrophic factor receptor a, cytokine-induced neutrophil chemotactic factor 1, cytokine-induced neutrophil chemotactic factor 2a, cytokine-induced neutrophil chemotactic factor 20, 0 endothelial cell growth factor, endothelin 1, epidermal growth factor, epithelial-derived neutrophil attractant, fibroblast growth factor 4; fibroblast growth factor 5, fibroblast growth factor 6, fibroblast growth factor 7, fibroblast growth factor 8, fibroblast growth factor 8b, fibroblast growth factor 8c, fibroblast growth factor 9, fibroblast growth factor 10, fibroblast growth factor acidic, fibroblast growth factor basic, glial cell line-derived neutrophic factor receptor al, glial cell line-derived neutrophic factor receptor a2, growth related protein, growth related protein a, growth related protein 0, growth related protein y, heparin binding epidermal growth factor, hepatocyte growth factor, hepatocyte growth factor receptor, insulin-like growth factor I, insulin-like growth factor receptor, insulin-like growth factor II, insulin-like growth factor binding protein, keratinocyte growth factor, leukemia inhibitory factor, leukemia inhibitory factor receptor a, nerve growth factor, nerve growth factor receptor, neurotrophin-3, neurotrophin-4, placenta growth factor, placenta growth factor 2, platelet derived endothelial cell growth factor, platelet derived growth factor, platelet derived growth factor A chain, platelet derived growth factor AA, platelet derived growth factor AB, platelet derived growth factor B chain, platelet derived growth factor BB, platelet derived growth factor receptor a, platelet derived growth factor receptor 0, pre-B cell growth stimulating factor, stem cell factor, stem cell factor receptor, transforming growth factor a, transforming growth factor 0, transforming growth factor (31, transforming growth factor (31.2, transforming growth factor 02, transforming growth factor 03, transforming growth factor 05, latent transforming growth factor (31, transforming growth factor (3 binding protein I, transforming growth factor (3 binding protein II, transforming growth factor (3 binding protein III, tumor necrosis factor receptor type I, tumor necrosis factor receptor type II, urokinase-type plasminogen activator receptor, vascular endothelial growth factor, and chimeric proteins and biologically or immunologically active fragments thereof.

Immunomodulatory agents used for treatment can be administered in a plurality of doses. It is conteinplated that the agents are administered in the combination methods according to the invention at a low dose, that is, at a dose lower than conventionally used in clinical situations where the agent or therapy is administered alone, because the P13K6 selective nature of the inhibitors of the invention increases the therapeutic index (i.e., the specificity) of the inventive combination therapies. Lowering the dose of the agent or therapy administered to an individual decreases the incidence of adverse effects associated with higher dosages, and can thereby improve the quality of life of a patient undergoing treatment.
Further benefits include improved patient compliance, and a reduction in the number of hospitalizations needed for the treatment of adverse effects. Additionally, the specificity of the methods of the invention are advantageous in that they permit treatment at higher doses of the PI3K8 selective inhibitor(s) than nonselective inhibitors such as LY294002 and wortmannin, further maximizing the therapeutic efficacy of the inventive methods In another aspect, methods may include administering an inliibitor with one or more other agents which either enhance the activity of the inhibitor or compliment its activity or use in treatment. Such additional factors and/or agents may produce a synergistic effect with an inhibitor of the invention, or to minimize side effects.
Methods of the invention contemplate use of a selective PI3K6 inhibitor compound having formula (I) or pharmaceutically acceptable salts and solvates thereof:

Rb ~R3 N
/

wherein A is an optionally substituted monocyclic or'bicyclic ring system containing at least two nitrogen atoms, and at least one ring of the system is aromatic;

X is selected from the group consisting of C(Rb)2, CH2CHRb, and CH=C(Rb);

Y is selected from the group consisting of null, S, SO, SO2, NH, 0, C(=0), OC(=O), C(=0)O, and NHC(=O)CH2S;

Rl and R2, independently, are selected from the group consisting of hydrogen, C1-6alkyl, aryl,.heteroaryl,.halo, NHC(=O)C1-3alkyleneN(Ra)2, NO2, ORa, CF3, OCF3, N(Ra)2, CN, OC(=O)Ra, C(=0)Ra, C(=O)ORa, arylORb, Het, NRaC(=O)C1_3alkyleneC(=O)ORa, arylOC1-3alkyleneN(Ra)2, arylOC(=0)Ra, C1-4alkyleneC(=O)ORa, OC 1_4alkyleneC(=O)ORa, C 1_4alkyleneOC 1-4alkyleneC(=O)ORa, C(=O)NRaSO2Ra, C1_4alkyleneN(Ra)2, C2_ 6alkenyleneN(Ra)2, C(=0)NRaC1_4alkyleneORa, C(=O)NRaC1-4alkyleneHet, OC2-4alkyleneN(Ra)2, OC1_4alkyleneCH(ORb)CH2N(Ra)2, OC1-4alkyleneHet, OC2-4alkyleneORa, OC2_4alkyleneNRaC(=0)ORa, NRaC1-4alkyleneN(Ra)2, NRaC(=O)Ra, NRaC(=O)N(Ra)2, N(S02C1-4alkyl)2, NRa(S02C1-4alkyl), S02N(Ra)2, OSO2CF3, C1-3alkylenearyl, C1-4alkyleneHet, C1-6alkyleneORb, C1_ 3alkyleneN(Ra)2, C(=O)N(Ra)2, NHC(=O)C1-3alkylenearyl, C3-8cycloalkyl, C3-8heterocycloalkyl, arylOC1-3alkyleneN(Ra)2, arylOC(=O)Rb, NHC(=O)Cl-3alkyleneC3_8heterocycloalkyl, NHC(=O)C1-3alkyleneHet, OC1-4alkyleneOCl_ q.alkyleneC(=O)ORb, C(=O)C1-4alkyleneHet, and NHC(=O)haloC1-6alkyl;

or RI and R2 are taken together to form a 3- or 4-membered alkylene or alkenylene chain component of a 5- or 6-membered ring, optionally containing at least one heteroatom;

R3 is selected from the group consisting of optionally substituted 1lydrogen, C1_6alkyl, C3-8cycloalkyl, C3_8heterocycloalkyl, C1-4alkylenecycloalkyl;
C2-6alkenyl, C1-3alkylenearyl, ary1C1-3alkyl, C(=O)Ra, aryl, heteroaryl, C(=O)ORa, C(=O)N(Ra)2, C(=S)N(Ra)2, SO2Ra, SO2N(Ra)2, S(=O)Ra, S(=O)N(Ra)2, C(=O)NRaC 1-4alkyleneORa, C(=O)NRaC 1_4alkyleneHet, C(=O)C 1_4alkylenearyl, C(=O)C1-4alkyleneheteroaryl, C1-4alkylenearyl optionally substituted with one or more of halo, SO2N(Ra)2, N(Ra)2, C(=O)ORa, NRaSO2CF3, CN, NO2, C(=O)Ra, ORa, C 1_4alkyleneN(Ra)2, and OC 1_4alkyleneN(Ra)2, C 1-4alkyleneheteroaryl, C

4alkyleneHet, C 1-4alkyleneC(=O)C 1-4alkylenearyl, C 1_4alkyleneC(=O)C 1-4alkyleneheteroaryl, C 1-q.alkyleneC(=O)Het, C 1-4alkyleneC(=O)N(Ra)2, C 1-4alkyleneORa, C1-4alkyleneNRaC(=0)Ra, C1-4alkyleneOC1_4alkyleneORa, Cl-4alkyleneN(Ra)2, C 1_4alkyleneC(=0)ORa, and C 1-4alkyleneOC 1-4alkyleneC(=O)ORa;

Ra is selected from the group consisting of hydrogen, C1_6alkyl, C3-8cycloalkyl, C3-8heterocycloalkyl, C1-3alkyleneN(Rc)2, aryl, ary1C1-3alkyl, Cl-3alkylenearyl, heteroaryl, heteroarylC1-3alkyl, and C1-3alkyleneheteroaryl;

or two Ra groups are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;
Rb is selected from the group consisting of hydrogen, C1-6alkyl, heteroCl_3alkyl, C1-3alkyleneheteroC1-3alkyl, arylheteroC1-3alkyl, aryl, heteroaryl, ary1C 1-3 alkyl, heteroarylC 1_3 alkyl, C 1_3 alkylenearyl, and C 1-3 alkylenehetero aryl;

Rc is selected from the group consisting of hydrogen, C1-6alkyl, C3_ gcycloalkyl, aryl, and heteroaryl; and, Het is a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1_4alkyl or C(=0)ORa.

As used herein, the term "alkyl" is defined as straight chained and branched hydrocarbon groups containing the indicated number of carbon atoms, typically methyl, ethyl, and straight chain and branched propyl and butyl groups. The hydrocarbon group can contain up to 16 carbon atoms, for example, one to eight carbon atoms. The term "alkyl" includes "bridged alkyl," i.e., a C6-C16 bicyclic or polycyclic hydrocarbon group, for example, norbomyl, adarnantyl, bicyclo[2.2.2]octyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, or decahydronaphthyl.
The term "cycloalkyl" is defined as a cyclic C3-Cg hydrocarbon group, e.g., cyclopropyl, cyclobutyl, cyclohexyl, and cyclopentyl.

The term "alkenyl" is defined identically as "alkyl," except for containing a carbon-carbon double bond. "Cycloalkenyl" is defined similarly to cycloalkyl, except a carbon-carbon double bond is present in the ring.

The ternl "alkylene" is defined as an alkyl group having a substituent.
For example, the term "C1_3alkylenearyl" refers to an alkyl group containing one to three carbon atoms, and substituted with an aryl group.

The term "heteroCi_3alkyP" is defined as a C1_3alkyl group further containing a heteroatom selected from 0, S, and NRa. For example, -CH2OCH3 or -CHZCH2SCH3. The term "arylheteroC1_3alkyl" refers to an aryl group having a heteroCi_3alkyl substituent.

The term "halo" or "halogen" is defined herein to include fluorine, bromine, chlorine, and iodine.

The term "aryl," alone or in combination, is defined herein as a monocyclic or polycyclic aromatic group, e.g., phenyl or naphthyl. Unless otherwise indicated, an "aryl" group can be unsubstituted or substituted, for example, with one or more, and in particular one to three, halo, alkyl, phenyl, hydroxyalkyl, alkoxy, 5 alkoxyalkyl, haloalkyl, nitro, and amino. Exemplary aryl groups include phenyl, naphthyl, biphenyl, tetrahydronaphthyl, chlorophenyl, fluorophenyl, aminophenyl, methylphenyl, methoxyphenyl, trifluoromethylphenyl, nitrophenyl, carboxyphenyl, and the like. The terms "ary1C1_3alkyl" and "heteroarylC1_3alkyl" are defined as an aryl or heteroaryl group having a C1_3alkyl substituent.

10 The term "heteroaryl" is defined herein as a monocyclic or bicyclic ring system containing one or two aromatic rings and containing at least one nitrogen, oxygen, or sulfur atom in an aromatic ring, and which can be unsubstituted or substituted, for example, with one or more, and in particular one to three, substituents, like halo, alkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkyl, nitro, and 15 amino. Examples of heteroaryl groups include thienyl, furyl, pyridyl, oxazolyl, quinolyl, isoquinolyl, indolyl, triazolyl, isothiazolyl, isoxazolyl, imidizolyl, benzothiazolyl, pyrazinyl, pyrimidinyl, thiazolyl, and thiadiazolyl.

The term "Het" is defined as monocyclic, bicyclic, and tricyclic groups containing one or more heteroatoms selected from the group consisting of oxygen, 20 nitrogen, and sulfur. A"Het" group also can contain an oxo group (=0) attached to the ring. Nonlimiting examples of Het groups include 1,3-dioxolane, 2-pyrazoline, pyrazolidine, pyrrolidine, piperazine, a pyrroline, 2H-pyran, 4H-pyran, morpholine, thiopholine, piperidine, 1,4-dithiane, and 1,4-dioxane.

Alternatively, methods of the invention contemplate use of a PI3K8 selective inhibitor compound having formula (II) or pharmaceutically acceptable salts and solvates thereof:
N
N~ Rs N N
iN
R N
(II) wherein R4, R5, R6, and R7, independently, are selected from the group consisting of hydrogen, C1-6alkyl, aryl, heteroaryl, halo, NHC(=O)C1_ 3alkyleneN(Ra)2, NO2, ORa, CF3, OCF3, N(Ra)2, CN, OC(=O)Ra, C(=O)Ra, C(=O)ORa, arylORb, Het, NRaC(=O)C1-3alkyleneC(=O)ORa, arylOCl-3alkyleneN(Ra)2, arylOC(=O)Ra, C1_4alkyleneC(=O)ORa, OC1_ 4alkyleneC(=O)ORa, C1-4alkyleneOC1-4alkyleneC(=O)ORa, C(=O)NRaSO2Ra, C 1-4alkyleneN(Ra)2, C2_6alkenyleneN(Ra)2, C(=O)NRaC 1-4alkyleneORa, C(=O)NRaC1-4alkyleneHet, OC2_4alkyleneN(Ra)2, OC1-4alkyleneCH(ORb)CH2N(Ra)2, OC1-4a1ky1eneHet, OC2-4alkyleneORa, OC2-4alkyleneNRaC(=0)ORa, NRaC1_4alkyleneN(Ra)2, NRaC(=O)Ra, NRaC(=O)N(Ra)2, N(S02C1-4alkyl)2, NRa(S02C 1 -4alkyl), SO2N(Ra)2, OSO2CF3, C1-3alkylenearyl, C1-4alkyleneHet, C1_6alkyleneORb, C1-3alkyleneN(Ra)2, C(=0)N(Ra)2, NHC(=0)C1-3alkylenearyl, C3_8cycloalkyl, C3_ gheterocycloalkyl, arylOC1_3alkyleneN(Ra)2, arylOC(=O)Rb, NHC(=O)C1-3alkyleneC3_8heterocycloalkyl, NHC(=O)C1_3alkyleneHet, OC1-4alkyleneOC1-4alkyleneC(=0)ORb, C(=0)C1_4alkyleneHet, and NHC(=O)haloCl-6alkyl;
R8 is selected from the group consisting of hydrogen, C1-6alkyl, halo, CN, C(=O)Ra, and C(=0)ORa;

Xl is selected from the group consisting of CH (i.e., a carbon atom having a hydrogen atom attached thereto) and nitrogen;

Ra is selected from the group consisting of hydrogen, C1_6alkyl, C3-8cycloalkyl, C3-8heterocycloalkyl, C1-3alkyleneN(R92, aryl, arylCl_3alkyl, C1-3alkylenearyl, heteroaryl, heteroarylC1-3alkyl, and C1-3alkyleneheteroaryl;

or two Ra groups are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;

Rc is selected from the group consisting of hydrogen, Ci-6alkyl, C3-8cycloalkyl, aryl, and lieteroaryl; and, Het is a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1-4alkyl or C(=O)ORa.

In yet another embodiment, methods of the invention include use of a selective inhibitor of PI3K8 compound having formula (III) or pharmaceutically acceptable salts and solvates thereof:
Rl o Rl l O

N

HN N
N
N
~NH
(M) wherein R9, R10, Rll, and R12, independently, are selected from the group consisting of hydrogen, C1_galkyl, aryl, heteroaryl, halo, NHC(=O)C1_ 3alkyleneN(Ra)2, NO2, ORa, CF3, OCF3, N(Ra)2, CN, OC(=O)Ra, C(=O)Ra, C(=O)ORa, arylORb, Het, NRaC(=O)C1-3alkyleneC(=O)ORa, arylOCl_ 3 alkyleneN(Ra)2, arylOC(=O)Ra, C 1-4alkyleneC(=O)ORa, OC 1_ 4alkyleneC(=0)ORa, C1_4alkyleneOC1_4alkyleneC(=O)ORa, C(=O)NRaSO2Ra, C 1_4alkyleneN(Ra)2, C2-6alkenyleneN(Ra)2, C(=O)NRaC 1_4alkyleneORa, C(=O)NRaC 1-4alkyleneHet, OC2_4alkyleneN(R a)2, OC 1_ 4alkyleneCH(ORb)CH2N(Ra)2, OC 1_4alkyleneHet, OC2-4alkyleneORa, OC2-4alkyleneNRaC(=0)ORa, NRaC1_4alkyleneN(Ra)2, NRaC(=O)Ra, NRaC(=O)N(Ra)2, N(SO2C1-4alkyl)2, NRa(S02C1-4alkyl), S02N(Ra)2, OSO2CF3, C1_3alkylenearyl, C1_4alkyleneHet, C1-6alkyleneORb, Cl_ 3alkyleneN(Ra)2, C(=O)N(Ra)2, NHC(=O)C1-3alkylenearyl, C3-8cycloalkyl, C3_ gheterocycloalkyl, arylOC1_3alkyleneN(Ra)2, arylOC(=0)Rb, NHC(=O)C1_ 3alkyleneC3_gheterocycloalkyl, NHC(=O)C1_3alkyleneHet, OC1_4alkyleneOCl_ 4alkyleneC(=0)ORb, C(=O)C1_4alkyleneHet, and NHC(=O)haloC1_6alkyl;
R13 is selected from the group consisting of hydrogen, C1-6alkyl, halo, CN, C(=O)Ra, and C(=O)ORa;

Ra is selected from the group consisting of hydrogen, C1-6alkyl, C3-8cycloalkyl, C3-8heterocycloalkyl, C1-3alkyleneN(Rc)2, aryl, arylC1-3alkyl, Cl-3alkylenearyl, heteroaryl, heteroarylC1-3alkyl, and C1-3alkyleneheteroaryl;

or two Ra groups are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;

Rc is selected from the group consisting of hydrogen, C1-6alkyl, C3-8cycloalkyl, aryl, and heteroaryl; and, Het is a 5- or 6-membered heterocyclic ring, saturated or partially or f-ully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1-4alkyl or C(=O)ORa. More specifically, methods of the invention embrace use of a selective inhibitor selected from the group consisting of 2-(6-aminopurin-9-ylmethyl)-3-(2-chlorophenyl)-6,7-dimethoxy-3H-quinazolin-4-one; 2-(6-aminopurin-o-ylmethyl)-6-bromo-3-(2-chlorophenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-o-ylmethyl)-3-(2-chlorophenyl)-7-fluoro-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-6-chloro-3-(2-chlorophenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-chlorophenyl)-5-fluoro-3H-quinazolin-4-one; 2-(6-aminopurin-o-ylmethyl)-5-chloro-3-(2-chloro-phenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-chlorophenyl)-5-methyl-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-8-chloro-3-(2-chlorophenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-biphenyl-2-yl-5-chloro-3H-quinazolin-4-one; 5-chloro-2-(9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-chloro-3-(2-fluorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-chloro-3-(2-fluorophenyl)-3H-quinazolin-4-one; 3-biphenyl-2-yl-5-chloro-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 5-chloro-3-(2-methoxyphenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-fluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-6,7-dimethoxy-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 6-bromo-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-8-trifluoromethyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-one; 3-(2-chlorophenyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-benzo[g]quinazolin-5 one; 6-chloro-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 8-chloro-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-7-fluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-7-nitro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-one; 3-(2-chlorophenyl)-6-hydroxy-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-10 4-one; 5-chloro-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-methyl-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-6,7-difluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-6-fluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-isopropylphenyl)-5-methyl-15 ' 3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 3-(2-fluorophenyl)-5-methyl-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-chloro-3-o-tolyl-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-chloro-3-(2-methoxy-phenyl)-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-3-cyclopropyl-5-methyl-20 3H-quinazolin-4-one; 3-cyclopropylmethyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclopropylmethyl-5-methyl-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-3-cyclopropylmethyl-5-methyl-3H-quinazolin-4-one; 5-methyl-3-phenethyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(2-amino-9H-25 purin-6-ylsulfanylmethyl)-5-methyl-3-phenethyl-3H-quinazolin-4-one; 3-cyclopentyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclopentyl-5-methyl-3H-quinazolin-4-one; 3-(2-chloropyridin-3 -yl)-5 -methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one;
2-(6-aminopurin-9-ylmethyl)-3-(2-chloropyridin-3-yl)-5-methyl-3H-quinazolin-4-one; 3-methyl-4-[5-methyl-4-oxo-2-(9H-purin-6-ylsulfanylmethyl)-4H-quinazolin-yl]-benzoic acid; 3-cyclopropyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclopropyl-5-methyl-3H-quinazolin-4-one; 5-methyl-3-(4-nitrobenzyl)-2-(9H-purin-6-ylsulfanylmethyl)-quinazolin-4-one; 3-cyclohexyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclohexyl-5-methyl-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-3-cyclo-hexyl-5-methyl-3H-quinazolin-4-one; 5-methyl-3-(E-2-phenylcyclopropyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-fluoro-2-[(9H-purin-6-ylamino) methyl]-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)methyl]-3-(2-chlorophenyl)-5-fluoro-3H-quinazolin-4-one; 5-methyl-2-[(9H-purin-6-ylamino)methyl]-3-o-tolyl-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)methyl]-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-[(2-fluoro-9H-purin-ylainino)methyl]-5-methyl-3-o-tolyl-3H-quinazolin-4-one; (2-chlorophenyl)-dimethylamino-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 5-(2-benzyloxyethoxy)-3-(2-chlorophenyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 6-aminopurine-9-carboxylic acid 3-(2-chlorophenyl)-5-fluoro-oxo-3,4-dihydro-quinazolin-2-ylmethyl ester; N-[3-(2-chlorophenyl)-5-fluoro-4-oxo-3,4-dihydro-quinazolin-2-ylmethyl]-2-(9H-purin-6-ylsulfanyl)-acetamide; 2-[1-(2-fluoro-9H-purin-6-ylamino)ethyl]-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-[ 1-(9H-purin-6-ylamino)ethyl]-3-o-tolyl-3H-quinazolin-4-one; 2-(6-dimethylaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(2-methyl-6-oxo-1,6-dihydro-purin-7-ylmethyl)-3-o-tolyl-3H-quinazolin-4-one;

methyl-2-(2-methyl-6-oxo-1,6-dihydro-purin-9-ylmethyl)-3-o-tolyl-3H-quinazolin-one; 2-(amino-dimethylaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-one; 2-(2-amino-9H-purin-6-y1sulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-one; 2-(4-amino-1,3,5-triazin-2-y1sulfanylmethyl)-5-methyl-3-o-to1y1-3H-quinazolin-, 4-one; 5-methyl-2-(7-methyl-7H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(2-oxo-1,2-dihydro-pyrimidin-4-ylsulfanylmethyl)-3-o-tolyl-quinazolin-4-one; 5-methyl-2-purin-7-ylmethyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-purin-9-ylmethyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(9-methyl-9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 2-(2,6-Diamino-pyrimidin-4-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-(5 -methyl-[ 1,2,4]triazolo [ 1,5-a]pyrimidin-7-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(2-methylsulfanyl-9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 2-(2-hydroxy-9H-purin-6-ylsulfanylmethyl)-5-methyl-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(1-methyl-1 H-imidazol-2-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-3-o-tolyl-2-(1H-[1,2,4]triazol-3-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(2-amino-6-chloro-purin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(6-aminopurin-7-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(7-amino-1,2,3-triazolo[4,5-d]pyrimidin-3-yl-methyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(7-amino-1,2,3-triazolo[4,5-d]pyrimidin-1-yl-methyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(6-amino-9H-purin-2-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(2-amino-6-ethylamino-pyrimidin-4-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one;
2-(3-amino-5-methylsulfanyl-1,2,4-triazol-1-yl-methyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(5-amino-3-methylsulfanyl-1,2,4-triazol-1-ylmethyl)=5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(6-methylaminopurin-9-ylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 2-(6-benzylaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(2,6-diaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one;- 3-isobutyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; N-{2-[5-Methyl-4-oxo-2-(9H-purin-6-ylsulfanylmethyl)-4H-quinazolin-3-yl]-phenyl}-acetamide; 5-methyl-3-(E-2-methyl-cyclohexyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-[5-methyl-4-oxo-2-(9H-purin-6-ylsulfanylmethyl)-4H-quinazolin-3-yl]-benzoic acid; 3-{2-[(2-dimethylaminoethyl)methylamino]phenyl} -5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quin-azolin-4-one; 3-(2-chlorophenyl)-5-methoxy-2-(9H=
20, purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-(2-morpholin-4-yl-ethylamino)-2-(9H-purin-6-y1su1fanylmethyl)-3H- quiiiazolin-4-one; 3-benzyl-5-methoxy-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-, 9-ylmethyl)-3-(2-benzyloxyphenyl)-5-methyl-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-hydroxyphenyl)-5-methyl-3H-quinazolin-4-one; 2-(1-(2-amino-9H-purin-6-ylamino)ethyl)-S-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-[1-(9H-purin-6-ylamino)propyl]-3-o-tolyl-3H-quinazolin-4-one; 2-(1-(2-fluoro-9H-purin-6-ylamino)propyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(1-(2-amino-9H-purin-6-ylamino)propyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(2-benzyloxy-l-(9H-purin-6-ylamino)ethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one;

(6-aminopurin-9-ylmethyl)-5-methyl-3-{2-(2-(1-methylpyrrolidin-2-yl)-ethoxy)-phenyl}-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-(3-dimethylamino-propoxy)-phenyl)-5-methyl-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-methyl-3-(2-prop-2-ynyloxyphenyl)-3H-quinazolin-4-one; 2-{2-(1-(6-aminopurin-9-ylmethyl)-5-methyl-4-oxo-4H-quinazolin-3-yl]-phenoxy}-acetamide; 2-[(6-aminopurin-9-yl)methyl]-5-methyl-3-o-tolyl-3-hydroquinazolin-4-one; 3-(3,5-difluorophenyl)-5-methyl-2-[(purin-6-ylamino)methyl]-3-hydroquinazolin-4-one;

(2,6-dichlorophenyl)-5-methyl-2-[(purin-6-ylamino)methyl]-3-hydroquinazolin-4-one; 3-(2-Fluoro-phenyl)-2-[ 1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3-hydroquinazolin-4-one; 2-[1-(6-aminopurin-9-yl)ethyl]-3-(3,5-difluorophenyl)-5-methyl-3-hydroquinazolin-4-one; 2-[ 1-(7-Amino-[ 1,2,3]triazolo[4,5-d]pyrimidin-3-yl)-ethyl]-3-(3,5-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 5-chloro-3-(3,5-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 6-fluoro-3-phenyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-fluoro-3-phenyl-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(2,3-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3-chloro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)-methyl]-3-(3,5-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-{2-[(2-diethylamino-ethyl)-methyl-amino]-phenyl}-5-methyl-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 5-chloro-3-(2-fluoro-phenyl)-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 5-chloro-2-[(9H-purin-6-ylamino)-methyl]-3-o-tolyl-3H-quinazolin-4-one; 5-chloro-3-(2-chloro-phenyl)-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 6-fluoro-3-(3-fluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-chloro-3-(3-fluoro-phenyl)-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[1-(2-fluoro-9.H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-2-[1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[ 1-(7H-pyrrolo [2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[ 1-(2-amino-9H-purin-6-ylamino)-propyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(7H-pyrrolo[2,3-d]pyrimidin-ylamino)-propyl]-3H-quinazolin-4-one; 2-[1-(2-fluoro-9h-purin-6-ylamino)-propyl]-5-methyl-3-phenyl-3h-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-methyl-3-phenyl-3H-quinazolin-4-one; 2-[2-benzyloxy-l-(9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-2-benzyloxy-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[2-benzyloxy-l-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one;
2-[2-benzyloxy-l-(2-fluoro-9H-purin-6-ylamino)-etlryl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 3-(4-fluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(4-fluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(4-fluoro-phenyl)-2-[1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3H-quinazolin-4-one;3-(4-fluoro-phenyl)-5-methyl-2-[

(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3Hquinazolin-4-one; 5-methyl-3-phenyl-2-[1-(7FI-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3-fluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;
2-[ 1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-fluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3-fluoro-phenyl)-5-methyl-2-[1-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-yl)-pyrrolidin-2-yl]-3H-quinazolin-4-one; 2-[2-hydroxy-l-(9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[phenyl-(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)-phenyl-methyl]=5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[(2-fluoro-9H-purin-6-ylamino)-phenyl-methyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[phenyl-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-methyl]-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3Hquinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-fluoro-3-phenyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-chloro-3-phenyl-3H-quinazolin-4-one; [5-(5-methyl-4-oxo-3-phenyl-3,4-dihydro-quinazolin-2-yl)-5-(9Hpurin-6-ylamino)-pentyl]-carbamic acid benzyl ester; [5-(2-amino-9H-purin-6-ylamino)-5-(5-methyl-4-oxo-3-phenyl-3,4-dihydro-quinazolin-2-yl)-pentyl]-carbamic acid benzyl ester;
[4-(5-methyl-4-oxo-3-phenyl-3,4-dihydro-quinazolin-2-yl)-4-(9H-purin-6-ylamino)-butyl]-carbamic acid benzyl ester; [4-(2-amino-9H-purin-6-ylamino)-4-(5-methyl-4-oxo-phenyl-3,4-dihydro-quinazolin-2-yl)-butyl]-carbamic acid benzyl ester; 3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[5-amino-l-(9H-purin-6-ylamino)-pentyl]-5-methyl-3-phenyl-3H-quinazolin-4-one); 2-[5-amino-1 -(2-amino-9Hpurin-6-ylamino)-pentyl]-5-methyl-3-phenyl-3Hquinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-Dimethyl-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(2,6-dimethyl-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-5 quinazolin-4-one; 5-morpholin-4-ylmethyl-3-phenyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-morpholin-4ylmethyl-3-phenyl-3H-quinazolin-4-one; 2-[4-amino-l-(2-amino-9Hpurin-6-ylamino)-butyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-10 ylamino)-ethyl]-6-fluoro-3-phenyl-3H-quinazolin-4-one; 2-[2-tert-butoxy-l-(9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 3-(3-methyl-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-methyl-phenyl)-5-methyl-3Hquinazolin-4-one; 3-(3-chloro-phenyl)-5-methyl-2=[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-15 4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-chloro-phenyl)-5-methyl-3H-quinazolin-4-one; 2-[ 1-(2-amino-9H-purin-6-ylamino)-2-hydroxy-ethyl]-5-methyl-phenyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-fluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-difluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-20 5-fluoro-3-phenyl-3H-quinazolin-4-one; 5-chloro-3-(3-fluoro-phenyl)-2-[1-(9H-purin-6=ylamino)-ethyl]-3Hquinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-chloro-3-(3-fluoro-phenyl)-3H quinazolin-4-one; 3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-5-trifluoromethyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-methyl-2-[ 1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-(2,6-difluoro-25 phenyl)-5-methyl-2-[ 1-(9Hpurin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[
1-(2-amino-9H-purin-6-ylamino)-propyl]-3-(2,6-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3,5-dichloro-phenyl)-5-methyl-2-[1-(9H-purin-ylamino)-ethyl]-3Hquinazolin-4-one; 3-(2,6-dichloro-phenyl)-5-methyl-2-[1-(9FI-30 purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-dichloro-phenyl)-5-methyl-3H-quinazolin-4-one; 5-chloro-3-phenyl-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[l-(2-amino-9H-purin-6-ylamino)-propyl]-5-chloro-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-butyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-butyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[ 1-(2-amino-9H-purin-ylamino)-ethyl]-3-(3,5-dichloro-phenyl)-5-methyl-3H-quinazolin-4-one; 5-methyl-(3-morpholin-4-ylmethyl-phenyl)-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-3-(3-morpholin-4-ylmethyl-phenyl)-3H-quinazolin-4-one; 2-[1-(5-bromo-7Hpyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-2-[1-(5-methyl-7H-pyrrolo[2,3-a']pyrimidin-4-ylamino)-ethyl]-3-phenyl-3H-quinazolin-4-one; 2-[1-(5-fluoro-7H-pyrrolo [2,3 -d]pyrimidin-4-ylamino)-ethyl}-5-methyl-3-phenyl-3H
quinazolin-4-one; 2-[2-hydroxy-l-(9H-purin-6-ylamino)-ethyl]-3-phenyl-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[ 1-(2-amino-9H-purin-6-ylamino)-propyl]-3-(3,5-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H=quinazolin-4-one; 2-[1-(5-bromo-7HHpyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3-(3-fluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3-fluoro-phenyl)-5-methyl-2-[1-(5-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one; 3-phenyl-2-[ 1-(9H-,purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[ 1-(2-amino-9Hpurin-6-ylamino)-ethyl]-3-(3,5-difluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9Hpurin-6-ylamino)-propyl]-3-phenyl-3H-quinazolin-4-one; 6,7-difluoro-3-phenyl-2-[1-(9Hpurin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 6-fluoro-3-(3-fluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[4-diethylamino-l-(9H-purin-6-ylamino)-butyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[ 1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-one; 5-fluoro-2-[1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-3-phenyl-3H-quinazolin-one; 3-(3-fluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;

chloro-3-(3,5-difluoro-phenyl)-2-[ 1-(9H-purin-6-ylamino)-propyl}-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-Methyl-3-phenyl-2-[3,3,3-trifluoro-l-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-(3-hydroxy-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3-methoxy-phenyl)-5-methyl-2- { 1-[9H-purin-6-ylamino]-ethyl}-3H-quinazolin-4-one; 3-[3-(2-dimethylamino-ethoxy)-phenyl]-5-methyl-2- { 1-[9H-purin-6-ylamino]-ethyl} -3H-quinazolin-4-one; 3-(3-cyclopropylmethoxy-phenyl)-5 -methyl-2-{ 1-[9H-purin-6-ylamino]-ethyl} -3H-quinazolin-4-one; 5-methyl-3-(3-prop-2-ynyloxy-phenyl)- 2-{1-[9H-purin-6-ylamino]-ethyl}-3H-quinazolin-4-one; 2-{1-[2-amino-9H-purin-6-ylamino]ethyl}-3-(3-hydroxyphenyl)-5-methyl-3H-quinazolin-4-one; 2- { 1-[2-amino-9H-purin-6-ylamino]ethyl}-3-(3-methoxyphenyl)-5-methyl-3H-quinazolin-4-one; 2-{1-[2-amino-9H-purin-6-ylamino]ethyl} -3-(3-cyclopropylmethoxy-phenyl)-5-methyl-3H-quinazolin-4-one; 2-{1-[2-amino-9H-purin-6-ylamino]ethyl}-5-methyl-3-(3-prop-2-ynyloxy-phenyl)-3H-quinazolin-4-one; 3-(3-ethynyl-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-{5-methyl-4-oxo-2-[1-(9H-purin-ylamino)-ethyl]-4H-quinazolin-3-yl} -benzonitrile; 3- {5-methyl-4-oxo-2- { 1-[9H-purin-6-ylamino)-ethyl]-4H-quinazolin-3-yl}-benzamide; 3-(3-acetyl-phenyl)-5-methyl-2- { 1-[9H-purin-6-ylamino]-ethyl} -3H-quinazolin-4-one; 2-(3-(5-methyl-oxo-2-{1-[9H-purin-6-ylamino]-ethyl}=4H-quiriazolin-3-yl-phenoxy acetamide; 5-methyl-2-{1-[9H-purin-6-ylamino]-ethyl}-3-[3-(tetrahydropuran-4-yloxy)-phenyl]-3H-quinazolin-4-one; 3-[3-(2-methoxy-ethoxy)-phenyl]-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; .6-fluoro-2-[ 1-(9H-purin-6-ylamino)ethyl]-3-[3-(tetrahydro-pyran-4-yloxy)-phenyl]-3H-quinazolin-4-one; 3-[3-(3-dimethylamino-propoxy)-phenyl]-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;
2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-ethynyl-phenyl)-5-methyl-3H-quinazolin-4-one; 3- {2-[ 1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-4-oxo-quinazolin-3-yl} -benzonitrile; 3- {2-[ 1-(2-amino-9H-purin-6-ylamino)-ethyl]-methyl-4-oxo-4H-quinazolin-3-yl}-benzamide; 3-{2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-4-oxo-4H-quinazolin-3-yl}-benzamide; 5-methyl-3-(3-morpholin-4-yl-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-3-(3-morpholin-4-yl-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-[3-(2-methoxy-ethoxy)-phenyl]-5-methyl-3H-quinazolin-4-one; 2-[ 1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-[3-(2-dimethylamino-ethoxy)-phenyl]-5-methyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-but-3-ynyl]-5-methyl-3-phenyl-3H-quinazolin-4-one;
2-[ 1-(2-amino-9H-purin-6-ylamino)-but-3-ynyl]-5-methyl-3-phenyl-3H-quinazolin-one; 5-chloro-3-(3,5-difluoro-phenyl)-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-5-chloro-3-(3,5-difluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-chloro-3-(3,5-difluoro-phenyl)-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-6-fluoro-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-chloro-3-(2,6-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl}-5-chloro-3-(2,6-difluoro-phenyl)-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-yloxy)-ethyl]-3H-quinazolin-4-one;
and mixtures thereof..

Where a stereocenter is present, the methods can be practiced using a racemic mixture of the compounds or a specific enantiomer. In preferred emliodiments, the S-enantiomer of the'above compounds is utilized. Therefore, the present invention includes all possible stereoisomers and geometric isomers of the aforementioned compounds.

"Pharmaceutically acceptable salts" means any salts that are physiologically acceptable insofar as they are compatible with other ingredients of the formulation and not deleterious to the recipient thereof. Some specific preferred examples are: acetate, trifluoroacetate, hydrochloride, hydrobromide, sulfate, citrate, tartrate, glycolate, oxalate.

In one embodiment, the invention contemplates an article of manufacture comprising a phosphoinositide 3-kinase delta (PI3KS) selective inhibitor, and a label indicating a method of use according to any one of the methods of the invention. In a related embodiment, the invention provides for use of a composition comprising at least one PI3K8 selective inhibitor in the manufacture of a medicament for treating or preventing a condition associated with undesirable mast cell activity.

Administration of prodrugs are also contemplated. The term "prodrug"
as used herein refers to compounds that are rapidly transformed in vivo by hydrolysis to, for example, a compound having a structural formula described herein.
Prodrug design is discussed generally in Hardma et al. (Eds.), Goodnzan and Gilnian's The Plaarmacological Basis of Therapeutics, 9tla ed., pp. 11-16 (1996). A thorough discussion is provided in Higuchi et al., Prodrugs as Novel Delivery Systenas, Vol. 14, ASCD Symposium Series, and in Roche (ed.), Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press (1987).
Briefly, administration of a drug is followed by elimination from the body or some biotransformation whereby biological activity of the drug is reduced or eliminated.
Alternatively, a biotransformation process can lead to a metabolic by-product, which is itself more active or equally active as compared to the drug initially administered.
Increased understanding of these biotransformation processes permits the design of so-called "prodrugs," which, following a biotransformation, become more physiologically active in their altered state. Prodrugs, therefore, encompass pharmacologically inactive compounds that are converted to biologically active metabolites.

To illustrate, prodrugs can be converted into a pharmacologically active form through hydrolysis of, for example, an ester or amide linkage, thereby introducing or exposing a functional group on the resultant product. The prodrugs can be designed to react with an endogenous compound to form a water-soluble conjugate that fizrther enhances the pharmacological properties of the compound, for example, increased circulatory half-life. Alternatively, prodrugs can be designed to undergo covalent modification on a functional group with, for example, glucuronic acid, sulfate, glutathione, amino acids, or acetate. The resulting conjugate can be inactivated and excreted in the urine, or rendered more potent than the parent compound. High molecular weight conjugates also can be excreted into the bile, subjected to enzymatic cleavage, and released back into the circulation, thereby effectively increasing the biological half-life of the originally adniinistered compound.

Additionally, compounds that selectively negatively regulate p110S
mRNA expression more effectively than they do other isozymes of the P13K
family, and that possess acceptable pharmacological properties are contemplated for use as PI3K8 selective inhibitors in the methods of the invention. Polynucleotides encoding human p110S are disclosed, for example, in Genbank Accession Nos. AR255866, NM
005026, U86453, U57843 and Y10055, the entire disclosures of which are incorporated herein by reference. See also, Vanhaesebroeck et al., Proc. Natl.
Acad.
Sci. 94:4330-4335 (1997), the entire disclosure of which is incorporated herein by reference. Representative polynucleotides encoding mouse pI lOS are disclosed, for example, in Genbank Accession Nos. BC035203, AK040867, U86587, and NM 008840, and a polynucleotide encoding rat p1106 is disclosed in Genbank Accession No. XM 345606, in each case the entire disclosures of which are incorporated herein by reference.

In one embodiment, the invention provides methods using antisense oligonucleotides which negatively regulate p110S expression via hybridization to messenger RNA (mRNA) encoding p110S. In one aspect, antisense oligonucleotides at least 5 to about 50 nucleotides in length, including all lengths (measured in number 5 of nucleotides) in between, which specifically hybridize to mRNA encoding p and inhibit mRNA expression, and as a result p110S protein expression, are contemplated for use in the methods of the invention. Antisense oligonucleotides include those comprising modified internucleotide linkages and/or those comprising modified nucleotides which are known in the art to improve stability of the 10 oligonucleotide, i.e.; make the oligonucleotide more resistant to nuclease degradation, particularly in vivo. It is understood in the art that, while antisense oligonucleotides that are perfectly complementary to a region in the target polynucleotide possess the highest degree of specific inhibition, antisense oligonucleotides that are not perfectly complementary, i.e., those which include a limited number of mismatches with 15 respect to a region in the target polynucleotide, also retain high degrees of hybridization specificity and therefore also can inhibit expression of the target mRNA. Accordingly, the invention contemplates methods using antisense oligonucleotides that are perfectly complementary to a target region in a polynucleotide encoding p110S, as well as methods that utilize antisense 20 oligonucleotides that are not perfectly complementary (i.e., include mismatches) to a target region in the target polynucleotide to the extent that the mismatches do not preclude specific hybridization to the target region in the target polynucleotide.
Methods for designing and optimizing antisense nucleotides are described in Lima et al., (JBiol Chein ;272:626-38. 1997), Kurreck et al., (Nucleic Acids Res.
;30:1911-8.
25 2002) and U.S. Patent No. 6,277,981, which are incorporated herein by reference.
Exemplary antisense compounds are described in International Patent Publication WO
01/05958, which is incorporated herein by reference.

The invention further contemplates rimethods utilizing ribozyme inhibitors which, as is known in the art, include a nucleotide region which specifically 30 hybridizes to a target polynucleotide and an enzymatic moiety that digests the target polynucleotide. Specificity of ribozyme inhibition is related to the length the antisense region and the degree of complementarity of the antisense region to the target region in the target polynucleotide. The methods of the invention therefore contemplate ribozyme inhibitors comprising antisense regions from 5 to about nucleotides in length, including all nucleotide lengths in between, that are perfectly complementary, as well as antisense regions that include mismatches to the extent that the mismatches do not preclude specific hybridization to the target region in the target p 1 l 08-encoding polynucleotide. Ribozymes useful in methods of the invention include those comprising modified internucleotide linkages and/or those comprising modified nucleotides which are known in the art to improve stability of the oligonucleotide, i.e., make the oligonucleotide more resistant to nuclease degradation, particularly in vivo, to the extent that the modifications do not alter the ability of the = ribozyme to specifically hybridize to the target region or diminish enzymatic activity of the molecule. Because ribozymes are enzymatic, a single molecule is able to direct digestion of multiple target molecules thereby offering the advantage of being effective at lower concentrations than non-enzymatic antisense oligonucleotides.
Preparation and use of ribozyme technology is described in U.S. Patent Nos.
6,696,250, 6,410,224, 5,225,347, the entire disclosures of which are incorporated herein by reference.

The invention also coritemplates use of methods in which RNAi technology is utilized for inhibiting pl 108 expression. In one aspect, the invention provides double-stranded RNA (dsRNA) wherein one strand is complementary to a target region in a target p1108-encoding polynucleotide. In general, dsRNA
molecules of this type are less than 30 nucleotides in length and referred to in the'art as short interfering RNA (siRNA). The invention also contemplates, however, use of dsRNA molecules longer than 30 nucleotides in length, and in certain aspects of the invention, these longer dsRNA molecules can be about 30 nucleotides in length up to 200 nucleotides in length and longer, and including all length dsRNA molecules in between. As with other RNA inhibitors, complementarity of one strand in the dsRNA
molecule can be a perfect match with the target region in the target polynucleotide, or may include mismatches to the extent that the mismatches do not preclude specific hybridization to the target region in the target p1106-encoding polynucleotide. As with other RNA inhibition technologies, dsRNA molecules include those comprising modified intemucleotide linkages and/or those comprising modified nucleotides which are known in the art to improve stability of the oligonucleotide, i.e., make the oligonucleotide more resistant to nuclease degradation, particularly in vivo.

Preparation and use of RNAi using double stranded (dsRNA) (Fire et al., Nature 391:
806-811. 1998) or short-interfering RNA (siRNA) sequences (Yu et al., Proc Natl Acad Sci USA. 99:6047-52. 2002) compounds are further described in U.S. Patent Application No. 20040023390, the entire disclosure of which is incorporated herein by reference.

The invention further contemplates methods'wherein inhibition of pl 108 is effected using RNA lasso technology. Circular RNA lasso inhibitors are highly structured molecules that are inherently more resistant to degradation and therefore do not, in general, include or require modified intemucleotide linkage or modified nucleotides. The circular lasso structure includes a region that is capable of hybridizing to a target region in a target polynucleotide, the hybridizing region in the lasso being of a length typical for other RNA inhibiting technologies. As with other RNA inhibiting technologies, the hybridizing region in the lasso may be a perfect match with the target region in the target polynucleotide, or may include mismatches to the extent that the mismatches do not preclude specific hybridization to the target region in the target pl 106-encoding polynucleotide. Because RNA lassos are circular and form tight topological linkage with the target region, inhibitors of this type are generally not displaced by helicase action unlike typical antisense oligonucleotides, and therefore can be utilized as dosages lower than typical antisense oligonucleotides.
20, Preparation and use of RNA lassos is described in U.S. Patent 6,369,038, the entire disclosure of which is incorporated herein by reference.

The inhibitors of the invention may be covalently or noncovalently associated with a carrier molecule, such as a linear polymer (e.g., polyethylene glycol, polylysine, dextran, etc.), a branched-chain polymer (see U.S. Patents 4,289,872 and 5,229,490; PCT Publication WO 93/21259 published 28 October 1993); a lipid; a cholesterol group (such as a steroid); or a carbohydrate or oligosaccharide.
Specific examples of carriers for use in the pharmaceutical compositions of the invention include carbohydrate=based polymers, such as trehalose, mannitol, xylitol, sucrose, lactose, sorbitol, dextrans, such as cyclodextran, cellulose, and cellulose derivatives.
Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated.

Other carriers include one or more water soluble polymer attachments such as polyoxyethylene glycol, or polypropylene glycol as described U.S.
Patent Nos: 4,640,835, 4,496,689, 4,301,144, 4,670,417, 4,791,192 and 4,179,337.
Still other useful carrier polymers known in the art include monomethoxy-polyethylene glycol, poly-(N-vinyl pyrrolidone)-polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol, as well as mixtures of these polymers.
Derivatization with bifunctional agents is useful for cross-linking a compound of the invention to a support matrix or to a carrier. One such carrier is polyethylene glycol (PEG). The PEG group may be of any convenient molecular weight and may be straight chain or branched. The average molecular weight of the PEG can range from about 2 kDa to about 100 kDa, in another aspect from about kDa.to about 50 kDa, and in a further aspect from about.5 kDa to about 10 kDa.
The PEG groups will generally be attached to the compounds,of the invention via acylation, reductive alkylation, Michael addition, thiol alkylation or other chemoselective conjugation/ligation methods through a reactive group on the PEG
moiety (e.g., an aldehyde, amino, ester, thiol, haloacetyl, maleimido or hydrazino group) to a reactive group on the target inhibitor compound (e.g., an aldehyde, amino, ester, thiol, a-haloacetyl, maleimido or hydrazino group). Cross-linking agents can include, e.g., esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiobis (succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane. Derivatizing agents such as methyl-3-[(p-azidophenyl)dithio]propioirnidate yield pliotoactivatable intermediates that are capable of forming crosslinks in the presence of light.
Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos.
3,969,287;
3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 may be employed for inhibitor immobilization.

The pharmaceutical compositions of the invention may also include compounds derivatized to include one or more antibody Fc regions. Fc regions of antibodies comprise monomeric polypeptides that may be in dimeric or multimeric forms linked by disulfide bonds or by non-covalent association. The number of intermolecular disulfide bonds between monomeric subunits of Fc molecules can be from one to four depending on the class (e.g., IgG, IgA, IgE) or subclass (e.g., IgG1, IgG2, IgG3, IgAl, IgGA2) of antibody from which the Fc region is derived. The term "Fc" as used herein is generic to the monomeric, dimeric, and multimeric forms of Fc molecules, with the Fc region being a wild type structure or a derivatized structure.
The pharmaceutical compositions of the invention may also include the salvage receptor binding domain of an Fe molecule as described in WO 96/32478, as well as other Fc molecules described in WO 97/34631.

Such derivatized moieties preferably improve one or more characteristics of the inhibitor compounds of the invention, including for example, biological activity, solubility, absorption, biological half life, and the like.
Alternatively, derivatized moieties result in compounds that have the same, or essentially the same, characteristics and/or properties of the compound that is not derivatized. The moieties may alternatively eliminate or attenuate any undesirable side effect of the compounds and the like.

Compounds that compete with an inhibitor compound described herein for binding to PI3K8 are also contemplated for use in the invention. Methods of identifying compounds which competitively bind with PI3K8, with respect to the compounds specifically provided herein, are well known in the art.

In view of the disclosures above, therefore, the term "inhibitor" as used herein embraces compounds disclosed, compounds that compete with disclosed compounds for PI3K8 binding, and in each case, conjugates and derivatives thereof.

Methods include administration of an inhibitor to an individual in need, by itself, or in combination as described herein, and in each case optionally including one or more suitable diluents, fillers, salts, disintegrants, binders, lubricants, glidants, wetting agents, controlled release matrices, colorants/flavoring, carriers, excipients, buffers, stabilizers, solubilizers, other materials well known in the art and combinations thereof.

Any pharmaceutically acceptable (i.e., sterile and non-toxic) liquid, semisolid, or solid diluents known in the art that serve as pharmaceutical vehicles, excipients, or media may be used. Exemplary diluents include, but are not limited to, polyoxyethylene sorbitan monolaurate, magnesium stearate, calcium phosphate, mineral oil, cocoa butter, and oil of theobroma, methyl- and propylhydroxybenzoate, talc, alginates, carbohydrates, especially mannitol, a-lactose, anhydrous lactose, cellulose, sucrose, dextrose, sorbitol, modified dextrans, gum acacia, and starch.

Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the present inhibitor compounds. See, e.g., Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack 5 Publishing Co., Easton, PA 18042) pages 1435-1712 which are herein incorporated by reference.

Pharmaceutically acceptable fillers can include, for example, lactose, microcrystalline cellulose, dicalcium phosphate, tricalcium phosphate, calcium sulfate, dextrose, mannitol, and/or sucrose.

10 Inorganic salts including calcium triphosphate, magnesium carbonate, and sodium chloride may also be used as fillers in the pharmaceutical compositions.
Amino acids may be used, such as use in a buffer formulation of the pharmaceutical compositions.

Disintegrants may be included in solid dosage formulations of the 15 inhibitors. Materials used as disintegrants include but are not limited to starch including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxyrnethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge, corn starch, potato starch, and bentonite may all be used as disintegrants in the 20 pharmaceutical compositions. Other disintegrants include insoluble cationic exchange resins. Powdered gums including powdered gums such as agar, Karaya or tragacanth may be used as disintegrants and as binders. Alginic acid and its sodium salt are also useful as disintegrants.

Binders may be used to hold the therapeutic agent together to form a 25 hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include crystalline cellulose, cellulose derivatives such as methyl cellulose (MC), etYzyl cellulose (EC) and carboxymethyl cellulose (CMC), acacia, corn starch, and/or gelatins Polyvinyl pyrrolidone (PVP) and hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to 30 granulate the therapeutic.

An antifriction agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process. Lubricants may be used as a layer between the therapeutic and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils, talc, and waxes.
Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added.
Suitable glidants include starch, talc, pyrogenic silica and hydrated silicoaluminate.

To aid dissolution of the therapeutic into the aqueous environment, a surfactant might be added as a wetting agent. Natural or synthetic surfactants may be used. Surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate, and dioctyl sodium sulfonate. Cationic detergents such as benzalkonium chloride and benzethoniuin chloride may be used. Nonionic detergents that can be used in the pharmaceutical formulations include lauromacrogol 400, polyoxyl,40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. These surfactants could be present in the pharmaceutical compositions of the invention either alone or as a mixture in different ratios.

Controlled release formulation may be desirable. The inhibitors of the invention could be incorporated into an inert matrix which permits release by either diffusion or leaching mechanisms, e.g., gums. Slowly degenerating matrices may also be incorporated into the pharmaceutical formulations, e.g., alginates, polysaccharides.
Another form of controlled release is a method based on the Oros therapeutic system (Alza Corp.), i.e., the drug is enclosed in a semipermeable membrane which allows water to enter and push the inhibitor compound out through a single small opening due to osmotic effects. Some enteric coatings also have a delayed release effect.

Colorants and flavoring agents may also be included in the pharmaceutical compositions. For example, the inhibitors of the invention may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.

The therapeutic agent can also be administered in a film coated tablet.
Nonenteric materials for use in coating the pharmaceutical compositions include methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl-methyl cellulose, sodium carboxy-methyl cellulose, povidone and polyethylene glycols. Enteric materials for use in coating the pharmaceutical compositions include esters of phthalic acid. A mix of materials might be used to provide the optimum film coating. Film coating manufacturing may be carried out in a pan coater, in a fluidized bed, or by compression coating.

Compositions can be administered in solid,. semi-solid, liquid or gaseous form, or may be in dried powder, such as lyophilized form. The pharniaceu.tical compositions can be packaged in forms convenient for delivery, including, for example,-capsules, sachets, cachets, gelatins, papers, tablets, capsules, ointments, granules, solutions, inhalants, aerosols, suppositories, pellets, pills, troches, lozenges or other forms known in the art. The type of packaging will generally depend on the desired route of administration. Implantable sustained release formulations are also contemplated, as are transdermal fonnulations.

Methods of the invention contemplate administration of inhibitor compounds by various routes. Such pharmaceutical compositions may be for administration for injection, or for oral, nasal, transdermal or other forms of administration, including, e.g., by intravenous, intradermal, intramuscular, intramammary, intraperitoneal, intratracheal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., aerosolized drugs) or subcutaneous injection (including depot administration for long term release e.g., embedded under the splenic capsule, brain, or in the cornea); by sublingual, anal, vaginal, or by surgical implantation, e.g., embedded under the splenic capsule, brain, or in the cornea. The treatment may consist of a single dose or a plurality of doses over a period of time. In general, the methods of the invention involve administering effective amounts of an inhibitor of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or camers, as described above. As is understood in the art, a chosen route of administration may dictate the physical form of the compound being delivered.

In one aspect, the invention provides methods for oral administration of a pharmaceutical composition of the invention. Oral solid dosage forms are described generally in Remington's Pharmaceutical Sciences, 18th Ed. 1990 (Mack Publishing Co. Easton PA 18042) at Chapter 89. Solid dosage forms include tablets, capsules, pilis, troches or lozenges, and cachets or pellets. Also, liposomal or proteinoid encapsulation may be used to formulate the present compositions (as, for example, proteinoid microspheres reported in U.S. Patent No. 4,925,673).
Liposomal encapsulation may include liposomes that are derivatized with various polymers. (e.g., U.S. Patent No. 5,013,556). In general, the formulation will include a compound of the invention and inert ingredients which protect against degradation in the stomach and which pennit release of the biologically active material in the intestine.

The inhibitors can be included in the formulation as fine multiparticulates in the form of granules or pellets of particle size about 1 mm. The formulation of the material for capsule adininistration could also be as a powder, 15' lightly compressed plugs or even as tablets. The capsules could be prepared by compression.

Also contemplated herein is pulmonary delivery of the present inhibitors in accordance with the invention. According to this aspect of the invention, the inhibitor is delivered to the lungs of a mammal while inhaling and traverses-across the lung epithelial lining to the blood stream.

Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art. Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent nebulizer, manufactured by Mallinckrodt, Inc., St. Louis, Missouri;
the Acorn H nebulizer, manufactured by Marquest Medical Products, Englewood, Colorado; the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, North Carolina; and the Spinhaler powder inhaler, manufactured by Fisons Corp., Bedford, Massachusetts.

All such devices require the use of formulations suitable for the dispensing of the inventive compound. Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to diluents, adjuvants and/or carriers useful in therapy.

When used in pulmonary administration methods, the inhibitors of the invention are most advantageously prepared in particulate form with an average particle size of less than 10 m (or microns), for example, 0.5 to 5 m, for most effective delivery to the distal lung.

Formulations suitable for use with a nebulizer, either jet or ultrasonic, will typically comprise the inventive compound dissolved in water at a concentration range of about 0.1 to 100 mg of inhibitor per mL of solution, 1 to 50 mg of inhibitor per mL of solution, or 5 to 25 mg of inhibitor per mL of solution. The formulation may also include a buffer. The nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the inhibitor caused by atomization of the solution in forining the aerosol.

Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the inventive inhibitors suspended in a propellant with the aid of a surfactant. The propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorod"ifluoromethane, dichlorotetrafluoro ethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof. Suitable surfactants include sorbitan trioleate and soya lecithin. Oleic acid may also be useful as a surfactant.
Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing the inventive compound and may also include a bulking agent or diluent, such as lactose, sorbitol, sucrose, mannitol, trehalose,'or xylitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation.

Nasal delivery of the inventive compound is also contemplated. Nasal delivery allows the passage of the inhibitor to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung. Formulations for nasal delivery may include dextran or cyclodextran. Delivery via transport across other mucous membranes is also contemplated.

Toxicity and therapeutic efficacy of the PI3K8 selective compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50%
of the population) and the ED50 (the dose therapeutically effective in 50% of the 5 population). Additionally, this information can be determined in cell cultures or experimental animals additionally treated with other therapies such as radiation, chemotherapeutic agents, and antiangiogenic agents.

In practice of the methods of the invention, the pharmaceutical compositions are generally provided in doses ranging from 1 pg compound/kg body 10 weight to 1000 mg/kg, 0.1 mg/kg to 100 mg/kg, 0.1 mg/kg to 50 mg/kg, and 1 to 20 mg/kg, given in daily doses or in equivalent doses at longer or shorter intervals, e.g., every other day, twice weekly, weekly,,or twice or three times daily. The inhibitor compositions may be administered by an initial bolus followed by a continuous infusion to maintain therapeutic circulating levels of drug product. Those of ordinary 15 skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient. The frequency of dosing will depend on the pharmacokinetic parameters of the agents and the route of administration. 'The optimal pharmaceutical formulation will be determined by one skilled in the art depending upon the route of 20 administration and desired dosage. See for example, Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042) pages 1435-1712, the disclosure of which is hereby incorporated by reference. Such formulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agents. Depending on the route of administration, a 25 suitable dose may be calculated according to body weight, body surface area or organ size. Further refinement of the calculations necessary to determine the appropriate dosage for treatment involving each of the above mentioned formulations is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein, as well as the 30 pharmacokinetic data observed in the human clinical trials discussed above.
Appropriate dosages may be ascertained through use of established assays for determining blood levels dosages in conjunction with appropriate physician, considering various factors which modify the action of drugs, e.g. the drug's specific activity, the severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. As studies are conducted, further information will emerge regarding the appropriate dosage levels and duration of treatment for various diseases and conditions.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 shows a count of inflammatory cell infiltration (total cells and differential cellular component) of BAL fluids isolated from OVA-sensitized/challenged mice treated with p 1105 inhibitor IC87114 or control in BAL
fluid. Macrophage (Mac), lymphocyte (Lym), neutrophils (Neu), and eosinophils (Eos) were counted. Bars represent mean SEM from 6 independent experiments.
#, p < 0.05 vs. SAL+SAL; p < 0.05 vs. OVA+SAL.

Figure 2 depicts total lung inflammation, defined as the average of the peribronchial and perivascular inflammation scores, in OVA-sensitized/challenged mice treated with IC87114 or control. Bars represent mean SEM from 6 independent experiments. #, p < 0:05 vs. SAL+SAL; *, p < 0.05 vs. OVA+SAL.

Figure 3 shows airway responsiveness to aerosolized methacholine measured in unrestrained, conscious OVA-sensitized/challenged mice treated with IC87114 or control, at 72 h after the last challenge. Readings of breathing parameters were taken for 3 min after each nebulization during which Penh values were determined. Data represent mean SEM from 6 independeiit experiments. #, p <
0.05 vs. SAL+SAL; *, p < 0.05 vs. OVA+SAL.

EXAMPLES
The following examples are provided to illustrate the invention, but are not intended to limit the scope thereof.

DEGRANULATION

Mast cells are centrally important in allergic inflammation of the airways. Mast cell activation is mediated by antigen/IgE cross-linking of the FcERI
receptor, complement components and cytokines activation, leading to a spontaneous release of preformed histamine and proteases from secretory granules. These activated cells then transcribe, translate and secrete proinflammatory cytokines and chemokines which lead to increased inflammation.

Non-specific P13-kinase inhibitors such as wortmannin have been shown to inhibit mast cell degranulation from rat basophilic leukemia cells (RBL-2H3) (Kitani et al., Biochem Biophys Res Commun. 183:48-54, 1992). However, the role for specific P13-kinase subunits in regulating mast cell degranulation remains unclear. [Tkaczyk et al., JBiol Claem. 278:48474-84 (2003); Windmiller et al., JBiol Cltem. 278:11874-8 (2003)].

To determirie the role of P13-kinase p 1105 subunit in regulating mast cell activity, the ability of specific p 1108 inhibitors to regulate human or mouse bone marrow derived mast cell degranulation was assessed.

Bone marrow cells from mouse femur were cultured in mouse IL-3-containing RPMI 1640 medium supplemented with 10% heat-inactivated FBS
(Invitrogen Life Technologies, Carlsbad, CA), 2 mM glutamine, and 50 M 2-ME
in humidified 95% air/5% CO2 at 37 C [Saito, H., F. et al., J. Irnmunol. 138:3927-(1987)]. More than 95% pure mast cells (bone marrow-derived mast cells (BMMC)) were obtained after 5 wk of culture.
BMMCs were sensitized by an overnight incubation with 0.5-1 g/ml antidinitrophenyl (DNP) IgE mAb, washed once in Tyrode buffer (112 mM NaCl, 2.7 mM KCl, 0.4 mM NaHaPO4, 1.6 mM CaCl2, 1 mM MgC12, 10 mM Hepes [pH 7.5], 0.05% gelatin, 0.1 % glucose), resuspended in Tyrode buffer to 1 x 107 cells/ml and incubated with pl 10-delta inhibitor or vehicle control (0.3% DMSO) for 30 minutes.
Cells were then stimulated by polyvalent antigen, 0.1-100 ng/ml DNP conjugates of human serum albumin (DNP-HSA) for 45 minutes. Cell suspensions were briefly centrifuged to separate Tyrode solution from cell pellets. Histamine secreted into the Tyrode solution during 45-min stimulation was measured by enzyme immunoassay (Cayman Chemical Company, Ann Arbor, MI). Percent inhibition of histamine release by an inhibitor was expressed by the following formula: (1 -(%
histamine release in the presence of an inhibitor)/(% histamine release from control cells)) x 100%.

Results of the ELISA indicate that mast cells pretreated with IC87114 (5 M) reduce the amount of histamine in the cell supernatant by approximately 40%
compared to vehicle control, and increase the amount of histamine detected in the cell pellet accordingly. This demonstrates that inhibition ofp110S kinase reduces the amount of histamine released from mast cell storage granules which has utility as an effective therapy for decreasing histamine release from mast cell activation mediated ' through FcsRI.

CYTOKINE SECRETION BY MAST CELLS

A significant effect of FcsRI crosslinking of mast cells is the induction of numerous inflammatory cytokines such as IL-2, IL-3, IL-4, IL-6, IL-13, GM-CSF, and TNF-a. Both TNF-a and IL-2 are known to play a role in late phase hypersensitivity reactions like those in asthma and allergy [Galli et al., Fundamental In2naunology, 4th Ed., Paul ed., Lippincott-Raven Publishers, Philadelphia, PA, (1998); p. 1127-1174]. To determine the effect p1108 kinase inhibitors have on the downstream effects of mast cell activation, the ability of a selective p1108 inhibitor to modulate the secretion of inflammatory cytokines after mast cell activation was examined.

BMMCs were sensitized by an overnight incubation with 0.5-1 g/ml antidinitrophenyl (DNP) IgE mAb, washed once in BMMC medium in the absence of IL-3 and resuspended in the same medium at 1 x 107 cells/ml. Cells were incubated with p110S inhibitor or vehicle control (0.3% DMSO) for 30 minutes and then stimulated by polyvalent antigen, 1-100 ng/ml DNP conjugates of human serum albumin (DNP-HSA) for 20 hours. The cell supernatant was collected and the levels of inflammatory cytokines assessed by ELISA (BD PharMingen or Endogen). The cytokines measured include TNF-a, IL-6, IL-2, and granulocyte/monocyte-colony stimulating factor (GM-CSF).

Results show that stimulation with 10 ng/ml antigen and addition of only 2 M IC87114 to mast cells decreases the amount of TNF-a secreted by mast cells to undetectable levels. Additionally, 100 ng/ml of antigen plus 3 M
inhibitor inhibits TNF-a production from activated mast cells by 85%.

Assessment of IL-6 levels after addition of the p110S kinase inhibitor demonstrates similar effects. Addition of IC87411 to activated mast cells (10 ng/mi antigen) showed a dose dependent reduction in IL-6 production, from 6000 pg/ml with no inhibitor, reduced to approximately 2000 pg/ml IL-6 in the presence of inhibitor. Bone marrow mast cells isolated from p1108 knockout mice were also assessed for IL-6 secretion levels after activation by antigen (100 ng/ml).
Addition of p1108 inhibitor (up to 10 gM) had no effect on levels of IL-6 in knockout cells, indicating that the effect of the inhibitor is through inhibition of the protein kinase p1108isoform. GM-CSF secretion by activated mast cells was also inhibited by approximately 45% after addition of the p1106 inhibitor.

' Levels of the inflammatory cytokine IL-2 were also measured from antigen stimulated BMMC. BMMC activated with 10 ng/ml antigen showed a dose response inhibition of IL-2 secretion upon addition of p110S inhibitor, with levels decreased from control values of approximately 10 pg/ml to < 4 pg/ml IL-2 in the presence of 20 M inhibitor, a 2.5 fold reduction in IL-2 secretion.

Chemokines play a significant role in attracting cells to the site of inflammation thereby promoting the inflammatory response. Antigen activated BM
mast cells were assessed for chemokine production in the presence and absence of p110S inhibitor using Multi-Analyte Profile Technology (Rules-Based Medicine, Inc.
Austin, TX). The effects ofp1108 inhibitor (5 M) on chemokine production from activated mast cells resulted in approximately 57% inhibition of eotaxin levels, approximately 37% inhibition of lymphotactin, approximately 60% inhibition of macrophage-derived chemokine (MDC) secretion, and approximately 35% inhibition in the levels of macrophage-inflammatory proteins MIP-la and MIP-1(3.

These results demonstrate that specific inhibition of p110S reduces the amount of inflammatory cytokine and chemokine secreted as a result of antigen cross linking of FcsRI on the surface of mast cells. The ability to target and inhibit pI 108 activation in these stimulated mast cells provides a method for an improved, specific method for treatment of mast cell related disorders, and a useful therapeutic for downregulating aberrant mast cell activation.

IN ANTIGEN ACTIVATED MAST CELLS

P13K p 1105 is involved in a complex signaling pathway leading to phospholipase C activation and influx of calcium into the cell. Akt is a downstream target to P13 kinases that gets phosphorylated at serine 243 in response to 10 activation. The phosphorylation event is essential to the kinase activity of Akt. Akt activation has previously been associated with secretion of cytokines from activated rat basophilic leukemia cells or cells isolated from tyrosine kinase knockout mice [Kitaura et al., J. Exp. Med. 192:729-39 (2000)]. Broad inhibition of class Ia PI3Ks in certain cell types, such as endothelium, using LY294002 has been shown to not 15 only to reduce phosphorylation of Akt in response to TNF-a, but also in non-cytokine stimulated cells, as these lipid kinases are essential for both cell motility and survival.
[See Madge et al., J Biol. Chem., 275:15458-15465 (2000)]. Because Akt activation occurs through several different biochemical pathways, it was necessary to determine the role P13K p 1108 played in triggering Akt phosphorylation in activated mast cells.
20 To assess the effects of PI3K inhibitor on downstream signaling, BMMC were stimulated with a dose range of antigen and incubated with IC87114 as above with 0, 2 M, 5 M, 20 gM or 50 M inhibitor. Cells were lysed as described above and phsophoserine or phosphotyrosine levels for several p1106 downstream effectors were measured by Western blot. Antibodies used in the Western analysis 25 include: Akt pS473; 3-phosphoinositide-dependent kinase (PDK1) pS241; pERK;
pJNK; Lyn pY396 and Bruton's tyrosine kinase (Btk) pY223. Antibodies were obtained from Cell Signaling Technology (Beverly, MA), Upstate Biotechnology (Lake Placid, NY), Zymed, and Santa Cruz Biotechnology..

Western blot analysis demonstrated that p 1105 inhibitor as low as 5 30 M decreased Akt phosphorylation after 10 minutes of antigen stimulation and significantly inhibited phosphorylation after 30 minutes of antigen stimulation. The 50 gM inhibitor dose again significantly inhibited S243 Akt phosphorylation after 2 minutes of antigen stimulation, and completely abolished phosphorylation at 10 or 30 minutes of antigen stimulation. Analysis of other downstream effectors showed that antigen/IgE stimulation induced phosphorylation of PDK1, ERK, JNK, Lyn and Btk, but addition of pl 108 inhibitor did not affect the phosphorylation levels of any of these molecules. Additionally, overall tyrosine or serine phosphorylation for the cell was not affected by the p 1106 inhibitor. Protein levels of all molecules tested remained constant indicating that the decrease in Akt phosphorylation was specifically due to inhibition of p1106 activity.

These results indicate that selective inhibition of P13 K pl 108 in activated mast cells specifically decreases the levels of Akt phosphorylation without affecting phosphorylation levels of other related downstream tyrosine or serine kinases. This indicates that pl 108 selective inhibitor regulation of Akt, which is involved in mast cell cytokine secretion, will provide an effective therapeutic method for modulating ongoing inflammation in an already developed mast cell di"sorder by reducing cellular infiltrate to the site of inflammation, or may act preventatively in disorders such as allergy or asthma to prevent the onset of aii immune reaction.

ALLERGEN-INDUCED AIRWAY INFLAMMATION LEADS TO

P13K activity is stimulated after antigen challenge in a murine model of allergic asthma, and administration of wortmannin or LY294002, two broad-spectrum inhibitors of PI3Ks, attenuate inflammation and airway hyperresponsiveness (AHR) (Ezeamuzie et al., Am JRespir Crit Care Med 164:1633-39, 2001; Kwak et al., J. Clin. Invest 111:1083-92, 2003). However, these inhibitors do not distinguish among the four class I PI3Ks (Davies et al., Biochem J. 351:95-105, 2000;
Sadhu et al., J. Immunol 170:2647-54, 2003) and also broadly impact multiple cell types that express these kinases. Previous studies have indicated a crucial role for p110S in B
and T cell antigen receptor signaling and activation (Okkenhaug et al., Science 297:1031-34, 2002; Clayton et al., JExp Med 196:753-63, 2002) and, neutrophil migration and activation (Puri et al., Blood 103:3448-56, 2004; Sadhu et al., Biochem Biopltys Res Commun 308:764-69, 2003). In addition, pl 106 was reported to be essential for allergen-IgE-induced mast cell degranulation and vascular permeability (Ali et al., Nature 431:1007-11, 2004). This study did not determine if blockade of activity of this leukocyte and endothelial cell specific P13K isoform, p110S
alone would be sufficient to attenuate allergic inflammation and AHR. To examine the role of p110S in the pathogenesis of asthma the effects of administration of a p110S
inhibitor was investigated in animal models of asthma.

Female BALB/c mice, 8-10 weeks of age and free of murine-specific pathogens, were obtained from the Korean Research Institute of Chemistry Technology (Daejon, Korea). Mice were housed throughout the experiments in a laminar flow cabinet, and were maintained on standard laboratory chow ad libitum.
All experimental animals used in this study were under a protocol approved by the Institutional Animal Care and Use Committee of the Chonbuk National University Medical School. Mice were sensitized on days 1 and 14 by intraperitoneal injection of 20 g ovalbumin (OVA) (Sigma-Aldrich, St. Louis, MO) emulsified in 1 mg of aluminum hydroxide (Pierce Chemical Co., Rockford, IL) in a total volume of 200 l, as previously described (Kwak et al., supra). On days 21, 22, and 23 after the initial sensitization, the mice were challenged for 30 min with an aerosol of 3%
(wt/vol) OVA in saline (or with saline as a control) using an ultrasonic nebulizer (NE-U12, Omron, Japan). IC87114 (0.1 or 1 mg/kg body weight/day) or DMSO (vehicle control) diluted with 0.9% NaCl, was administered in a volume of 50 l by intratracheal instillation two times to each animal, once on day 21 (1 h before the first 20, airway challenge with OVA) and the second time on day 23 (3 h after the last airway challenge with OVA). All statistical comparisons were performed using one-way ANOVA followed by the Fisher's test.. Significant differences between groups were determined using the unpaired Student's t test. Statistical significance was set at p <
0.05.

In OVA-exposed mice, class I P13K activity was measured using protein extracts from lung tissue homogenates prepared in the presence of protease inhibitors, as described previously (Kwak et al., supra). Protein concentrations were determined using the Bradford reagent (Bio-Rad, Hercules, CA) and P13K
activity in the tissue extracts was quantified by PIP3 competition enzyme inuiiunoassays according to the manufacturer's protocol (Echelon, Inc., Salt Lake City, UT).

levels increased from approximately 15 pmol/ ml pre-challenge to approximately pmol/ml at 1 hr, approximately 100 pmol/ml at 24 hr, and approximately 160 pmol/ml at 48 hrs and 72 hrs, indicating that class 1 P13K activity increased approximately 4.6-6.1-, 9.5-, and 9.6-fold, respectively, after OVA inhalation, compared with the pre-challenge period. In contrast, no significant changes in P13K activity were observed after saline inhalation.

Activation of these kinases has been linked to phosphorylation of Ser-473 of Akt, an event crucial for Akt enzymatic activation (Alessi et al., Curr. Biol.
7:261-69, 1997). Akt levels were measured by Western blot. Protein extracts from lung tissue homogenates (30 g/lane) were electrophoresed in polyacrylamide gels (Invitrogen Life Technologies, Carlsbad, CA), transferred electrophoretically to a PVDF membrane (Imxnobilon-P; Millipore, Billerica, MA), and incubated overnight at 4 C with anti phoshposerine antibodies as described above. Consistent with increased P13K activity, levels of p-Akt protein in the lung tissues were also increased at 72 h after OVA inhalation, as detected by Western blot, compared with levels in the control animals that received saline inhalation. No significant changes in total Akt protein levels were observed. Intratracheal administration of a selective p110b inhibitor, IC8 7114, blocked Akt phosphorylation. The level of phosphorylation was similar to saline control levels indicating that p1108 contributed significantly to the overall class I P13K activity in allergen-induced Akt activation.

While PIP3 formation and Akt phosphorylation indicated that the P13K
family plays a role in allergy and asthma, administration of the p1108 specific inhibitor described herein demonstrates that the important P13K for allergy and asthma is pl 105.

25, IN BAL FLUID

The experiments above established that p110S mediates allergen-induced Akt activation. To determine the effect of p1106 kinase in eosinophil recruitment following allergen challenge; the number of inflammatory cells in the lung was assessed in OVA-challenged animals.

Mice were administered ovalbumin as described in Example 4 above and bronchialveolar lavage (BAL) fluid was collected 72 h after the last OVA
aerosol challenge, and total leukocyte and differential cell counts were performed. At hours after the last challenge, mice were sacrificed with an overdose of pentobarbital-Na (100 mg/kg of body weight, administered intraperitoneally). Blood was drawn by puncture of the vena cava and centrifuged. Serum was shock frozen in liquid nitrogen and stored at -70 C for IgE measurements. BAL was performed as described (Kwak et al., supra). Briefly, the chest cavity was exposed to allow for expansion, after which the trachea was carefully intubated and the catheter secured with ligatures.
Pre-warmed 0.9% NaCl solution was slowly infused into" tlie lungs and withdrawn.
Total BAL cells were counted using a hemocytometer. Differential cell counts were obtained from BAL cells spun down onto slides with a cytocentrifuge (Shannon Scientific Ltd., Cheshire, United Kingdom) and treated with Diff-Quik solution (Dade Diagnostics of Puerto Rico Inc. Aguada, Puerto Rico). Two independent, blinded investigators counted the cells using a microscope. Approximately 400 cells, in each of four different random locations were counted. The mean number from the two investigators was used to estimate the cell differentials. For cytokine and leukotriene measurements, supematants of BAL were shock frozen in liquid nitrogen and stored at -70 C until use.

OVA inhalation significantly (p < 0.05) increased the absolute numbers of eosinophils, lymphocytes, and neutrophils, as compared with saline control (Figure 1). Intratracheal administration of IC87114 (0.1 mg/kg) reduced the number of eosinophils, lymphocytes, and neutrophils detected in BAL fluids at hours post challenge by 79.8%, 63.5%, and 80%, respectively, compared withmice treated with vehicle control. In contrast, the number of macrophages was not affected by IC87114. No eosinophils were found in unsensitized control animals, whereas many eosinophils were present in BAL fluids of allergen-treated mice.
Reduction in the total cell number recovered in BAL fluid in IC87114-treated mice as compared with vehicle control, was mainly due to a significant (p < 0.05) reduction in eosinophils in tlle IC87114-treated mice. These results indicate that p1106 activity contributed to eosinophil recruitment during the allergic inflammatory response.

To determine if p 1108 affects OVA-induced tissue eosinophilia, mucus production, and airway inflammation, lung tissue was extracted and histological analysis performed.

Lung tissues were collected 72 h after the last OVA challenge. Mice were sacrificed and the lungs and trachea were filled intratracheally with a fixative (0.8% formalin, 4% acetic acid) using a ligature around the, trachea. Lungs were removed and lung tissues were fixed with 10% (vol/vol) neutral buffered formalin.
The specimens were dehydrated and embedded in paraffin. For histological examination, 4 gm sections of fixed embedded tissues were cut on a Leica model 2165 rotary microtome (Leica Microsystems Nussloch GinbHõ Nussloch, Germany), 5 placed on glass slides, deparaffinized, and stained with hematoxylin 2, cosin-Y
(Richard-Allan Scientific, Kalamazoo, MI) and periodic acid-Schiff (PAS).
Three independent blinded investigators graded the inflammation score. The degree of peribronchial and perivascular inflammation was evaluated on a subjective scale of 0 to 3, as described in Kwak et al (supra). A value of 0 was adjudged when no 10 inflammation was detectable, a value of 1 for occasional cuffing with inflammatory cells, a.value of 2 for most bronchi or vessels surrounded by thin layer (one to five cells) of inflammatory cells, and a value of 3 when most bronchi or vessels were surrounded by a thick layer (more than five cells) of inflammatory cells.

Histological analyses revealed typical pathologic features of asthma-15 like inflammation in the OVA-exposed mice. In contrast to the saline controls, OVA-exposed mice showed numerous inflammatory cells in the peribronchiolar zone and accumulation of mucus and cellular debris within the lumen of the bronchioles.
In contrast, IC87114-treated mice showed substantial attenuation in the eosinophil-rich leukocyte infiltration in the peribronchiolar regions and in the amount of debris 20 present in the lumen. In addition, representative sections of each group were stained with periodic acid-Schiff (PAS) for detection of goblet cells. As compared to the control, OVA-exposed mice exhibited severe goblet cell hyperplasia in the airways, which was markedly reduced by IC87114-treatment. The inflammation scores of peribronchial, perivascular regions as well as total lung were increased significantly 25 (p < 0.05) at 72 h after OVA inhalation compared with scores after saline inhalation (Figure 2). The increased lung inflammation observed after OVA inhalation was reduced by more than 50% by administration of the p110fi inhibitor.

These results indicate that p1108 inhibitor significantly reduces allergen-induced leukocyte influx, airway inflammation, and goblet cell hyperplasia.

ALLERGEN INDUCED AIRWAY INFLAMMATION

Eosinophil accumulation and subsequent activation in bronchial tissues is known to play a critical role in the pathogenesis of allergic airway inflammation (Busse et al., NEngl JMed 344:35062, 2001; Humbles et al., Science 305:1776-79, 2004). Eosinophil transmigration into the airways is a multistep process that is orchestrated by Th2 cytokines (IL-4, IL-5, and IL-13), and coordinated by specific chemokines such as eotaxin.in combination with adhesion molecules such as VCAM-1 and VLA-4 (10, 11). IL-13 is a potent inducer of eotaxin expression in airway epithelial cells (Tigani et al., Eur JPharmacol 433:217-23, 2001) Given the essential role of Th2 cytokines in evoking allergic inflammatory responses, the concentrations of IL=4, IL-5, and IL-13 were measured from BAL fluid as well as in'the lung tissue of OVA-challenged mice that received either p1108 inhibitor or vehicle control.

Effects of IC87114 on Cytokine protein levels Levels of IL-1(3, TNF-a, IL-4, IL-5, IL-13, and RANTES were quantified in the supematants of BAL fluids by enzyme immunoassays according to the manufacturer's protocol (IL-1(3, TNF-a, IL-4, and IL-5; Endogen, Inc., Wobum, MA; IL-13 and RANTES; R&D Systems, Inc., Minneapolis, MN). The lower detection limit for IL-10, TNFa, IL-4, IL-5, IL-13, and RANTES in these assays was 3, 10, 5, 5, 1.5, and 2 pg/ml, respectively.

OVA-challenge induced significant (p < 0.05) increases in the concentrations of all three IL-4, IL-5, IL- 13 cytokines detected by protein-blotting in lung tissues as well as by ELISA of BAL fluids compared with the levels detected after saline challenge. The increased levels of these cytokines in both lung tissue and in BAL fluids were significantly (p < 0.05) reduced by IC87114.

Enzyme immunoassays revealed that levels of IL-1(3 and TNFa in BAL fluids were also increased significantly (p < 0.05) at 72 h after OVA
inhalation compared with the levels after saline inhalation, from approximately 100 pg/ml TNFa and 15 pg/ml IL-1(3 in control animals up to approximately 280 pg/ml TNFa and pg/ml IL-1(3. IC87114 reduced the increased levels of these proinflammatory cytokines by more than 50%, down to approximately 140 pg/ml TNFa and 15 pg/ml IL-1(3. One of the responses to these cytokines is the induction of leukocyte-endothelial adhesion molecules. Indeed, levels of ICAM-1 and VCAM-1 proteins in the lung tissue were increased significantly (p < 0.05) at 72 h after OVA
inhalation and these levels were substantially reduced by the administration of IC87114.

Effects of IC87114 on eotaxin and RANTES protein levels in lung tissues and BAL
fluids of OVA sensitized and -challenged mice Western blot analysis revealed that protein levels of the chemokines eotaxin and RANTES in the lung tissue were increased significantly (p < 0.05) at 72 h after OVA inhalation compared with the saline control. Administration of reduced the increased levels of these chemokines by more than 50%. In addition, enzyme immunoassays revealed that increased levels of RANTES in BAL fluids at 10. h after, OVA inhalation were also significantly (p < 0.05) reduced by treatment. , Effect of IC87114 on serum IgE levels'and LTC4 release in BAL fluid IL-4 and IL-13 are important in directing B cell growth, differentiation, and secretion of IgE (Emson et 1., JExp Med 188:399-404, 1998). The biological activities of IgE are mediated through high affinity IgE receptors (FccRI) on mast cells and basophils. Cross-linking of FcsRI initiates multiple signaling cascades leading to cellular degranulation and activation (Nadler et al., Adv Im.rnunol 76:325-55, 2000: Kawakami et al., Nat Rev Ibnmunol 2:77386, 2002).

In order to determine whether IC87114 could modify OVA-specific Th2 response in vivo in mice, circulating IgE antibody levels were analyzed 72 hours following OVA challenge.

OVA-specific IgE levels were measured by capture ELISA as previously described (MacLean et al., Jlmmunol 165:6568-75, 2000). Briefly, microtiter plates were coated with 2 g/ml of purified monoclonal anti-mouse IgE
25, (BD PharMingen, San Diego, CA). After blocking with PBS-10% FCS, appropriate dilutions of serum samples in PBS-10% FCS were added to the plate and incubated for 2 h at room temperature. After washing with PBS-Tween, biotinylated OVA
(10 g/ml) and HRP-conjugated streptavidin were added to the wells and incubated for 1 hour., The plates were washed followed by addition of the HRP substrate, 3,3'5,5'-tetramethylbenzidine substrate (TMBS, Sigma Chemical Co.). After incubation for 30 min in the dark at room temperature, the plates were read at 450 nm on a microplate reader (Molecular Dynamics, Sunnyvale, CA). Total serum IgE was measured by capture ELISA in a manner similar to the detection of OVA-specific IgE. A biotinylated rat anti-mouse IgE (PharMingen) was used to detect captured IgE
in place of the biotinylated OVA.

Substantial elevation in total IgE and OVA-specific IgE was observed in serum from OVA-challenged mice (approx. 12 ng/ml total IgE, approx. 3 ng/ml OVA-Specific IgE) as compared with untreated mice (approx. 3 ng/ml total IgE, less than 1 ng/ml OVA-specific IgE). IC87114 significantly (p < 0.05) lowered total circulating IgE levels in,a dose-dependent manner (approx. 6ng/ml total IgE
and less than 1.5 ng/ml OVA-specific IgE). In agreement with the inhibitory effect on total IgE level, IC87114 at 0.1 and 1 mg/kg significantly (p < 0.05) reduced OVA-specific IgE levels by 63 and 72%, respectively.

BAL fluid was obtained 72 h after the last OVA challenge and assayed for LTC4. Levels of LTC4 were quantified in the, supematants of BAL fluids by enzyme,immunoassay according to the,manufacturer's protocol (Cayman Chemical Co., Ann Arbor,,Ml). The lower detection limit for LTC4 in this assay was 10 pg/ml.
The BAL fluid levels of the LTC4 were 3.1-fold higher in the OVA-sensitized/challenged mice (approximately 40 pg/ml) than in the mice receiving saline only (approximately 13 pg/ml) (p < 0.05 compared to saline). IC87114 (0.1 and mg/kg),significantly (p < 0.05) inhibited LTC4 levels by 37 and 50%, respectively decreasing LTC4 levels to approximately 26 pg/ml and 21 pg/ml, respectively.
The amounts of LTC4 in the BAL fluid of OVA-sensitized/challenged mice treated with vehicle control were not significantly different from those of the saline control group.

HYPERRESPONSIVENESS (AHR) The results above indicate that administration of p110S specific inhibitor effectively inhibited many side effects associated with airway inflammation.
To determine the effect of IC87114 on the development of AHR in an experimental animal model, OVA-treated mice were induced with AHR and treated with p1106 inhibitor.

Sensitized BALB/c mice challenged with 3% OVA aerosol for 30 min daily for 3 consecutive days developed AHR to inhaled methacholine. Three days after the last ovalbumin challenge, airway responsiveness was measured in unrestrained conscious animals using whole body plethysmography (All Medicus Co., Seoul, Korea), as described previously (Kwak et al., supra).

Briefly, in whole body plethysmography, respiratory function is measured in conscious, freely moving mice in chambers which allow animals to move freely within the chamber while respiratory function is measured. Each chamber is connected to a bias flow regulator to supply a smooth, constant flow of fresh air during testing. A transducer'attached to each chamber detects pressure changes that occur as the animal breathes. Inspiration and expiration are recorded by establishing start-inspiration and end-inspiration as the box pressure/time curve crosses the zero point. Start of an inspiration is determined by extrapolating from a straight line drawn from two levels of the rising inspiratory phase of the box pressure signal.
Time of inspiration (TI) is defined as, the time from the start of inspiration to the end of inspiration; time of expiration (TE) as the time from the end of inspiration to the start of the next inspiration. The maximum box pressure signal occurring during one breath in a negative or positive direction is defined as peak inspiratory pressure (PIP) or peak expiratory pressure (PEP), respectively. Recordings of every 10 breaths are extrapolated to define the respiratory rate in breaths per minute. The relaxation time (Tr) is defined as ihe time of pressure decay to 36% of the total expiratory pressure signal (area under the box pressure signal in expiration). This may thus serve as a correlate to the time constant (RC) of the decay of the volume signal to 36%
of the peak volume in passive expiration. During bronchoconstriction, the main alteration in the signal occurs during early expiration and leads to changes in the waveform of the box pressure signal. Other parameters measured include Tidal Volume (ml), Respiratory Rate (breaths per minutes), Minute Volume (tidal volume multiplied by respiratory rate, ml/min), Inspiratory Time (sec), Expiratory Time (sec), Peak Inspiratory Flow (ml/sec), and Peak Expiratory Flow (ml/sec).

Airway responsiveness was assessed as percent increase of Penh (Helmann et al., Anz JRespir Cr it Care Med 156:766-75, 1997; Chong et al., J
Pharrnacol Toxicol Methods 39:163-68, 1998). Readings were obtained at baseline and after exposure to aerosolized saline or methacholine (2.5 to 50 mg/ml).
Data were collected and averaged for 3 min after each nebulization. Enhanced pause (Penh), calculated as: (expiratory time/relaxation time-1) x (peak expiratory flow/peak inspiratory flow), according to the manufacturers' protocol. Results are expressed as the percentage increase of Penh following challenge with each concentration of 5 methacholine, where the baseline Penh (after saline challenge) is expressed as 100%.
Airway responsiveness was substantially increased in the OVA-challenged group in response to methacholine inhalation ascompared with the saline-challenged group (Figure 3). Administration of IC87114 to OVA-sensitized mice prior to OVA challenge showed a significant (p < 0.05) attenuation in Penh measured 10 at all methacholine levels tested suggesting a role for pl 108 in immune-mediated events leading to airway hyperresponsiveness in vivo. These results are contemplated to be associated with reduction in Th2 cytokine production, tissue eosinophilia, and mast cell activation, following p110S inhibition.

Allergic airway inflammation and AHR development involve multiple 15, inflammatory cells and a wide array of mediators. The results above demonstrate that p110S inhibition effectively reduced OVA-induced Th2 cytokine production, pulmonary eosinophilia, serum IgE levels, goblet cell liyperplasia, and AHR in a mouse asthma model. These findings indicate that p l 106 plays an important role in the pathogenesis of allergic asthma and that specific inhibitors of p110S are useful 20 ' therapeutics for the treatment of asthma and airway hyperresponsiveness.

DEGRANULATION
25 Mast cells and basophils express FcsRI, the high affinity receptor for IgE, and play a central role for IgE-associated immediate hypersensitivity reactions and allergic disorders. Cross-linking of FcsRI-bound IgE with multivalent antigen initiates the activation of mast cells and basophils, resulting in the degranulation from these cells. To determine the effects of p110S inhibitors on mast cell/basophil 30 degranulation, the release of serotonin by degranulation of rat basophil leukemia cells (RBL-2H3) was measured in the presence of p110S-specific inhibitors.

RBL-2H3 cells are grown in monolayer cultures in media. RBL cells were grown to confluence in 25 ml Eagle minimum essential medium containing 16% FCS

(EMEM-16) in a 75-cm2 tissue culture flask. Medium was removed via aspiration and cells washed with 3 ml trypsin-EDTA to remove trypsin inhibitor present in serum. Trypsin-EDTA was then added 37 C for 5 min to remove the adherent cells.
Cells are removed from the flask, washed in EMEM-16 and collected by centrifugation at 1000 rpm for 5 minutes. Cells were washed a second time and cell pellet resuspended in EMEM-16. Cells were then plated in a 24 well plate at a concentration of 4 x 105 cells/ml, and cultured with 25 gl of 1 mCi/ml 3H-labeled serotonin (0.5 Ci/ml final) and 1 g/ml anti-DNP IgE overnight at 37 C. Cell media was aspirated from the wells and cells washed twice by adding 500 l of PBS to the well and inverting the plate onto a stack of paper towels. A final volume of 200 [t1 PBS was added and cells equilibrate <_2 min in a 37 C water bath, and 10 l DNP-albumin was added to each well (10 ng/ml final) and incubated for the 10 to 30 min at 37 C.
The reaction was stopped by transferring the buffer from each well into a liquid scintillation vial. Wells were washed two times using 500 l of 1%
Triton X-100 in PBS incubated 10 min at room temperature, and the liquid transferred to the vial for measuring. 10 ml of liquid scintillation cocktail is added to the sample vials and the level of radioactivity counted in a liquid scintillation counter, calculating the percentage of serotonin released by the cells. In samples cultured with either p1108 inhibitor IC87114 or a second p110S-specific inhibitor was added over a range of doses to determine the efficacy of each dose.
Measurement of radioactivity levels in control and p1108-treated cells demonstrated that at IC87114 at a concentration of 2.5 micromolar showed 50%
inhibition of serotonin release.
These results indicate that p110S specific inhibitors are effective at reducing mast cell degranulation mediated by Fc-receptor cross-linking, providing a means for reducing these affects during allergic reactions.

RESPONSES
As shown above, p110S inhibitors were effective at reducing the levels of mast cell degranulation mediated by IgE crosslinking. Mast cell degranulation plays a significant role in mediating allergic reactions and other type I
hypersensitivity responses in vivo. To examine the effects of p1108 inhibitors on mast cell degranulation in vivo, an animal model of dermal hypersensitivity was used.
To sensitize, the shaved dorsal skin of Lewis rats was injected with either saline or anti-DNP monoclonal IgE (1.25 to 25 ng in 50 l per site) intradermally.
Forty-eight hours after IgE sensitization, 1 ml saline solution containing the antigen (1 ng/ml DNP-BSA) and 0.5% Evan's Blue dye was intravenously injected, and 30 min later the rats were euthanized. Skin edema was assessed by measuring the size of the wheal at the site of injection using a caliper. The skin around the wheal was also excised and the amounts of extravasated dye were measured as previously described 10, (Inagaki et al., 1986). A single dose of IC87114 (20 or 60 mg/kg) or PEG-400 (used as vehicle control) was administered orally to the rats 1 hour before antigen administration. Ketotifen (10 mg/kg) was intraperitoneally injected 30 minutes before a.ntigen challenge. Blood samples were drawn immediately after the wheal size measurements and plasma concentration of IC87114 was determined by determined , 15 liquid-liquid extraction by liquid chromatography/mass. spectroscopy as described previously (Puri et al., Blood,103:3448-3456, 2004).
Type I hypersensitivity responses showed a dose dependent response to IC87114, decreasing to approximately 70% of control at a dose of 20 mg/kg, to approximately 55% at a dose of 60 mg/kg. The positive control ketotifen (10 mg/kg) 20 decreased sensitivity responses to approximately 35% of the control response.
These results indicate that p1108 plays a role in mediating type I
hypersensitivity reactions in sensitized animals, indicating that administration of p1108 inhibitors may reduce or prevent type I sensitivity reactions DEGRANULATION
To determine the effect of p110S inhibitors on human mast cells, cells were isolated from human cord blood, differentiated to mast cell lineage and assayed 30 for degranulation and histamine release in the presence of p1108 inhibitors.

CD34+ human cord blood cells were isolated and differentiated using stem cell factor and IL-4 following the protocol set out in Hsieh et al. (JExp Med.
193:123-33, 2001), in the presence and absence of methylcellulose (lida et al, Blood 97:1016-22, 2001). Cells were cultured for approximately 5 weeks and were harvested by centrifugation at 1000 rpm for 3 minutes. Cells were resuspended in fresh medium (RPMI, IMDM or methylcellulose) at 1 x 106 cell/ml containing IgE
at g/ml. Cells were primed for 5-days before testing for activation by anti-IgE

5 cross-linking.

To assess mast cell activation by IgE cross-linking, cells were harvested by centrifugation 2 minutes at 1000 rpm, and washed twice in Dulbecco's-phosphate buffered saline (D-PBS). Cells were then resuspended at a concentration of 2.5 x 105 cells/ml in D-PBS. To 95 l of cells, 5 l of either 200 M
IC87114 or 10 ketoprofen in 3% DMSO, and cells were incubated at 37 C for 30 minutes.
After 30 minutes, 2~t1 of a 1:20 dilution of anti-IgE for a final concentration of 1:1000 IgE, and cells incubated in the presence of anti-IgE for 45 minutes. Cells were then centrifuged as above and the supernatant collected. An equivalent amount of D-PBS
was added to the cells and the cells were freeze-thawed 3-times to lyse the cells. The lysed cells were then centrifuged 4 minutes at 10,000 rpm and analyzed for histamine release by histamine ELISA.

Histamine was measured in both the cell supernatant and the cell pellet. In this experiment IC87114 p1105 inhibitor reduced histamine release by approximately 18 % at a 10 M concentration (based on normalization of total control release of 100%).

These results show that p1108 exhibits an effect on human mast cells as well as in murine mast cells, indicating that p110b inhibitors are useful regulators of histamine release and other mast cell activity in the treatment of patient with diseases or conditions mediated by aberrant mast cell activity.

Numerous modifications and variations in the invention as set forth in the above illustrative examples are expected to occur to those skilled in the art.
Consequently only such limitations as appear in the appended claims should be placed on the invention.

Claims (33)

1. A method for inhibiting an activity of mast cells comprising the step of administering to an individual a selective inhibitor of phosphoinositide 3-kinase delta (PI3K.delta.) in an amount effective to inhibit said activity.
2. The method of claim 1 wherein the mast cell activity being inhibited is mast cell migration.
3. The method of claim 1 wherein the mast cell activity being inhibited is mast cell degranulation.
4. The method of claim 1 wherein the mast cell activity being inhibited is mast cell proliferation.
5. The method of claim 1 wherein the mast cell activity being inhibited is secretion of a cytokine, chemokine or growth factor.
6. The method of claim 1 wherein the mast cell activity being inhibited is expression of a cytokine, chemokine or growth factor.
7. The method of claim 5 or 6 wherein the cytokine is TNF-.alpha..
8. The method of claim 5 or 6 wherein the cytokine is IL-6.
9. The method of claim 5 or 6 wherein the cheinokine is eotaxin, MIP-1.alpha., MIP-1 J3, MDC-1, MCP-1 or lymphotactin.
10. A method of reducing lymphocyte infiltration to a site of inflammation in a condition associated with undesirable mast cell activity comprising the step of administering to an individual a selective inhibitor of phosphoinositide 3-kinase delta (PI3K.delta.) in an amount effective to reduce or prevent lymphocyte infiltration to said site of inflammation in an amount effective to reduce lymphocyte recruitment signaling by mast cells in said individual.
11. A method for treating or preventing a condition associated with undesirable mast cell activity in an individual, comprising the step of administering a selective inhibitor of phosphoinositide 3-kinase delta (PI3K.delta.) in an amount effective for treating or preventing a condition associated with undesirable mast cell activity.
12. The method of claim 11 wherein the mast cell activity inhibited is mast cell migration, mast cell proliferation, mast cell degranulation, or expression or secretion of cytokines, chemokines or growth factors from mast cells.
13. The method of claim 11 wherein the condition is an IgE-mediated condition.
14. The method of claim 11 wherein the condition is asthma, an allergic reaction, or an autoimmune disease.
15. The method of claim 14 wherein the allergic reaction is type I
hypersensitivity, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, or allergic asthma.
16. The method of claim 14 wherein the autoimmune disease is bullous pemphigoid or chronic urticaria.
17. The method of any one of claims 11-16, wherein the PI3K.delta. selective inhibitor is administered in an amount effective to inhibit Akt phosphorylation in said mast cells.
18. The method of any one of claims 11-16, further comprising administering an immunomodulatory agent to the individual.
19. The method of claim 18, wherein the immunomodulatory agent is administered over a plurality of doses.
20. The method of claim 18, wherein the immunomodulatory agent is a glucocorticoid or corticosteroid, an immunosuppressant, an antihistamine, an aminosalicylate, a steroid hormone, a non-steroidal anti-inflammatory drug (NSAID), a sympathomimetic, or an analgesic.
21. The method of claim 18, wherein the immunosuppressant is an azathioprine, cyclosporine, cyclophosphamide, methotrexate, or penicillamine.
22. The method of claim 18, wherein the glucocorticoid is cortisone, dexamethosone, hydrocortisone, methylprednisolone, prednisolone, prednisone, or budesonide.
23. The method of any one of claims 11-16, wherein the PI3K6 selective inhibitor is administered over a plurality of doses.
24. A method of reducing or preventing mast cell proliferation in an individual having a condition associated with undesirable mast cell activity, comprising:

administering to said individual a therapeutically effective amount of a combination therapy comprising a selective inhibitor of phosphoinositide 3-kinase delta (PI3K6) and an immunomodulatory agent.
25. A method of reducing or preventing lymphocyte infiltration to a site of inflammation in an individual having a condition associated with undesirable mast cell activity, comprising:

administering to said individual a therapeutically effective amount of a combination therapy comprising a selective inhibitor of phosphoinositide 3-kinase delta (P13K6) and a immunomodulatory agent.
26. A method of increasing the therapeutic index of an immunomodulatory agent administered to an individual to treat a condition associated with undesirable mast cell activity, comprising:

administering to said individual a combination therapy comprising an immunomodulatory agent and an amount of a selective inhibitor of phosphoinositide 3-kinase delta (PI3K8) effective to increase the therapeutic index of the immunomodulatory agent.
27. The method of any one of claims 24-26 wherein the condition is asthma, an allergic reaction, or an autoimmune disease.
28. The method of any one of claims 24-26 wherein the allergic reaction is type I hypersensitivity, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, or allergic asthma.
29 The method of any one of claims 24-26 wherein the autoimmune disease is bullous pemphigoid, or chronic urticaria.
30. The method of any one of claims 1-16 or 24-26 wherein the selective PI3K8 inhibitor is a compound having formula (I) or pharmaceutically acceptable salts and solvates thereof:

wherein A is an optionally substituted monocyclic or bicyclic ring system containing at least two nitrogen atoms, and at least one ring of the system is aromatic;
X is selected from the group consisting of C(Rb)2,CH2CHRb, and CH=C(Rb);

Y is selected from the group consisting of null,S,SO,SO2,NH,0,C(=0), OC(=0), C(=O)O, and NHC(=0)CH2S;

Rl and R2, independently, are selected from the group consisting of hydrogen, C 1-6alkyl, aryl, heteroaryl, halo, NHC(=0)C 1-3 alkyleneN(Ra)2, NO2, ORa, CF3, OCF3, N(Ra)2, CN, OC(=O)Ra, C(=0)Ra, C(=O)ORa, arylORb, Het, NRaC(=O)C1-3alkyleneC(=0)ORa, arylOC1-3alkyleneN(Ra)2, arylOC(=0)Ra, C1-4alkyleneC(=0)ORa, OC1-4alkyleneC(=0)ORa, C1-4alkyleneOC1-4alkyleneC(=0)ORa, C(=O)NRaSO2Ra, C1-4alkyleneN(Ra)2, C2-6alkenyleneN(Ra)2, C(=O)NRaC1-4alkyleneORa, C(=O)NRaC1-4alkyleneHet, OC2-4alkyleneN(Ra)2, OC1-4alkyleneCH(ORb)CH2N(Ra)2, OC1-4alkyleneHet, OC2-4alkyleneORa, OC2-4alkyleneNRaC(=0)ORa, NRaC1-4alkyleneN(Ra)2, NRaC(=O)Ra, NRaC(=0)N(Ra)2, N(S02C1-4alkyl)2s NRa(S02C1-4alkyl), SO2N(Ra)2, OSO2CF3, C1-3alkylenearyl, C1-4alkyleneHet, C1-6alkyleneORb, C1-3alkyleneN(Ra)2, C(=O)N(Ra)2, NHC(=O)C1-3alkylenearyl, C3-8cycloalkyl, C3-8heterocycloalkyl, arylOC1-3alkyleneN(Ra)2, arylOC(=O)Rb, NHC(=0)C1-3 alkyleneC3-8heterocycloalkyl,NHC(=O)C 1-3 alkyleneHet, OC 1-4alkyleneOC l-4alkyleneC(=O)ORb, C(=O)C 1-4alkyleneHet, and NHC(=O)haloC1-6alkyl;

or Rl and R2 are taken together to form a 3- or 4-membered alkylene or alkenylene chain component of a 5- or 6-membered ring, optionally containing at least one heteroatom;

R3 is selected from the group consisting of optionally substituted hydrogen, C1-6alkyl, C3-8cycloalkyl, C3-8heterocycloalkyl, C1-4alkylenecycloalkyl, C2-6alkenyl, C1-3alkylenearyl; arylC1_3alkyl, C(=O)Ra, aryl, heteroaryl, C(=O)ORa, C(=O)N(Ra)2, C(=S)N(Ra)2, SO2Ra, SO2N(Ra)2, S(=O)Ra, S(=O)N(Ra)2, C(=O)NRaC 1-4alkyleneORa, C(=O)NRaC 1-4alkyleneHet, C(=O)C 1-4alkylenearyl, C(=O)C1-4alkyleneheteroaryl, C1-4alkylenearyl optionally substituted with one or more of halo, S02N(Ra)2, N(Ra)2, C(=O)ORa, NRaSO2CF3,CN,NO2, C(=O)Ra, ORa,C1-4alkyleneN(Ra)2, and OC1-4alkyleneN(Ra)2, C1-4alkyleneheteroaryl, C1-4alkyleneHet, C1-4alkyleneC(=O)C 1-4alkylenearyl, C1-4alkyleneC(=0)C 1-4alkyleneheteroaryl, C1-4alkyleneC(=O)Het, C1-4alkyleneC(=0)N(Ra)2, C1-4alkyleneORa, C1-4alkyleneNRaC(=O)Ra, C1-4alkyleneOC1-4alkyleneORa, C1-4alkyleneN(Ra)2, C1-4alkyleneC(=O)ORa, and C1-4alkyleneOC 1-4alkyleneC(=O)ORa;

Ra is selected from the group consisting of hydrogen, C1-6alkyl, C3-8cycloalkyl,C3-8heterocycloalkyl,C1-3alkyleneN(Rc)2, aryl, arylC1-3a1ky1, C1-3alkylenearyl, heteroaryl, heteroarylC1-3 alkyl, and C1-3 alkyleneheteroaryl;

or two Ra groups are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;

Rb is selected from the group consisting of hydrogen, C1-6alkyl, heteroCl_ 3alkyl, C1-3alkyleneheteroC1-3alkyl, arylheteroC1-3alkyl, aryl, heteroaryl, arylCl-3alkyl, heteroarylCl-3alkyl, C1-3alkylenearyl, and C1-3alkyleneheteroaryl;

Rc is selected from the group consisting of hydrogen, C1-6alkyl, C3-8cycloalkyl, aryl, and heteroaryl; and, Het is a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1-4alkyl or C(=O)ORa.
31. The method of any one of claims 1-16 or 24-26 wherein the selective PI3K8 inhibitor is selected from the group consisting of: 2-(6-aminopurin-9-ylmethyl)-3-(2-chlorophenyl)-6,7-dimethoxy-3H-quinazolin-4-one; 2-(6-aminopurin-o-ylmethyl)-6-bromo-3-(2-chlorophenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-o-ylmethyl)-3-(2-chlorophenyl)-7-fluoro-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-6-chloro-3-(2-chlorophenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-chlorophenyl)-5-fluoro-3H-quinazolin-4-one; 2-(6-aminopurin-o-yhnethyl)-5-chloro-3-(2-chloro-phenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-chlorophenyl)-5-methyl-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-8-chloro-3-(2-chlorophenyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-biphenyl-2-yl-5-chloro-3H-quinazolin-4-one; 5-chloro-2-(9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-chloro-3-(2-fluorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-chloro-3-(2-fluorophenyl)-3H-quinazolin-4-one; 3-biphenyl-2-yl-5-chloro-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 5-chloro-3-(2-methoxyphenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-fluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-6,7-dimethoxy-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 6-bromo-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-8-trifluoromethyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-one; 3-(2-chlorophenyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-benzo[g]quinazolin-one; 6-chloro-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 8-chloro-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-7-fluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-7-nitro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-one; 3-(2-chlorophenyl)-6-hydroxy-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 5-chloro-3-(2-chlorophenyl)-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-methyl-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-6,7-difluoro-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-6-fluoro-2-(9H-purin-6-yl-sulfanylmethyl)-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-isopropylphenyl)-5-methyl-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-one; 3-(2-fluorophenyl)-5-methyl-2-(9H-purin-6-yl-sulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-chloro-3-o-tolyl-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-5-chloro-3-(2-methoxy-phenyl)-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-3-cyclopropyl-5-methyl-3H-quinazolin-4-one;

3-cyclopropylmethyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclopropylmethyl-5-methyl-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-3-cyclopropylmethyl-5-methyl-3H-quinazolin-4-one; 5-methyl-3-phenethyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-5-methyl-3-phenethyl-3H-quinazolin-4-one; 3-cyclopentyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclopentyl-5-methyl-3H-quinazolin-4-one; 3-(2-chloropyridin-3-yl)-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-chloropyridin-3-yl)-5-methyl-3H-quinazolin-4-one; 3-methyl-4-[5-methyl-4-oxo-2-(9H-purin-6-ylsulfanylmethyl)-4H-quinazolin-3-yl]-benzoic acid; 3-cyclopropyl-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclopropyl-5-methyl-3H-quinazolin-4-one; 5-methyl-3-(4-nitrobenzyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 3-cyclohexyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-cyclohexyl-5-methyl-3H-quinazolin-4-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-3-cyclo-hexyl-5-methyl-3H-quinazolin-4-one; 5-methyl-3-(E-2-phenylcyclopropyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-fluoro-2-[(9H-purin-6-ylamino)methyl]-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)methyl]-3-(2-chlorophenyl)-5-fluoro-3H-quinazolin-4-one; 5-methyl-2-[(9H-purin-6-ylamino)methyl]-3-o-tolyl-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)methyl]-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-[(2-fluoro-9H-purin-6-ylamino)methyl]-5-methyl-3-o-tolyl-3H-quinazolin-4-one; (2-chlorophenyl)-dimethylamino-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one;

5-(2-benzyloxyethoxy)-3-(2-chlorophenyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 6-aminopurine-9-carboxylic acid 3-(2-chlorophenyl)-5-fluoro-oxo-3,4-dihydro-quinazolin-2-ylmethyl ester; N-[3-(2-chlorophenyl)-5-fluoro-4-oxo-3,4-dihydro-quinazolin-2-ylmethyl]-2-(9H-purin-6-ylsulfanyl)-acetamide; 2-[1-(2-fluoro-9H-purin-6-ylamino)ethyl]-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-[1-(9H-purin-6-ylamino)ethyl]-3-o-tolyl-3H-quinazolin-4-one; 2-(6-dimethylaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(2-methyl-6-oxo-1,6-dihydro-purin-7-ylmethyl)-3-o-tolyl-3H-quinazolin-4-one;

methyl-2-(2-methyl-6-oxo-1,6-dihydro-purin-9-ylmethyl)-3 -o-tolyl-3H-quinazolin-4-one; 2-(amino-dimethylaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-one; 2-(2-amino-9H-purin-6-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-one; 2-(4-amino-1,3,5-triazin-2-ylsulfanylmethyl)-5-methyl-3-o-tolyl.-3H-quinazolin-4-one; 5-methyl-2-(7-methyl-7H-purin-6-ylsulfanylmethyl)-3-o-to1y1-3H-quinazolin-4-one; 5-methyl-2-(2-oxo-1,2-dihydro-pyrimidin-4-ylsulfanylmethyl)-3-o-tolyl-quinazolin-4-one;. 5-methyl-2-purin-7-ylmethyl-3-o-tolyl-3H-quinazolin-4-one;

methyl-2-purin-9-ylmethyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(9-methyl-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 2-(2,6-Diamino-pyrimidin-4-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(5-methyl-[1,2,4]triazolo[1,5-a]pyrimidin-7-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(2-methylsulfanyl-9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 2-(2-hydroxy-9H-purin-6-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(1-methyl-lH-imidazol-2-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-3-o-tolyl-2-(1H-[1,2,4]triazol-3-ylsulfanylmethyl)-quinazolin-4-one; 2-(2-amino-6-chloro-purin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(6-aminopurin-7 ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-one; 2-(7-amino-1,2,3-triazolo[4,5-d]pyrimidin-3-yl-methyl)-5-methyl-3-o-tolyl-quinazolin-4-one; 2-(7-amino-1,2,3-triazolo[4, 5-d]pyrimidin-1-yl-methyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(6-amino-9H-purin-2-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(2-amino-6-ethylamino-pyrimidin-4-ylsulfanylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(3-amino-5-methylsulfanyl-1,2,4-triazol-1-yl-methyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(5-amino-3 -methylsulfanyl-1,2,4-triazol-1-ylmethyl)-5 -methyl-3 -o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(6-methylaminopurin-9-ylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 2-(6-benzylaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(2,6-diaminopurin-9-ylmethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3-o-tolyl-3H-quinazolin-4-one; 3-isobutyl-5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; N-{2-[5-Methyl-4-oxo-2-(9H-purin-6-ylsulfanylmethyl)-4H-quinazolin-3-yl]-phenyl}-acetamide; 5-methyl-3-(E-2-methyl-cyclohexyl)-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-[5-methyl-4-oxo-2-(9H-purin-6-ylsulfanylmethyl)-4H-quinazolin-3-yl]-benzoic acid; 3-{2-[(2-dimethylaminoethyl)methylamino]phenyl} -5-methyl-2-(9H-purin-6-ylsulfanylmethyl)-3H-quin-azolin-4-one; 3-(2-chlorophenyl)-5-methoxy-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 3-(2-chlorophenyl)-5-(2-morpholin-4-yl-ethylamino)-2-(9H-purin-6-ylsulfanylmethyl)-3H- quinazolin-4-one; 3-benzyl-5-methoxy-2-(9H-purin-6-ylsulfanylmethyl)-3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-benzyloxyphenyl)-5-methyl-3H-quinazolin-4-one; 2-(6-aminopurin-ylmethyl)-3-(2-hydroxyphenyl)-5-methyl-3H-quinazolin-4-one; 2-(1-(2-amino-9H-purin-6-ylamino)ethyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 5-methyl-2-[1-(9H-purin-6-ylamino)propyl]-3-o-tolyl-3H-quinazolin-4-one; 2-(1-(2-fluoro-9H-purin-ylamino)propyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(1-(2-amino-9H-purin-ylamino)propyl)-5-methyl-3-o-tolyl-3H-quinazolin-4-one; 2-(2-benzyloxy-l-(9H-purin-6-ylamino)ethyl)-5-methyl-3-o-to1y1-3H-quinazolin-4-one; 2-(6-aminopurin-ylmethyl)-5-methyl-3- {2-(2-(1-methylpyrrolidin-2-yl)-ethoxy)-phenyl} -3H-quinazolin-4-one; 2-(6-aminopurin-9-ylmethyl)-3-(2-(3-dimethylamino-propoxy)-phenyl)-5-methyl-3H-quinazolin-4-one; 2-(6-aminopurin-9.-ylmethyl)-5-methyl-3-(2-prop-2-ynyloxyphenyl)-3H-quinazolin-4-one; 2-{2-(1-(6-aminopurin-9-ylmethyl)-5-methyl=4-oxo-4H-quinazolin-3-y1]-phenoxy}-acetamide; 5-chloro-3-(3,5-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[1-(9H-purin-ylamino)-propyl]-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 6-fluoro-3-phenyl-2-[1-(9H-purin-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[ 1-(9H-purin-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(2,3-difluoro-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3-chloro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)-methyl]-3-(3,5-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-{2-[(2]-diethylamino-ethyl)-methyl-amino]-phenyl} -5 -methyl-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-one; 5-chloro-3-(2-fluoro-phenyl)-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 5-chloro-2-[(9H-purin-6-ylamino)-methyl]-3-o-tolyl-3H-quinazolin-4-one; 5-chloro-3-(2-chloro-phenyl)-2-[(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one;
6-fluoro-3-(3-fluoro-phenyl)-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;
2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-chloro-3-(3-fluoro-phenyl)-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-2-[1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[1-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H
quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-propyl}-3H-quinazolin-4-one; 2-[1-(2-fluoro-9h-purin-6-ylamino)-propyl]-5-methyl-3-phenyl-3h-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-methyl-3-phenyl-3H-quinazolin-4-one; 2-[2-benzyloxy-l-(9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-2-benzyloxy-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[2-benzyloxy-l-(7H-pyrrolo [2,3-d]pyrimidin-4-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one;
2-[2-benzyloxy-l-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H
quinazolin-4-one; 3-(4-fluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(4-fluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(4-fluoro-phenyl)-2-[1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-5-methyl-3H-quinazolin-4-one;3-(4-fluoro-phenyl)-5-methyl-2-[

(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-( 7Hpyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one;

(3-fluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;
2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-fluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3-fluoro-phenyl)-5-methyl-2-[1-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-yl)-pyrrolidin-2-yl]-3H-quinazolin-4-one; 2-[2-hydroxy-l-(9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[phenyl-(9H-purin-6-ylamino)-methyl]-3H-quinazolin-4-one; 2-[(2-amino-9H-purin-6-ylamino)-phenyl-methyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[(2-fluoro-9H-purin-6-ylamino)-phenyl-methyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[phenyl-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-methyl]-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-fluoro-3-phenyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5 -chloro-3-phenyl-3H-quinazolin-4-one; [5-(5-methyl-4-oxo-3-phenyl-3,4-dihydro-quinazolin-2-yl)-5-(9H purin-6-ylamino)-pentyl}-carbamic acid benzyl ester; [5-(2-amino-9H-purin-6-ylamino)-5-(5-methyl-4-oxo-3-phenyl-3,4-dihydro-quinazolin-2-yl)-pentyl}-carbamic acid benzyl ester;
[4-(5-methyl-4-oxo-3-phenyl-3,4-dihydro-quinazolin-2-yl)-4-(9H-purin-6-ylamino)-butyl]-carbamic acid benzyl ester; [4-(2-amino-9H-purin-6-ylamino)-4-(5-methyl-4-oxo-phenyl-3,4-dihydro-quinazolin-2-yl)-butyl]-carbamic acid benzyl ester; 3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[5=amino-l-(9H-purin-6-ylamino)-pentyl]-5-methyl-3-phenyl-3H quinazolin-4-one); 2-[5-amino-l-(2-amino-9H-purin-6-ylamino)-pentyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[ 1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-Dimethyl-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(2,6-dimethyl-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-morpholin-4-ylmethyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-morpholin-4ylmethyl-3-phenyl-3H-quinazolin-4-one; 2-[4-amino-l-(2-amino-9H-purin-6-ylamino)-butyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-6-fluoro-3-phenyl-3H-quinazolin-4-one; 2-[2-tert-butoxy-l-(9H-purin-6-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 3-(3-methyl-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-methyl-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3-chloro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl}-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-chloro-phenyl)-5-methyl-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-2-hydroxy-ethyl]-5-methyl-phenyl-3Fl-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-fluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-yla.mino)-ethyl]-3-(2,6-difluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-5-fluoro-3-phenyl-3H-quinazolin-4-one; 5-chloro-3-(3-fluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-chloro-3-(3-fluoro-phenyl)-3H-quinazolin-4-one; 3-phenyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-5-trifluoromethyl-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-methyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[ 1-(2-amino=9H-purin-6-ylamino)-propyl]-3-(2,6-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3,5-dichloro-phenyl)-5-methyl-2-[1-(9H-purin-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(2,6-dichloro-phenyl)-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(2,6-dichloro-phenyl)-5-methyl-3H-quinazolin-4-one; 5-chloro-3-phenyl-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[l-(2-amino-9H-purin-6-ylamino)-propyl]-5-chloro-3-phenyl-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-ylamino)-butyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-butyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-ylamino)-ethyl]-3-(3,5-dichloro-phenyl)-5-methyl-3H-quinazolin-4-one; 5-methyl-(3-morpholin-4-ylmethyl-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3-Hquinazolin-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-3-(3-morpholin-4-ylmethyl-phenyl)-3H-quinazolin-4-one; 2-[1-(5-bromo-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-5-methyl-3-pheny1-3H-quinazolin-4-one; 5-methyl-2-[1-(5-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3-phenyl-3H-quinazolin-4-one; 2-[1-(5-fluoro-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 2-[2-hydroxy-l-(9H-purin-6-ylamino)-ethyl]-3-phenyl-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-propyl]-3Hquinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-3-(3,5-difluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(5-bromo-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl]-3-(3-fluoro-phenyl)-5-methyl-3H-quinazolin-4-one; 3-(3-fluoro-phenyl)-5-methyl-2-[1-(5-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-ethyl}-3H-quinazolin-4-one; 3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3,5-difluoro-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-3-phenyl-3H-quinazolin-4-one; 6,7-difluoro-3-phenyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 6-fluoro-3-(3-fluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[4-diethylamino-l-(9H-purin-6-ylamino)-butyl]-5-methyl-3-phenyl-3H-quinazolin-4-one; 5-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-one; 5-fluoro-2-[ 1-(2-fluoro-9H-purin-6-ylamino)-ethyl]-3-phenyl-3H-quinazolin-4-one; 3-(3-fluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;

chloro-3-(3,5-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-one; 3-(2,6-difluoro-phenyl)-5-methyl-2-[1-(9H=purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(2,6-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-Methyl-3-phenyl-2-[3,3,3-trifluoro-l-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 3-(3-hydroxy-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-(3-methoxy-phenyl) -5-methyl-2-{1-[9H-purin-6-ylamino]-ethyl} -3H-quinazolin-4-one; 3-[3-(2-dimethylamino-ethoxy)-phenyl]-5-methyl-2-{1-[9H-purin-6-ylamino]-ethyl}-3H-quinazolin-4-one; 3-(3-cyclopropylmethoxy-phenyl)-5-methyl-2- {1-[9H=purin-6-ylamino]-ethyl}-3H-quinazolin-4-one; 5-methyl-3-(3-prop-2-ynyloxy-phenyl)- 2- { 1-[9H-purin-6-ylamino]-ethyl} -3H-quinazolin-4-one; 2- { 1-[2-amino-9H-purin-6-ylamino]
ethyl} -3 -(3-hydroxyphenyl)-5-methyl-3H-quinazolin-4-one; 2- {1-[2-amino-9H-purin-6-ylamino]ethyl} -3-(3-methoxyphenyl)-5-methyl-3H-quinazolin-4-one; 2- {1-[2-amino-9H-purin-6-ylamino]ethyl} -3-(3-cyclopropylmethoxy-phenyl)-5-methyl-3H-quinazolin-4-one; 2-{1-[2-amino-9H-purin-6-ylamino] ethyl} -5-methyl-3-(3-prop-ynyloxy-phenyl)-3H-quinazolin-4-one; 3-(3-ethynyl-phenyl)-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 3-{5-methyl-4-oxo-2-[1-(9H-purin-ylamino)-ethyl]-4H-quinazolin-3-y1}-benzonitrile; 3-{5-methyl-4-oxo-2-{1-[9H-purin-6-ylamino)-ethyl]-4H-quinazolin-3-yl}-benzamide; 3-(3-acetyl-phenyl)-5-methyl-2-{1-[9H-purin-6-ylamino]-ethyl}-3H-quinazolin-4-one; 2-(3-(5-methyl-4-oxo-2-{1-[9H-purin-6-ylamino]-ethyl},-4H-quinazolin-3-yl-phenoxy acetamide; 5-methyl-2-{1-[9H-purin-6-ylamino]-ethyl}-3-[3-(tetrahydropuran-4-yloxy)-phenyl]-3H-quinazolin-4-one; 3-[3-(2-methoxy-ethoxy)-phenyl]-5-methyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;6-fluoro-2-[1-(9H-purin-6-ylamino)ethyl]-3-[3-(tetrahydro-pyran-4-yloxy)-phenyl]-3H-quinazolin-4-one; 3-[3-(3-dimethylamino-propoxy)-phenyl]-5-methyl-2-[ 1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one;
2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-(3-ethynyl-phenyl)-5-methyl-3H-quinazolin-4-one; 3-{2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-4-oxo-quinazolin-3-yl}-benzonitrile; 3-{2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-4-oxo-4H-quinazolin-3-yl}-benzamide; 3-{2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-4-oxo-4H-quinazolin-3-yl}-benzamide; 5-methyl-3-(3-morpholin-4-yl-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-5-methyl-3-(3-morpholin-4-yl-phenyl)-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl]-3-[3-(2-methoxy-ethoxy)-phenyl]-5-methyl-3H-quinazolin-4-one; 2-[1-(2-amino-9.H-purin-6-ylamino)-ethyl]-3-[3-(2-dimethylamino-ethoxy)-phenyl]-5-methyl-3H quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-but-3-ynyl]-5-methyl-3-phenyl-3H-quinazolin-4-one;
2-[1-(2-amino-9H-purin-6-ylamino)-but-3-ynyl]-5-methyl-3-phenyl-3H-quinazolin-one; 5-chloro-3-(3,5-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-5-chloro-3-(3;5-difluoro-phenyl)-3H-quinazolin-4-orie; 2-[1-(2-amino-9H-purin-6-ylamino)-ethyl}-5-chloro-3-(3,5-difluoro-phenyl)-3H-quinazolin-4-one; 3-(3,5-difluoro-phenyl)-6-fluoro-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one; 5-chloro-3-(2,6-difluoro-phenyl)-2-[1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one; 2-[1-(2-amino-9H-purin-6-ylamino)-propyl]-5-chloro-3 -(2,6-difluoro-phenyl)-3H-quinazolin-4-one; 5-methyl-3-phenyl-2-[1-(9H-purin-6-yloxy)-ethyl]-3H-quinazolin-4-one:

and, pharmaceutically acceptable salts and solvates thereof.
32. An article of manufacture comprising a selective inhibitor of phosphoinositide 3-kinase delta (PI3K5) and a label indicating a method of use according to any one of claims 1-31.
33. Use of a composition comprising at least one selective inhibitor of PI3K8 in the manufacture of a medicament for treating or preventing a disorder involving undesirable mast cell activity.
CA002569406A 2004-06-04 2005-06-04 Methods for treating mast cell disorders Abandoned CA2569406A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US57694704P 2004-06-04 2004-06-04
US60/576,947 2004-06-04
PCT/US2005/019558 WO2005120511A1 (en) 2004-06-04 2005-06-04 Methods for treating mast cell disorders

Publications (1)

Publication Number Publication Date
CA2569406A1 true CA2569406A1 (en) 2005-12-22

Family

ID=34971708

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002569406A Abandoned CA2569406A1 (en) 2004-06-04 2005-06-04 Methods for treating mast cell disorders

Country Status (5)

Country Link
EP (1) EP1750715A1 (en)
JP (1) JP2008501707A (en)
CN (1) CN101123968A (en)
CA (1) CA2569406A1 (en)
WO (1) WO2005120511A1 (en)

Families Citing this family (102)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
DK1737831T3 (en) 2004-04-02 2013-08-19 Prana Biotechnology Ltd Neurologically active compounds
DK1761540T3 (en) 2004-05-13 2016-11-21 Icos Corp Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase DELTA
JP2008520744A (en) 2004-11-19 2008-06-19 ザ・レジェンツ・オブ・ザ・ユニバーシティ・オブ・カリフォルニア Anti-inflammatory pyrazolopyrimidine
WO2006089106A2 (en) * 2005-02-17 2006-08-24 Icos Corporation Phosphoinositide 3-kinase inhibitors for inhibiting leukocyte accumulation
GB2431156A (en) * 2005-10-11 2007-04-18 Piramed Ltd 1-cyclyl-3-substituted- -benzenes and -azines as inhibitors of phosphatidylinositol 3-kinase
JP2009514954A (en) 2005-11-04 2009-04-09 ハイドラ バイオサイエンシズ インコーポレイテッド Compounds for modulating TRPV3 function
CA2647391C (en) 2006-04-04 2015-12-29 The Regents Of The University Of California Kinase antagonists
NZ576278A (en) 2006-10-19 2011-12-22 Signal Pharm Llc Heteroaryl compounds, compositions thereof, and their use as protein kinase inhibitors
WO2008140750A1 (en) 2007-05-10 2008-11-20 Hydra Biosciences Inc. Compounds for modulating trpv3 function
TWI434849B (en) 2007-06-29 2014-04-21 Gilead Sciences Inc Modulators of toll-like receptor 7
WO2009046448A1 (en) 2007-10-04 2009-04-09 Intellikine, Inc. Chemical entities and therapeutic uses thereof
US20090131512A1 (en) * 2007-10-31 2009-05-21 Dynavax Technologies Corp. Inhibition of type I in IFN production
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US8703777B2 (en) 2008-01-04 2014-04-22 Intellikine Llc Certain chemical entities, compositions and methods
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
WO2009142772A2 (en) 2008-05-23 2009-11-26 Mastcell Pharmaceuticals, Inc. Methods and treatment for allergies and inflammation associated with gastrointestinal diseases
WO2010006086A2 (en) 2008-07-08 2010-01-14 Intellikine, Inc. Kinase inhibitors and methods of use
WO2010006072A2 (en) 2008-07-08 2010-01-14 The Regents Of The University Of California Mtor modulators and uses thereof
CA2738429C (en) 2008-09-26 2016-10-25 Intellikine, Inc. Heterocyclic kinase inhibitors
EP2358720B1 (en) 2008-10-16 2016-03-02 The Regents of The University of California Fused ring heteroaryl kinase inhibitors
US8110578B2 (en) 2008-10-27 2012-02-07 Signal Pharmaceuticals, Llc Pyrazino[2,3-b]pyrazine mTOR kinase inhibitors for oncology indications and diseases associated with the mTOR/PI3K/Akt pathway
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
PT2355828T (en) * 2008-11-13 2018-07-02 Gilead Calistoga Llc Therapies for hematologic malignancies
US9492449B2 (en) 2008-11-13 2016-11-15 Gilead Calistoga Llc Therapies for hematologic malignancies
KR101787309B1 (en) 2008-12-09 2017-10-18 길리애드 사이언시즈, 인코포레이티드 Modulators of toll-like receptors
BRPI1012333A2 (en) 2009-03-24 2016-03-29 Gilead Calistoga Llc atropisomers of 2-purinyl-3-tolyl-quinazolinones derivatives and methods of use
ES2548253T3 (en) * 2009-04-20 2015-10-15 Gilead Calistoga Llc Methods for the treatment of solid tumors
JP5789252B2 (en) 2009-05-07 2015-10-07 インテリカイン, エルエルシー Heterocyclic compounds and uses thereof
WO2011011550A1 (en) 2009-07-21 2011-01-27 Calistoga Pharmaceuticals Inc. Treatment of liver disorders with pi3k inhibitors
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
GB0918249D0 (en) 2009-10-19 2009-12-02 Respivert Ltd Compounds
NZ598933A (en) 2009-10-22 2013-04-26 Gilead Sciences Inc Derivatives of purine or deazapurine useful for the treatment of (inter alia) viral infections
EP3091021B1 (en) 2009-10-26 2019-08-28 Signal Pharmaceuticals, LLC Methods of synthesis and purification of heteroaryl compounds
JP5951600B2 (en) 2010-05-21 2016-07-13 インフィニティー ファーマシューティカルズ, インコーポレイテッド Compounds, compositions and methods for kinase regulation
UY33337A (en) 2010-10-18 2011-10-31 Respivert Ltd SUBSTITUTED DERIVATIVES OF 1H-PIRAZOL [3,4-d] PYRIMIDINE AS INHIBITORS OF PHOSFOINOSITIDE 3-KINASES
CA2817577A1 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
TWI592411B (en) 2011-02-23 2017-07-21 英特爾立秦有限責任公司 Combination of kinase inhibitors and uses thereof
AU2012284088B2 (en) 2011-07-19 2015-10-08 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
AR088218A1 (en) 2011-07-19 2014-05-21 Infinity Pharmaceuticals Inc USEFUL HETEROCICLICAL COMPOUNDS AS PI3K INHIBITORS
EP2751093A1 (en) 2011-08-29 2014-07-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
JP6342805B2 (en) 2011-09-02 2018-06-13 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Substituted pyrazolo [3,4-D] pyrimidine and uses thereof
TWI708605B (en) 2011-10-19 2020-11-01 標誌製藥公司 Treatment of cancer with tor kinase inhibitors
EA026390B1 (en) 2011-12-02 2017-04-28 СИГНАЛ ФАРМАСЬЮТИКАЛЗ, ЭлЭлСи PHARMACEUTICAL COMPOSITIONS OF 7-(6-(2-HYDROXYPROPAN-2-YL)PYRIDIN-3-YL)-1-((TRANS)-4-METHOXYCYCLOHEXYL)-3,4-DIHYDROPYRAZINO[2,3-b]PYRAZIN-2(1H)-ONE, SOLID FORMS THEREOF AND METHODS OF THEIR USE
EP2817029B1 (en) 2012-02-24 2019-07-10 Signal Pharmaceuticals, LLC Methods for treating non-small cell lung cancer using tor kinase inhibitor combination therapy
KR20140133590A (en) 2012-03-05 2014-11-19 길리아드 칼리스토가 엘엘씨 Polymorphic forms of (s)-2-(1-(9h-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3h)-one
NZ628072A (en) 2012-03-13 2015-08-28 Respivert Ltd Crystalline pi3 kinase inhibitors
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
SG10202003090VA (en) * 2012-08-08 2020-05-28 Kbp Biosciences Co Ltd PI3K δ INHIBITOR
BR112015006828A8 (en) 2012-09-26 2019-09-17 Univ California compound, or a pharmaceutically acceptable salt thereof; pharmaceutical composition; use of the compound; and method for modulating the activity of an ire1 protein
AU2013203714B2 (en) 2012-10-18 2015-12-03 Signal Pharmaceuticals, Llc Inhibition of phosphorylation of PRAS40, GSK3-beta or P70S6K1 as a marker for TOR kinase inhibitory activity
US9346812B2 (en) 2013-01-16 2016-05-24 Signal Pharmaceuticals, Llc Substituted pyrrolopyrimidine compounds, compositions thereof, and methods of treatment therewith
WO2014112646A1 (en) * 2013-01-21 2014-07-24 国立大学法人大阪大学 Phenoxyalkylamine compound
US9227977B2 (en) 2013-03-15 2016-01-05 Respivert Ltd. Phosphoinositide 3-kinase inhibitors
JO3279B1 (en) 2013-03-15 2018-09-16 Respivert Ltd 2-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-1h-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3-(2-(trifluoromethyl)benzyl)quinazolin-4(3h)-one derivatives and their use as phosphoinositide 3-kinase inhibitors
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
CA2908957C (en) 2013-04-17 2021-05-18 Signal Pharmaceuticals, Llc Combination therapy comprising a dihydropyrazino-pyrazine compound and an androgen receptor antagonist for treating prostate cancer
NZ629332A (en) 2013-04-17 2017-05-26 Signal Pharm Llc Treatment of cancer with dihydropyrazino-pyrazines
EP2986321A1 (en) 2013-04-17 2016-02-24 Signal Pharmaceuticals, LLC Combination therapy comprising a tor kinase inhibitor and a cytidine analog for treating cancer
MX2015014455A (en) 2013-04-17 2016-07-21 Signal Pharm Llc Combination therapy comprising a tor kinase inhibitor and n-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-yl amino)phenyl)acrylamide for treating cancer.
CA2908830C (en) 2013-04-17 2021-12-07 Signal Pharmaceuticals, Llc Treatment of cancer with dihydropyrazino-pyrazines
NZ629230A (en) 2013-04-17 2017-05-26 Signal Pharm Llc Pharmaceutical formulations, processes, solid forms and methods of use relating to 1-ethyl-7-(2-methyl-6-(1h-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1h)-one
AU2014253978B2 (en) 2013-04-17 2019-06-06 Signal Pharmaceuticals, Llc Combination therapy comprising a TOR kinase inhibitor and a 5-Substituted Quinazolinone Compound for treating cancer
JP6401250B2 (en) 2013-05-29 2018-10-10 シグナル ファーマシューティカルズ,エルエルシー 7- (6- (2-hydroxypropan-2-yl) pyridin-3-yl) -1-((trans) -4-methoxycyclohexyl) -3,4-dihydropyrazino [2,3-b] pyrazine-2 (1H) -one, pharmaceutical composition thereof in solid form, and method of use thereof
UY35675A (en) 2013-07-24 2015-02-27 Novartis Ag SUBSTITUTED DERIVATIVES OF QUINAZOLIN-4-ONA
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
PE20160685A1 (en) 2013-10-04 2016-07-23 Infinity Pharmaceuticals Inc HETEROCYCLIC COMPOUNDS AND USES OF THEM
AU2014364414A1 (en) 2013-12-20 2016-06-30 Gilead Calistoga Llc Polymorphic forms of a hydrochloride salt of (S) -2-(1-(9H-purin-6-ylamino) propyl) -5-fluoro-3-phenylquinazolin-4 (3H) -one
NZ736970A (en) 2013-12-20 2018-11-30 Gilead Calistoga Llc Process methods for phosphatidylinositol 3-kinase inhibitors
SG10201808053XA (en) 2014-03-19 2018-10-30 Infinity Pharmaceuticals Inc Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
NZ714742A (en) 2014-04-16 2017-04-28 Signal Pharm Llc Solid forms of 1-ethyl-7-(2-methyl-6-(1h-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1h)-one, compositions thereof and methods of their use
WO2015160882A1 (en) 2014-04-16 2015-10-22 Signal Pharmaceuticals, Llc SOLID FORMS COMPRISING 7-(6-(2-HYDROXYPROPAN-2YL) PYRIDIN-3-YL)-1-(TRANS)-4-METHOXYCYCLOHEXYL)-3, 4-DIHYDROPYRAZINO[2,3-b] PYRAZIN-2(1H)-ONE, AND A COFORMER, COMPOSITIONS AND METHODS OF USE THEREOF
US9737535B2 (en) 2014-04-16 2017-08-22 Signal Pharmaceuticals, Llc Methods for treating cancer using TOR kinase inhibitor combination therapy comprising administering substituted pyrazino[2,3-b]pyrazines
WO2015160975A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2015160880A1 (en) 2014-04-16 2015-10-22 Signal Pharmaceuticals, Llc SOLID FORMS COMPRISING 1-ETHYL-7-(2-METHYL-6-(1H-1,2,4-TRIAZOL-3-YL) PYRIDIN-3-YL)-3,4-DIHYDROPYRAZINO(2,3-b)PYRAZIN-2(1H)-ONE, AND A COFORMER, COMPOSITIONS AND METHODS OF USE THEREOF
US20170189409A1 (en) * 2014-05-27 2017-07-06 Almirall, S.A. Medical use
CN106459005A (en) 2014-06-13 2017-02-22 吉利德科学公司 Phosphatidylinositol 3-kinase inhibitors
WO2016001855A1 (en) 2014-07-04 2016-01-07 Lupin Limited Quinolizinone derivatives as pi3k inhibitors
US11116774B2 (en) 2014-07-11 2021-09-14 Gilead Sciences, Inc. Modulators of toll-like receptors for the treatment of HIV
US9623028B2 (en) 2014-07-14 2017-04-18 Signal Pharmaceuticals, Llc Methods of treating a cancer using substituted pyrrolopyrimidine compounds, compositions thereof
NZ629796A (en) 2014-07-14 2015-12-24 Signal Pharm Llc Amorphous form of 4-((4-(cyclopentyloxy)-5-(2-methylbenzo[d]oxazol-6-yl)-7h-pyrrolo[2,3-d]pyrimidin-2-yl)amino)-3-methoxy-n-methylbenzamide, compositions thereof and methods of their use
EA201790369A1 (en) 2014-09-16 2017-10-31 Джилид Сайэнс, Инк. SOLID FORMS OF THOUGH-RECEPTOR MODULATOR
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9957267B2 (en) 2015-07-01 2018-05-01 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
WO2017048702A1 (en) 2015-09-14 2017-03-23 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinone derivatives, process of making, compositions comprising, and methods of using the same
RU2750151C2 (en) 2015-11-20 2021-06-22 Форма Терапьютикс, Инк. Hypoxantines as inhibitors of ubiquitin-specific protease 1
WO2017161116A1 (en) 2016-03-17 2017-09-21 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as pi3k kinase inhibitors
WO2017214269A1 (en) 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
CN109640999A (en) 2016-06-24 2019-04-16 无限药品股份有限公司 Combination treatment
KR101932146B1 (en) * 2016-07-14 2018-12-24 주식회사 바이오웨이 Novel Quinazolinone derivatives as PI3K inhibitors, and pharmaceutical composition comprising the same
AU2017299513A1 (en) * 2016-07-19 2019-02-28 Arete Discoveries, Inc. Biomarkers for detection and treatment of mast cell activity-associated disorders
TW201813963A (en) 2016-09-23 2018-04-16 美商基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
TW201825465A (en) 2016-09-23 2018-07-16 美商基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
TW201815787A (en) 2016-09-23 2018-05-01 美商基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
BR112019027402A2 (en) 2017-06-22 2020-07-07 Celgene Corporation treatment of hepatocellular carcinoma characterized by infection with the hepatitis b virus
WO2019023278A1 (en) 2017-07-25 2019-01-31 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
CA3168103A1 (en) 2020-01-29 2021-08-05 Kamari Pharma Ltd. Compounds and compositions for use in treating skin disorders
WO2022256628A1 (en) * 2021-06-04 2022-12-08 La Jolla Institute For Immunology Suppressive t cell populations and methods of cancer immunotherapy
KR20230065591A (en) * 2021-11-05 2023-05-12 연세대학교 산학협력단 A Composition for Preventing or Treating Atopic Dermatitis Comprising an Inhibitor of AKT Signaling Pathway as an Active Ingredient

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI2223922T1 (en) * 2000-04-25 2016-04-29 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
US6667300B2 (en) * 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta

Also Published As

Publication number Publication date
WO2005120511A1 (en) 2005-12-22
CN101123968A (en) 2008-02-13
JP2008501707A (en) 2008-01-24
EP1750715A1 (en) 2007-02-14

Similar Documents

Publication Publication Date Title
CA2569406A1 (en) Methods for treating mast cell disorders
US20050054614A1 (en) Methods of inhibiting leukocyte accumulation
US20080287469A1 (en) Phosphoinositide 3-Kinase Inhibitors for Inhibiting Leukocyte Accumulation
US20050043239A1 (en) Methods of inhibiting immune responses stimulated by an endogenous factor
US20060106038A1 (en) Methods for treating and/or preventing aberrant proliferation of hematopoietic cells
US20060079538A1 (en) Methods for inhibiting angiogenesis
Piwowarski et al. Urolithins, gut microbiota‐derived metabolites of ellagitannins, inhibit LPS‐induced inflammation in RAW 264.7 murine macrophages
McNamara et al. Small-molecule inhibitors of the PI3K signaling network
US20050239809A1 (en) Methods for treating and preventing hypertension and hypertension-related disorders
JP6047149B2 (en) Combined pharmaceutical composition and use thereof
US8962633B2 (en) Methods of treatment and prevention of metabolic bone diseases and disorders
JP2004504259A (en) Method of inhibiting C-JUN expression using JAK-3 inhibitor
JP2002538107A (en) JAK-3 inhibitor for treating allergic disorders
JP2008501776A (en) Inhibition of phosphodiesterase 10 as a treatment for conditions associated with obesity and associated with metabolic syndrome
KR20060110301A (en) Cdk2 antagonists as short form c-maf transcription factor antagonists for treatment of glaucoma
CN110652514A (en) Pharmaceutical use of third generation EGFR inhibitor
US20040127575A1 (en) Method for counteracting a pathologic change in the beta-adrenergic pathway
Fortin et al. Effects of antisense oligodeoxynucleotides targeting CCR3 on the airway response to antigen in rats
US6800649B1 (en) Method for inhibiting c-jun expression using JAK-3 inhibitors
CN111093661A (en) Use of glutarimide derivatives for the treatment of diseases associated with abnormal activity of cytokines
JP2009542817A (en) New compounds
US11351156B2 (en) Combination treatment of pancreatic cancer
EA038527B1 (en) Method of treating a respiratory disease or condition, compositions and kits comprising a dual pi3k delta-gamma kinase inhibitor and a corticosteroid
US20160120872A1 (en) RAC1 Inhibitors for Inducing Bronchodilation

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead