Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PHOSPHATIDYLINOSITOL 3-KINASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2015/191754
Kind Code:
A2
Abstract:
The present application provides the compounds of formula (J), or pharmaceutically acceptable salts, isomers, tautomer, or a mixture thereof wherein n, m, R1, R2, R3, R4, and R5 are described herein. The compounds are inhibitors to the activities of phosphatidylinositol 3-kinase (ΡI3Κ) and are useful for treating conditions mediated by one or more PI3K isoforms. The present application further provides pharmaceutical compositions that include a compound of formula (I), or pharmaceutically acceptable salts, isomers, tautomer, or mixture thereof, and methods of using these compounds and compositions for treating conditions mediated by one or more ΡI3Κ isoforms.

Inventors:
KIM MUSONG (US)
YEUNG SUET CHUNG (US)
PERREAULT STEPHANE (US)
Application Number:
PCT/US2015/035161
Publication Date:
December 17, 2015
Filing Date:
June 10, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GILEAD SCIENCES INC (US)
International Classes:
C07D403/14
Foreign References:
US6800620B22004-10-05
US8435988B22013-05-07
US8673906B22014-03-18
US20130274253A12013-10-17
US8486941B22013-07-16
US8450321B22013-05-28
US8557803B22013-10-15
US4965288A1990-10-23
US4997854A1991-03-05
US4943593A1990-07-24
US5021456A1991-06-04
US5059714A1991-10-22
US5120764A1992-06-09
US5182297A1993-01-26
US5252608A1993-10-12
US20040248871A12004-12-09
US201261745437P2012-12-21
US3845770A1974-11-05
US4326525A1982-04-27
US4902514A1990-02-20
US5616345A1997-04-01
US5023252A1991-06-11
US4992445A1991-02-12
US5001139A1991-03-19
Other References:
RAMEH ET AL., J. BIOL. CHEM., vol. 274, 1999, pages 8347 - 8350
PANAYOTOU ET AL., TRENDS CELL BIOL., vol. 2, 1992, pages 358 - 60
OTSU ET AL., CELL, vol. 65, 1991, pages 91 - 104
HILES ET AL., CELL, vol. 70, 1992, pages 419 - 29
ENGELMAN, NAT. REV. CANCER, vol. 9, 2009, pages 550 - 562
FOSTER: "Deuterium Isotope Effects in Studies of Drug Metabolism", TRENDS PHARMACOL. SCI., vol. 5, no. 12, 1984, pages 524 - 527, XP025943358, DOI: doi:10.1016/0165-6147(84)90534-0
BERGE, JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, no. 1, January 1977 (1977-01-01)
J. MARCH: "Advanced Organic Chemistry, 4th ed.", 1992, JOHN WILEY AND SONS
AGNEW, CHEM. INTL. ED. ENGI, vol. 33, 1994, pages 183 - 186
FERRARA N.; ALITALO, K: "Clinical application of angiogenic growth factors and their inhibitors", NATURE MEDICINE, vol. 5, 1999, pages 1359 - 1364, XP002217702, DOI: doi:10.1038/70928
CHESON, B.D.; LEONARD, J.P.: "Monoclonal Antibody Therapy for B-Cell Non-Hodgkin's Lymphoma", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 359, no. 6, 2008, pages 613 - 626, XP002573821, DOI: doi:10.1056/NEJMra0708875
WIERDA, W.G.: "Current and Investigational Therapies for Patients with CLL", HEMATOLOGY, 2006, pages 285 - 294, XP055419284, DOI: doi:10.1182/asheducation-2006.1.285
MORTON, L.M. ET AL.: "Lymphoma Incidence Patterns by WHO Subtype in the United States, 1992-2001", BLOOD, vol. 107, no. 1, 2006, pages 265 - 276, XP055419287, DOI: doi:10.1182/blood-2005-06-2508
"Remington's Pharmaceutical Sciences. 17th Ed.", 1985, MACE PUBLISHING CO.
G.S. BANKER & C.T. RHODES,: "Modem Pharmaceutics. 3rd Ed.", MARCEL DEKKER, INC
Attorney, Agent or Firm:
HARTRUM, J. Elin et al. (Inc.333 Lakeside Driv, Foster City CA, US)
Download PDF:
Claims:
What is claimed:

1. A compound having the structure of formula (I):

wherein:

n is 1 , 2, or 3;

m is O or 1;

each R1 is independently selected from halo, cyano, optionally substituted alkyl, optionally substituted Ci-6 haloalkyl, optionally substituted Cj^ alkoxy, optionally substituted sulfonyl, optionally substituted C3.8 aryl, optionally substituted C3.8 heteroaryl, optionally substituted C3-8 cycloalkyl, and optionally substituted Cj.g heterocycloalkyl;

each R2 is independently selected from halo and optionally substituted C|-6 alkyl;

R3 is hydrogen, optionally substituted alkyl, optionally substituted C$.io aryl, or optionally substituted C3-8 cycloalkyl;

R4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms, wherein the heteroatoms are selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted C5- alkyl, and -NH2; and

R5 is hydrogen or optionally substituted alkyl, wherein R5 and R3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring; or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

2. The compound of claim 1 , wherein

n is 1 or 2;

m is 0 or 1 ;

each R5 is independently selected from halo, alkyl, and haloalkyl

each R2 is independently selected from Cj^ alkyl;

R3 is hydrogen, C\.6 alkyl, or C3.8 cycloalkyl;

63 R4 is a six- to twelve-membered heteroar l having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, -NH2, Ci^ haloalkyl, and Ci alkyi; and

R5 is hydrogen, methyl, ethyl, or propyl, or Rs and R3 together with the atoms to which they are attached optionally form a five-membered heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

3. The compound of claims 1 -2, wherein each R1 is independently selected from chloro, bromo, fluoro, methyl, ethyl, and propyl.

4. The compound of any of claims 1 -3, wherein each R: is independently selected from methyl, ethyl, and propyl.

5. The compound of any of claims 1 -4, wherein R3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, or cyclobutyl.

6. The compound of any of claims 1-5, wherein R5 is hydrogen, methyl, ethyl, or propyl.

7. The compound of any of claims 1 -6, R5 and R3 together with the atoms to which they are attached optionally form pyrrolidinyl.

8. The compound of any of claims 1-7, wherein R4 is a monocyclic heteroaryl having at least two nitrogen atoms, wherein R4 is substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and -NH2.

9. The compound of any of claims 1 -8, wherein R4 is pyrimidinyl substituted with two or three members selected from the group consisting of bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and -NH2.

10. The compounds of any of claims 1-19, wherein the compound is an (S)-enantiomer.

11. The compounds of any of claims 1-19, wherein the compound is an (R)-enantiomer.

12. The compound of claim 1 , wherein the compound is selected from the group consisting of:

(S)-3-(5-ammopyrazm-2-yl)-5-cM^

8-fluoroquinazolin-4(3H)-one;

64 (S)-2,4^iamino-6^(l-(3-(5-aminop>Tazin^

2-yi)ethyi)amino)pyriinidm^

(S)-4-amino-6-((l-(3-(5-aminopyrazin-2-yl)-5-chloro-8-fluoro-4-oxo-3,4-di yl)d- yl)amko)pyrimidine-5-cai1x>nitriie;

(S>2^1 -((3H-[1 ,2 3triazolol4,5^3pyrimidin-7-yl)am«»

fluoroquinazolin-4(3H)-one;

(S)-3^5-amijiop imii 2-y^^

ylamino)ethyl)quina2olin-4(3H)-one;

(S 2,4Kliammo^((H3-(5-aniinopyra2M^

yl)ethyl)amino)pyrimidine-5-caii)oiiitrile;

(S)-3-(5-aminopyrazin-2-yl)-5-chloK>-2-( 1 -((2,6-diamino-5-cWoropyrimidin-4- yl)amino)ethyl)quinazoHn-4(3H)-one;

(S)-3-<5-aiiiinopyrazin-2-yl)-5-chloix)-2-(cyclopTOpyi((2,6-diaram

yl)amino)methyl)quina2olin-4(3H)-one;

(S)-2^^iamino-6-(((3-(5-aminopyra2dn^

ylXcyclc^ropyl)methyl)ammo^

(SV2 4- Uamino-6-(((3-(5-aminopyrazm^

ylXcyclopropyl)methyl)an^

(S)-2,4^axnino~6 (K3 5-ami^

yl)ethyl)aniino)pyrimidine-5-caibonitrile;

(S)-3-(5-aminc yrazw-2-yl 5,8^icM

yl)amino)methyl)quinazolin-4(3H)-one;

(S)-3-(5-aminopyrazin-2-yt>-5,8-di ¾loro-2-( 1 ^(2,6^amino-5^1<m>pyrimidin-4- yl)amino)elhyl)q\jinazolin-4(3H> >ne;

(S)-2-amino-4-((l-(3-(5-aminc^yra¾n^

yl)ethyl)amkoHKdifiuofomethyl)pyriim

65 (S>2,4-Diamino^-( H3^5-amitwpyr^

yl)e hyl)amino)pyriiiudine-5-cai^nitri

(S)-3-(5-Aminopyrazm-2-yl 2^

di{luoroquinazolin-4(3H)-one;

(S)-2,4-Dian3diK>^^(l-(3^5-aininop razm

dihydroquina.∞Un-2-yl)e^

(S)-3 5-Aminopyrazm-2-yiy8 i^^^

6-fluoroquinazolm-4(3H)-one;

(S)-2-(l-((6-Amkc 5-cWoropyrirai<lin-4-yl)ami

chloroquinazolin-4{3H)-one;

(S)-2-(((6-Amino-5-cMow^vri^

5,8 lichloroquinazolijri-4(3H)-one; and

(Sy2-(((6-Amino-5-cWoropyrimidin^yi^

5-chJoroqwnazoUn-4(3H)-one;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

13. A pharmaceutical composition comprises the compound of any of claims 1 - 12 and at least one pharmaceutically acceptable vehicle.

14. A method of treating a disease or condition in a human in need thereof comprises the administering to the human a therapeutically effective amount of the compound of any of claims 1 -12, wherein the disease or condition is selected from cancer, hematologic malignancies, leukemias, lymphomas, myeloproliferative disorders, myelodysplastic syndromes, plasma cell neoplasms, solid tumor, inflammation, fibrosis, autoimmune disorders, allergic conditions, hypersensitivity, cardiovascular diseases, neurodegenerative diseases, renal disorders, viral infections, obesity, and autoimmune diseases.

15. The method of claim 14, wherein the disease or condition is selected from rheumatoid arthritis, osteoarthritis, atherosclerosis, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, asthma, chronic obstructive airways disease,

pneumonitis, dermatitis, alopecia, nephritis, vasculitis, atherosclerosis, Alzheimer's disease, hepatitis, primary biliary cirrhosis, sclerosing cholangitis, diabetes (including type I diabetes), acute rejection of transplanted organs, lymphomas, multiple myelomas, leukemias, pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, rectum cancer, liver cancer,

66 kidney cancer, stomach cancer, skin cancer, gastric cancer, esophageal cancer, head and neck cancer, melanoma, neuroendocrine cancers, CNS cancers (e.g., neuroblastoma), brain tumors (e.g., glioma, anaplastic oligodendroglioma, adult glioblastoma multiforme, and adult anaplastic astrocytoma), bone cancer, or soft tissue sarcoma. A method of inhibiting the activity of a phosphatidylinositol 3-kinase polypeptide by contacting the polypeptide with the compound of any of claims 1-12.

16. A method of inhibiting excessive or destructive immune reactions or growth or a proliferation of cancer cells, comprising administering an effective amount of the compound of any of claims 1-12.

17. A kit comprises the compound of any of claims 1 -12 or a label and or instructions for use.

18. The compound, a pharmaceutically acceptable salt, isomer, or a mixture thereof of any of claims 1-12 for use in therapy.

19. The compound, a pharmaceutically acceptable salt, isomer, or a mixture thereof of any of claims 1-12 for use in a method of treating of any of claims 14-15.

20. Use of the compound, a pharmaceutically acceptable salt, isomer, or a mixture thereof of any of claims 1-12 for the manufacture of a medicament for treatment of a disease or condition of any of claims 14-15.

67

Description:
PHOSPHATIDYL! INOSITOL 3 KINASE INHIBITORS

FIELD

[0001 ) The present application relates to novel compounds that selectively inhibit the activities of P13K isoforms and their uses in therapeutic treatments.

BACKGROUND

10002] Cell signaling via 3'-phosphorylated phosphoinositides has been implicated in a variety of cellular processes, e.g., malignant transformation, growth factor signaling,

inflammation, and immunity (Rameh et al.,J. Biol. Che , 274:8347-8350, 1999).

Phosphatidylinositoi 3 -kinase (PI 3 -kinase or PI3K) is responsible for generating these phosphorylated signaling products. PI3K was initially identified as a protein associated with viral oncoproteins and growth factor receptor tyrosine kinases that phosphory!ate

phosphatidylinositoi (PI) and its phosphorylated derivatives at the 3'-hydroxyl of the inositol ring(Panayotou era/., Trends Cell Biol, 2:358-60, 1992).

(0003] Three classes of the PI 3-kinase (PI3K) are proposed based on the substrate specificities. Class I PI3Ks phosphory!ate phosphatidylinositoi (PI), phosphandylinositol-4- phosphate, and phosphatidylinositol-4,5-biphosphate (PIP2) to produce phosphatidylinositol-3- phosphate (PIP), phosphatidylinositol-S^biphosphate, and phosphatidylinositol-3,4,5- triphosphate, respectively. Also, Class II PI3Ks phosphorylate PI and phosphatidylinositol-4- phosphate, and Class HI PI3Ks phosphorylate PI.

[0004] The initial purification and molecular cloning of PI 3-kinase revealed that it was a heterodimer consisting of p85 and pi 10 subunits (Otsu et al., Cell, 65:91-104, 1991; Hiles et al.. Cell, 70:419-29, 1992). Lata, four distinct Class I PI3Ks were identified and designated as PI3K α, β, δ, and γ isoforms. Each isoform consists of a distinct 110 kDa catalytic subunit and a regulatory subunit The catalytic subunits of PI3K a, β, and δ (i.e., pi 1 Oct, pi 10β, and pi 105, respectively) interacts, individually, with the same regulatory subunit p85, whereas the catalytic subunit of PI3K γ (pi 10γ) interacts with a distinct regulatory subunit plOI .

{0005} Studies have also showed that each PI3K isoform has distinct expression pattern. For example, PIK3CA which encodes PI3Kcc is frequently mutated in human cancers (Enge!man, Nat. Rev. Cancer, 9: 550-562, 2009). Also, PI3K8 is generally expressed in hematopoietic cells. Moreover, PI3K isoforms are shown to be associated with proliferation or survival signaling in cancers, inflammatory, or autoimmune diseases. As each PDK isoform has different biological function, P K isoforms are potential targets to treat cancer or disorder (US Patent Nos.

6,800,620; 8,435,988; 8,673,906; US Patent Application Publication No. US2013/0274253).

(0006) Therefore, there is a need for developing therapeutic agents that inhibit PDK isoforms to treat diseases, disorders, or conditions that are mediated by PDK.

SUMMARY

{0007} The present application provides novel compounds that are inhibitors of PI3K isoforms. The application also provides compositions, including pharmaceutical compositions, kits that include the compounds, and methods of using and making the compounds. The compounds provided herein are useful in treating diseases, disorders, or conditions that are mediated by PDK isoforms. The application also provides compounds for use in therapy. The application further provides compounds for use in a method of treating a disease, disorder, or condition that is mediated by PDK isoforms. Moreover, the application provides uses of the compounds in the manufacture of a medicament for the treatment of a disease, disorder or condition that is mediated by PDK isoforms.

(0008) The applications provides the compounds having the structure of formula (I):

wherein:

n is 1, 2, or 3;

m is O or 1;

each R l is independently selected from halo, cyano, optionally substituted alkyl, optionally substituted Cj haloalkyl, optionally substituted Ci^ alkoxy, optionally substituted suifbnyl, optionally substituted C3-8 aryl, optionally substituted C$4 heteroaryl, optionally substituted C 3 - 8 cycloalkyl, and optionally substituted C34 heterocycloalkyi;

each R 2 is independently selected from halo and optionally substituted C14 alkyl;

R 3 is hydrogen, optionally substituted C|. 6 alkyl, optionally substituted O aryl, or optionally substituted C3-8 cycloalkyl; R 4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two heteroatoms, wherein the heteroatoms are selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted Ct-6 alkyl, and - H2; and

R 5 is hydrogen or optionally substituted CM alkyl, wherein R s and R 3 together with the atoms to which they are attached optionally form a four- or dght-membered heterocyclic ring; or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

10009) The application also provides the compound having the structure of formula (I), wherein:

n is 1 or 2;

m is O or 1;

each R 1 is independently selected from halo, Ci^ alkyi, and C T _ haloalkyl;

each R 2 is independently Cj^ alkyl;

R 3 is hydrogen, Cj* alkyl, or C34 cycloalkyl;

R 4 is a six- to twelve-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, -NH2, haloalkyl, and Ci* alkyl; and

R 5 is hydrogen, methyl, ethyl, or propyl, or R 3 and R J together with the atoms to which they are attached optionally form a five-membered heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

10010] In one aspect, the compound of the present application having the structure of formula (I), wherein each R l is independently selected from chloro, bromo, fluoro, methyl, ethyl, and propyl, in some aspect, the compound of the present application having the structure of formula (I), wherein each R 2 is independently selected from methyl, ethyl, and propyl. In certain aspect, the compound of the present application having the structure of formula (I), wherein R 3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, or cyclobutyl. In other aspect, the compound of the present application having the structure of formula (1), wherein R 5 is hydrogen, methyl, ethyl, or propyl. In some other aspect, the compound of the present application having the structure of formula (I), wherein R 5 and R 3 together with the atoms to which they are attached optionally form pyrrolidinyl. In further aspect, the compound of the present application having the structure of formula (I), wherein R 4 is a monocyclic heteroaryl having at least two nitrogen atoms, wherein R 4 is substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and -NH . In some further aspect, the compound of the present application having the structure of formula (I), wherein R 4 is pyrimidinyl substituted with two or three members selected from the group consisting of bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and -Nl-fe.

10011] In certain embodiments, the PI3 inhibitors are the compounds selected from Table 1 , a pharmaceutically acceptable salt, isomer, or a mixture thereof. In additional embodiments, the compound is an (S)-enantiomer. In other embodiments, the compound is an (R)-enantiomer. In other additional embodiments, the compound is an atropisomer.

[0012] The application also provides a pharmaceutical composition that comprises a compound of formula (I), a pharmaceutically acceptable salt, isomer, or a mixture thereof, together with at least one pharmaceutically acceptable vehicle. Examples of a pharmaceutically acceptable vehicle may be selected from carriers, adjuvants, and excipients.

(0013) Further provided herein is a method of treating a disease, disorder, or condition in a human in need thereof by administering to the human a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof. Further provided is a compound of formula (I) for use in a method of treating a disease, disorder or condition that is mediated by PI3K isoforms. The application also provides the use of a compound of formula (I) in the manufacture of a medicament for the treatment of a disease, disorder or condition that is mediated by PI3K isoforms. In certain embodiments, the disease, disorder, or condition is associated or mediated by PI3 . In some embodiments, the disease, disorder, or condition is an inflammatory disorder. In other embodiments, the disease, disorder, or condition is a cancer.

(0014] Also provided herein is a method of inhibiting the activity of a phosphatidylinositol 3-kinase polypeptide by contacting the polypeptide with a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

(0015] Further provided is a method of inhibiting excessive or destructive immune reactions, comprising admiriistering an effective amount of a compound of formula (I) or a

pharmaceutically acceptable salt, isomer, or a mixture thereof. [0016| Also provided is a method of inhibiting growth or proliferation of cancer cells comprising contacting the cancer cells with an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

[0017| Also provided is a kit that includes a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof. The kit may further comprise a label and/or instructions for use of the compound in treating a disease, disorder, or condition in a human in need thereof. In some embodiments, the disease, disorder, or condition may be associated or mediated by PI3 activity.

[0018] Also provided are articles of manufacture that include a compound of formula (I) or a pharmaceutically acceptable salt, isomer, or a mixture thereof, and a container. In one embodiment, the container may be a vial, jar, ampoule, preloaded syringe, or an intravenous bag.

DETAILED DESCRIPTION

[0019] The following description sets forth exemplary methods, parameters and the like. Such description is not intended as a limitation on the scope of the present application but is instead provided as exemplary mbodiments.

[0020] As used herein, the following words, phrases and symbols are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise.

{00211 A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -CONHj is attached through the carbon atom. A dash at the front or end of a chemical group is a matter of convenience; chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning. A wavy line drawn through a line in a structure indicates a point of attachment of a group. Unless chemically or structurally required, no directionality is indicated or implied by the order in which a chemical group is written or named.

[0022] The prefix "C u v " indicates that the following group has from u to v carbon atoms. For example, "C h alky." indicates that the alkyl group has from 1 to 6 carbon atoms.

[0023) Reference to "about" a value or parameter herein includes (and describes)

embodiments that are directed to that value or parameter per se. In certain embodiments, the term "about" includes the indicated amount ± 10%. In other embodiments, the term "about" includes the indicated amount ± 5%. In certain other embodiments, the term "about" includes the indicated amount ± 1%. Also, to the term "about X" includes description of "X". Also, the singular forms "a" and "the" include plural references unless the context clearly dictates otherwise. Thus, e.g., reference to "the compound" includes a plurality of such compounds and reference to "the assay" includes reference to one or more assays and equivalents thereof known to those skilled in the art

(0024| "Alkyl " ' refers to an unbranched or branched saturated hydrocarbon chain. As used herein, alkyl has 1 to 20 carbon atoms (i.e., Cj.20 alkyl), 1 to 8 carbon atoms (i.e., C|.g alkyl), 1 to 6 carbon atoms (/.(?., C t ^ alkyl), or 1 to 4 carbon atoms (i.e., C alkyl). Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, pentyl, 2-pentyi, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyi, and 3-methylpenty!. When an alkyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons may be encompassed; thus, for example, "butyl" includes n-butyl, sec-butyl, isobutyi and t-butyl; "propyl' * includes n-propyl and isopropyl.

{0025] "Alkenyl" refers to an aliphatic group containing at least one carbon-carbon double bond and having from 2 to 20 carbon atoms (i.e., C2.20 alkenyl), 2 to 8 carbon atoms (i.e., C 2 -g alkenyl), 2 to 6 carbon atoms (i.e., C2- alkenyl), or 2 to 4 carbon atoms (i.e., C2-4 alkenyl). Examples of alkenyl groups include ethenyl, propenyl, butadienyl (including 1,2- butadienyl and 1,3-butadienyl).

(0026J "Alkynyr refers to an aliphatic group containing at least one carbon-carbon triple bond and having from 2 to 20 carbon atoms (i.e., C2.20 alkynyl), 2 to 8 carbon atoms {i.e., C2- 8 alkynyl), 2 to 6 carbon atoms (i.e., C2- 6 alkynyl), or 2 to 4 carbon atoms (i.e., C2. alkynyl). The term "alkynyl" also includes those groups having one triple bond and one double bond.

[0027] "Alkoxy" refers to the group "alkyl-O-". Examples of alkoxy groups include methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n- hexoxy, and 1,2-dimethylbutoxy.

(00281 "AcyP refers to a group -C(=0)R, wherein R is hydrogen, alkyl, cycloalkyl, heterocycloalky], aryl, heteroalkyl, or heteroaryl; each of which may be optionally substituted, as defined herein. Examples of acy include formyl, acetyl, cylcohexylcarbonyl,

cyclohexylmethyl-carbonyl, and benzoyl. 10029J "Amido" refers to both a "C-amido" group which refers to the group -C(«0)NR y R z and an "N-amido'* group which refers to the group -NR y C(= )R z , wherein R y and R z are independently selected from the group consisting of hydrogen, alkyl, aryl, haloalkyl, or heteroaryi; each of which ma be optionally substituted. f0030J "Amino" refers to the group -NR y R 2 wherein R y and R z are independently selected from the group consisting of hydrogen, alkyl, haloalkyl, aryl, or heteroaryi; each of which may be optionally substituted.

(0031 j "Aryl" refers to an aromatic carbocyclic group having a single ring (e.g. monocyclic) or multiple rings (e.g. bicyclic or tricyclic) including fused systems. As used herein, aryl has 6 to 20 ring carbon atoms (i.e., C$.20 *"Ύθ» 6 to 12 carbon ring atoms (i.e., C - 1 2 aryl), or 6 to 10 carbon ring atoms (i.e., C^io aryl). Examples of aryl groups include phenyl, naphthyl, fluorenyl, and anthry!. Aryl, however, does not encompass or overlap in any way with heteroaryi defined below. If one or more aryl groups are fused with a heteroaryi ring, the resulting ring system is heteroaryi.

10032] "Cyano' * or "carbonitrile" refers to the group -CN.

[0033] "CycloaJkyP * refers to a saturated or partially saturated cyclic alkyl group having a single ring or multiple rings including fused, bridged, and spiro ring systems. The term

"cycloalkyl" includes cycloalkenyl groups (i.e. the cyclic group having at least one alkenyl). As used herein, cycloalkyl has from 3 to 20 ring carbon atoms (i.e., C3.20 cycloalkyl), 3 to 12 ring carbon atoms (i.e.,€3.12 cycloalkyl), 3 to 10 ring carbon atoms (i.e., C3.10 cycloalkyl), 3 to 8 ring carbon atoms (i.e., C3-8 cycloalkyl), or 3 to 6 ring carbon atoms (i.e., Cj^ cycloalkyl). Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyi, and cyclohexyl.

{0034] "Halogen" or "halo" includes fiuoro. chloro, bromo, and iodo. "Haloalkyl" refers to an unbranched or branched alkyl group as defined above, wherein one or more hydrogen atoms are replaced by a halogen. For example, where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached. Dihaloalkyl and trihaloalkyl refer to alkyl substituted with two ("di") or three ("tri") halo groups, which may be, but are not necessarily, the same halogen. Examples of haloalkyl include difluoromethyl (-CHF2) and trifluoromethyl (-CF3). 10035} "Heteroalkyl" refers to an alkyt group in which one or more of the carbon atoms (and any associated hydrogen atoms) are each independently replaced with the same or different heteroatomic group. By way of example, 1 , 2 or 3 carbon atoms may be independently replaced with the same or different heteroatomic group. Heteroatomic groups include, but are not limited to, -NR-, -0-, -S-, -S(OK -S(0) , and the like, where R is H, alkyl, aryl, cycloalkyi,

heteroalkyl, heteroaryl or heterocycloalkyl, each of which may be optionally substituted.

Examples of heteroalkyl groups include -OCH3, -CH 2 OCH 3l -SCHj, -CH 2 SCH 3 , -NRCHj, and - CH2NRCH3, where R is hydrogen, alkyl, aryl, arylalkyl, heteroalkyl, or heteroaryl, each of which may be optionally substituted. As used herein, heteroalkyl include 1 to 10 carbon atoms, 1 to S carbon atoms, or 1 to 4 carbon atoms; and 1 to 3 heteroatoms, 1 to 2 heteroatoms, or 1 heteroatom.

[0036) * 'HeteroaryF' refers to an aromatic group having a single ring, multiple rings, or multiple fused rings, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur. As used herein, heteroaryl include 1 to 20 ring carbon atoms (i.e., C| -2 o heteroaryl).3 to 12 ring carbon atoms (i.e.,€3.12 heteroaryl), or 3 to 8 carbon ring atoms (i.e., C3-8 heteroaryl): and 1 to 5 heteroatoms, 1 to 4 heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups include pyrimidinyl, purinyf, pyridyl, pyridazinyl,

benzothiazolyi, and pyrazolyl. Heteroaryl does not encompass or overlap with aryl as defined above.

(0037] "Heterocycloalky refers to a saturated or unsaturated cyclic alkyl group, with one or more ring heteroatoms independently selected from nitrogen, oxygen and sulfur. The term "heterocycloalkyl" includes heterocycloalkenyl groups (i.e. the heterocycloalkyl group having at least one alkenyl). A heterocycloalkyl may be a single ring or multiple rings wherein the multiple rings may be fused, bridged, or spiro. As used herein, heterocycloalkyl has 2 to 20 ring carbon atoms (i.e.. C 2 .2o heterocycloalkyl), 2 to 12 ring carbon atoms (i.e., C 2 .| 2

heterocycloalkyl), 2 to 10 ring carbon atoms (i.e., C2-10 heterocycloalkyl), 2 to 8 ring carbon atoms (i.e., C2-8 heterocycloalkyl), 3 to 12 ring carbon atoms (i.e., C3-12 heterocycloalkyl), 3 to 8 ring carbon atoms (i.e., heterocycloalkyl), or 3 to 6 ring carbon atoms (i.e., C34

heterocycloalkyl); having 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring

heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, sulfur or oxygen. Examples of beterocycloalkyl groups include pvrrolidinyl, piperidinyl, piperazinyl, oxetanyl, dioxolanyl, azetidinyl, and morpholinyl.

[0038J "Hydroxy" or "hydroxy!" refers to the group -OH.

[0039J ^©" refers to the group (=0) or (O).

100401 "Sulfonyl" refers to the group -S(0>2 , where R is alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, heteroaryl, or aryl. Examples of sulfonyl are methylsulfonyl, ethylsulfonyl, phenylsulf nyl, and toluenesulfonyl.

(0041] Certain commonly used alternative chemical names may be used. For example, a divalent group such as a divalent "alkyl" group, a divalent "aryl" group, etc., may also be referred to as an "alkylene" group or an "alkylenyl" group, an "arylene" group or an "arylenyl" group, respectively. Also, unless indicated explicitly otherwise, where combinations of groups are referred to herein as one moiety, e.g. arylalkyl, the last mentioned group contains the atom by which the moiety is attached to the rest of the molecule.

{0042} The terms "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. Also, the term "optionally substituted" refers to any one or more hydrogen atoms on the designated atom or group may or may not be replaced by a moiety other than hydrogen.

(0043) The term "substituted" means that any one or more hydrogen atoms on the designated atom or group is replaced with one or more substituents other than hydrogen, provided that the designated atom ' s normal valence is not exceeded. The one or more substituents include, but are not limited to, alkyl, alkenyi, alkynyl, alkoxy, acyl, amino, amido, amidino, aryl, azido, carbamoyl, carboxyl, carboxyl ester, cyano, guanidino, halo, haloalkyt, heteroatkyl, heteroaryl, heterocycloalkyl, hydroxy, hydrazino, imino, oxo, nitro, alkylsulfinyl, sulfonic acid, alkylsulfonyl, thiocyanate, thiol, thione, or combinations thereof. By way of example, there may be one, two, three, four, five, or six substituents. Polymers or similar indefinite structures arrived at by defining substituents with further substituents appended ad infinitum (e.g., a substituted aryl having a substituted alkyl which is itself substituted with a substituted aryl group, which is further substituted by a substituted heteroalkyl group, etc.) are not intended for inclusion herein. Unless otherwise noted, the maximum number of serial substitutions in compounds described herein is three. For example, serial substitutions of substituted aryl groups with two other substituted aryl groups are limited to substituted aryl (substituted aryl) substituted aryl. Similarly, the above definitions are not intended to include impermissible substitution patterns (e.g., methyl substituted with 5 fluorines or heteroaryl groups having two adjacent oxygen ring atoms). Such impermissible substitution patterns are well known to the skilled artisan. When used to modify a chemical group, the term

•'substituted" may describe other chemical groups defined herein. For example, the terra

''substituted aryp includes, but is not limited to, ''alkylaryl.' * Unless specified otherwise, where a group is described as optionally substituted, any substituents of the group are themselves unsubstituted.

[0044] In some embodiments, the term "substituted alkyl" refers to an alkyl group having one or more substituents including hydroxy!, halo, alkoxy, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl. In additional embodiments, "substituted cycloalkyF' refers to a cycloalkyl group having one or more substituents including alkyl, haloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, halo, hydroxy!; "substituted aryl" refers to an aryl group having one or more substituents including halo, alkyl, haloalkyl, heterocycloalkyl, heteroaryl, alkoxy, and cyano, and

"substituted sulfonyl" refers to a group -S(0¼R, in which R is substituted with one or more substituents of alkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl. In other embodiments, the one or more substituents may be further substituted with halo, alkyl, haloalkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is substituted. In additional embodiments, the substituents may be further substituted with halo, alkyl, haloalkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is unsubstituted.

PI3 Inhibitor Compounds

(0045] The present application provides the compounds that function as inhibitors of PI3K isoforms. In one aspect, the ΡΓ3Κ inhibitors are the compounds having the structure of formula

(J):

wherein:

n is O, 1, 2, 3, or 4;

m is O, 1, or 2;

W is CH orN;

each R l is independently selected from halo, cyano, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkoxy, optionally substituted sulfonyL, optionally substituted aryl, optionally substituted hcteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;

each R 2 is independently selected from halo, optionally substituted alkyl, optionally substituted haloalkyl;

R 3 is hydrogen, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted cycloalkyl, or optionally substituted aryl;

R 4 is heteroaryl optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted alkyl, and - NH 2 ; and

R 5 is hydrogen or optionally substituted alkyl, wherein R s and R 3 together with the atoms to which they are attached optionally form a heterocyclic ring;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

{0046] The application also provides the compounds having the structure of formula (1) that function as inhibitors of PI3K isofbrms. In one embodiment, the PDK inhibitors are the compounds of the formula (I) having the structure of

(I)

wherein:

n is 1, 2, or 3;

m is O or 1;

each R 1 is independently selected from halo, cyano, optionally substituted CM alkyl, optionally substituted CM haloalkyl, optionally substituted Cj -$ alkoxy, optionally substituted sulfonyi, optionally substituted aryl, optionally substituted C34 heteroaryl, optionally substituted C3-8 cycloalkyl, and optionally substituted C 3 . g heterocycloalkyl;

each R 2 is independently selected from halo and optionally substituted C1 alkyl;

R 3 is hydrogen, optionally substituted alkyl, optionally substituted Q-io aryl» or optionally substituted C34 cycloalkyl;

R 4 is a six- to twelve-membered heteroaryl having at least one aromatic group and at least two hetcroatoms, wherein the heteroatoms are selected from N, O, or S, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, optionally substituted haloalkyl, optionally substituted Ci^ alkyl, and -N¾; and

R 5 is hydrogen or optionally substituted Cj* alkyl, wherein R 5 and R 3 together with the atoms to which they are attached optionally form a four- or eight-membered heterocyclic ring; or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

{0047} In some embodiment, the compounds have the structure of formula (I) wherein

n is 1 or 2;

m is O or 1;

each R l is independently selected from halo, Ct- alkyl, and C14 haloalkyl;

each R 2 is C \ * alkyl;

R 3 is hydrogen, alkyl, or CM cycloalkyl;

R 4 is a six- to twelve-membered heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, -NH 2 , Ct^ haloalkyl, and Ci. 6 alkyl; and

R s is hydrogen, methyl, ethyl, or propyl, or R s and R 3 together with the atoms to which they are attached optionally form a five-membcred heterocyclic ring

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

(0048} In some embodiment, the compound having the structure of the formula (I) wherein: n is 1 or 2;

m is O or 1;

each R 1 is independently selected from chloro, bromo, fluoro, methyl, ethyl, and propyl; each R 2 is independently selected from methyl, ethyl, and propyl;

R 3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, or cyclobutyl;

R 4 is a six- to twelve-membered monocyclic heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the monocyclic heteroaryl is substituted with two or three members independently selected from bromo, chloro, fiuoro, cyano, methyl, ethyl, propyl, and -NH 2 ; and

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

{0049} In some embodiment, the compound having the structure of the formula (I) wherein: n is 1 or 2;

m is O;

each R l is independently selected from chloro, bromo, fiuoro, methyl, ethyl, and propyl;

R 3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, or cyclobutyl;

R 4 is a six- to twelve-membered bicyclic heteroaryl having at least one aromatic ring, at least two nitrogen atoms, and at least one heteroatoms selected from N, O, and S, wherein the bicyclic heteroaryl is optionally substituted with one or two members independently selected from bromo, chloro, fiuoro, cyano, methyl, ethyl, propyl, and -NI¾; and

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

10050] In some embodiment, the compound having the structure of the formula (1) wherein: n is 1 or 2;

m is O;

each R 1 is independently selected from chloro, bromo, fiuoro, methyl, ethyl, and propyl; R 4 is pyzimidinyl substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, and -N¾; and

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

10051] In some additional embodiment, the compounds have the structure of the formula (I) wherein:

nis 1 or 2;

m is O or 1;

each R ! is independently selected from chloro, bromo. fiuoro, methyl, ethyl, and propyl; each R 2 is independently selected from methyl, ethyl, and propyl;

R 3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, or cyclobutyl;

R 4 is a six- to twelve-membered monocyclic heteroaryl having at least one aromatic ring and at least two nitrogen atoms, wherein the monocyclic heteroaryl is substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, fluoromethyl, difluororaethyl. trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, fluoropropyi, difluoropropyl, trifluoropropyl, and - H 2 ; and

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

(0052] In some embodiment, the compounds have the structure of the formula (I) wherein: n is 1 or 2;

m is O;

each l is independently selected from chloro, bromo, fluoro, methyl, ethyl, and propyl;

R 3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, or cyclobutyl;

R 4 is a six- to twelve-membered bicyclic heteroaryl having at least one aromatic ring, at least two nitrogen atoms, and at least one heteroatoms selected from N, O, and S, wherein the bicyclic heteroaryl is optionally substituted with one or two members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, fluoropropyi, difluoropropyl, trifluoropropyl, and -NH 2 ; and

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

{0053] In some embodiment, the compounds have the structure of the formula (I) wherein: n is 1 or 2;

m is O;

each R 1 is independently selected from chloro, bromo, fluoro, methyl, ethyl, and propyl;

R 4 is pyrimidinyl substituted with two or three members independently selected from bromo, chloro, fluoro, cyano, methyl, ethyl, propyl, fluoromethyl, difluoromethyl,

trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, fluoropropyi, difluoropropyl, trifluoropropyl and -N¾; and

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

(0054] In some embodiment, the compounds have the structure of the formula (I) wherein: n is I or 2;

m is O;

each R' is independently selected from chloro, bromo, fluoro, methyl, ethyl, and propyl; R 4 is pyrimidinyt substituted with two or three members, each of which is independently selected from fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difiuoroethyl, trifluoroethyl, fluoropropyl, difluoropropyl, trif!uoropropyl and -NH 2 ; and

R s is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof. 10055] Also, the compounds of formulae (J) or (1) may have the structure of formula (la):

wherein

each R la , R lb , R ,c , and R !d is independently selected from hydrogen, fluoro, chloro, bromo, and iodo;

each R 23 and R* is independently selected from hydrogen, methyl, ethyl, and propyl; and

R 4 is selected from

or a pharmaceutically acceptable salt, isomer, or a mixture thereof, wherein R is optionally substituted with one, two, or three members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and -N¾; and

R 3 and R 5 are described herein;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

10056] In certain embodiment, the compounds have the structure of formula (la), wherein each R la , l , R lc , and R w is independently selected from hydrogen, fluoro, chloro, bromo, and iodo;

each R 2a and R» is independently selected from hydrogen, methyl, ethyl, and propyl; and

R 3 is selected from hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, and cyclobutyl; R 4 is selected from

R s is hydrogen;

or a pharmaceutically acceptable salt isomer, or a mixture thereof.

[0057) In certain embodiment, the compounds have the structure of formula (la), wherein each R.'\ R lb , R lc , and R ,d is independently selected from hydrogen, fluoro, chloro, bromo, and iodo;

each R 2 " and R 2b is independently selected from hydrogen, methyl, ethyl, and propyl; R 3 is selected from hydrogen, methyl, ethyl, propyl, butyl, cyclopropyl, and cyclobutyl;

R 5 is hydrogen;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof. (0058] The compounds of the formulae (J) or (I) may also have the structure of formula (lb):

R", R ,b , R ,c , R ,d , R 2 ", R 2b , R 3 and R 5 are defined herein;

is O, 1, or 2; and

R * is independently selected from halo, cyano, -NH2, and Ci^ alkyl;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

(0059J In certain embodiment, the compounds have the structure of the formula (lb), wherein

R , R lb , R lc , R ld , R 2 *, R a , R 3 and R 5 are defined herein;

p is O, 1, or 2; and

each R 4 " is independently selected from halo, cyano, -NH2, Cj haloalkyl, and Cj. 6 alkyl; or a pharmaceuticaUy acceptable salt, isomer, or a mixture thereof.

(0060) In certain embodiment, the compounds have the structure of the formula (lb), wherein

each R , R ,b , R 1c , R w is independently selected from hydrogen, fluoro, chioro, bromo, and iodo;

each R 2 * and R 2b is independently selected from hydrogen, methyl, ethyl, and propyl; and

R 3 is selected from hydrogen, methyl, ethyl, propyl, butyl, cyciopropyl, and cyclobutyl; p is 1 or 2; and

each R 4 * is independently selected from bromo, chioro, fluoro, cyano, methyl, ethyl, propyl, fiuoromethyi, difluoromethyl, trifiuoromethy!, fluoroethyi, difluoroethyl, trifluoroethyl, iiuoropropyl, ditluoropropyl, trifluoropropyl, and -NH 2 ;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

[0061] In certain embodiment, the compounds have the structure of the formula (lb), wherein

each R ! \ R lb , R lc , R m is independently selected from hydrogen, fluoro, chioro, bromo, and iodo;

each R and R 2b is independently selected from hydrogen, methyl, ethyl, and propyl; and

R 3 is selected from hydrogen, methyl, ethyl, propyl, butyl, cyciopropyl, and cyclobutyl; p is 1 or 2; and each R * is independently selected from fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, fluoropropyl, difluoropropyl, trifluoropropyl, and - N¾;

or a pharmaceutically acceptable salt, isomer, or a mixture thereof.

10062] In one embodiment, n is 0. In some embodiments, n is 1, 2, 3, or 4. In some embodiments, n is 1, 2, or 3. In other embodiments, n is 1 or 2. In certain embodiments, n is 1 and R' moiety may be located on any position of the phenyl of the quinazolinone ring. In another embodiment, n is 2. Both R 1 substituents or moieties may be the same or different. Two R l moieties may be located on any two positions of the phenyl of the quinazolinone ring. By way of example, the first R l may be ortho, meta, or para to the second R\ In yet another embodiment, n is 3. All R 1 substituents or moieties may be the same or different, or two R l may be the same and different from the third R 1 . Three R 1 moieties may be located on any three positions of the phenyl of the quinazolinone ring. For example, the first R 1 may be ortho to the second R 1 , and the first R 1 may be para to the third R 1 . In yet another embodiment, n is 4. All R 1 substituents may be the same or different, three R 1 may be the same and different from the fourth R ! , two R 1 may be the same and different from the third and the fourth R 1 .

10063] In some other embodiments, each R 1 is independently halo, cyano, optionally substituted C^alkyl, optionally substituted Ci^haloalkyl, optionally substituted Ci^alkoxy, hydroxy, optionally substituted C3- cycloalkyl, optionally substituted C3^heterocycloalkyl, optionally substituted C^io ryl, optionally substituted C^gheteroaryl, or optionally substituted Ci alkyisulfbnyl. In certain embodiments, each R 1 is independently halo, cyano, optionally substituted CM alky), optionally substituted C haloalkyl, optionally substituted C alko y, optionally substituted C3- 6 cycloalkyl, or optionally substituted C alkyisulfbnyl. In other embodiments, each R 1 is independently halo, cyano, C haloalkyl, C alkyl, or C

alkyisulfbnyl. In certain embodiments, each R 1 is independently selected from fiuoro, chloro, iodo, bromo, cyano, methyl, ethyl, propyl, butyl, fluoromethyl, fluoroethyl, difluoromethyl, difluoroethyl, trifluoromethyl, trifluoroethyl, methylsulfony), ethylsulfonyl, or propylsulfonyl. In some embodiments, each R 1 is independently fluoro, chloro, iodo, cyano, methyl, ethyl, difluoromethyl (-CHF 2 ), trifluoromethyl (-CF 3 ), methoxy, methylsulfonyl (-SO 2 CH 3 ),

cyclopropylmethyl, or cyclopropyl. In one embodiment, each R 1 is independently fluoro, chloro, cyano, methylsulfonyl, methyl, or trifluoromethyl. {0064] In certain embodiments, m is 0. In some embodiments, m is 0, 1 , or 2. In some other embodiments, m is 1 or 2. When m is 1 , R 2 substituent or moie y may be located on any position of the pyrazine ring. When m is 2, both R 2 substituents may be the same or different.

[0065] In certain embodiments, each R 2 is independently halo, cyano, optionally substituted C alkyl, and optionally substituted C haloalkyl. In some embodiments, each R 2 is independently halo, cyano, C alkyl, and C haloalkyl. In some other embodiments, each R 2 is independently fluoro, chloro, iodo, methoxy, ethoxy, propoxy, butoxy, pentoxy, hexoxy, fluoromethyl (e.g. -CH2F), difluoromethyl (e.g. -CHF2), trifluoromethyl (e.g. -CF3), fluoroethyl, difluoroethyl, triiluoroethyi, methyl, ethyl, propyl, or butyl. In one embodiment, each R 2 is independently fluoro, chloro, methyl, -CHF2, or -CF3.

[0066} In certain embodiment, R 3 is hydrogen, optionally substituted CM alkyl, optionally substituted C 3 - 8 cycloalkyl, or optionally substituted C wo aryl. In one embodiment, R 3 is hydrogen, CM alkyl, C34 cycloalkyl, or Ce-ιο aryl, wherein C alkyl is optionally substituted with C alkoxy, C*.io «ryl, or C34 cycloalkyl, wherein C alkoxy is optionally substituted with CVio aryl. In additional embodiments, R 3 is C alkyl, C 3 ^ cycloalkyl, or C^i aryl, wherein C\. 4 alkyl is optionally substituted with CM alkoxy, CVio aryl, or C3-6 cycloalkyl, wherein CM alkoxy is optionally substituted with phenyl, cyclopropyl, or cyclobutyl. In some embodiments, R J is hydrogen, methyl, ethyl, propyl, butyl, methoxy, ethoxy, cyclopropylmethyl,

cyclopropylbutyl, cyclobutylmethyl, cyclopropylethyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, wherein methoxy and ethoxy is substituted with phenyl, cyclopropyl or cyclobutyl. In other embodiments, R 3 is methyl, ethyl, cyclopropylmethyl, or cyclopropyl.

[0067] In additional embodiments, R 5 is hydrogen or optionally substituted C alkyl. In some embodiments, R 5 is hydrogen or C alkyl. In certain embodiments, R s is hydrogen, methyl, ethyl, propyl or butyl. In certain other embodiments, R 5 is hydrogen.

[0068] In further embodiments, R 3 and R 5 with the atoms to which they are attached (e.g. carbon and nitrogen, respectively) optionally form a heterocyclic ring which is optionally substituted with halo. In other embodiments, the R 3 -R 5 heterocyclic ring is a four- to seven- membered ring. In some other embodiments, the R*-R s heterocyclic ring is a four- to seven- membered ring optionally substituted with fluoro, chloro, bromo, or iodo. In certain other embodiments, the R 3 -R 5 heterocyclic ring is azepanyl, azetidinyl, piperidinyl, and pyrrolidinyi. In some other embodiments, the R 3 -R 3 heterocyclic ring is pyrrolidinyi. In one other embodiment, the R 3 -R 5 heterocyclic ring is a five-membered heterocycloalkyl substituted with ha!o. In other additional embodiments, the R 3 -R 5 heterocyclic ring is pyrrolidinyl substituted with fluoro, chloro, bromo, or iodo.

{0069) In certain embodiments, R 3 is hydrogen, optionally substituted Cj.6 alkyf, optionally substituted C34 cycloalkyl, or optionally substituted CVio aryl. In one embodiment, R 3 is hydrogen, C34 cycloalkyl, CV10 aryl, or C alkyl which is optionally substituted with hydroxyl, Ce-io arylC alkoxy, or C3-6 cycloalkyl. Jn some embodiments, R 3 is hydrogen, methyl, ethyl, propyl, butyl, cyclopropylmethyl, cyclopropylethyl, cyclobutylmethyi, cyclobutylethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -CH2OH, -C2H 4 OH, -C3H6OH,

benzyloxymethyl (i.e.

10070J Tn further embodiments, R 3 and R 5 with the atoms to which they are attached (e.g. carbon and nitrogen, respectively) optionally form a heterocyclic ring. In other embodiments, the R 3 -R 5 heterocyclic ring is a three- to eight-membered heterocycloalkyl (i.e. heterocycloalkyl having three to eight ring members and at least one ring member is a heteroatom). In other embodiments, the R 3 -R s heterocyclic ring is a four- to seven-membered heterocycloalkyl (Le. heterocycloalkyl having four to seven ring members and at least one ring member is a heteroatom). In one embodiment, the R 3 -R 3 heterocyclic ring is a five-membered

heterocycloalkyl. bi certain other embodiments, the R 3 -R 5 heterocyclic ring is C 3 4

heterocycloalkyl. In some other embodiments, the R 3 -R 5 heterocyclic ring is pyrrolidinyl. In one other embodiment, the R 3 -R s heterocyclic ring is a five-membered heterocycloalkyl substituted with one or two members of halo. In other additional embodiments, the R 3 -R 5 heterocyclic ring is pyrrolidinyl substituted with one member of fluoro, chloro, bromo, or iodo.

[0071] In one embodiment, R 4 is heteroaryl having at least two nitrogen atoms and at least one aromatic ring, wherein R 4 heteroaryl is optionally substituted with one, two, or three members independently selected from halo, cyano, -NH 2 , Cj^alkyl, and Ci^haloalkyl. In certain embodiments, R 4 heteroaryl is a six- to twelve-membered heteroaryl (i.e. heteroaryl having six to twelve ring members). In certain embodiments, R 4 heteroaryl is a six- to ten- membered heteroaryl (i.e. heteroaryl having six to ten ring members). R 4 heteroaryl may be a monocyclic or bicyclic heteroaryl. In some embodiments, R 4 heteroaryl is a monocyclic heteroaryl having at least two nitrogen atoms. In certain embodiments, R 4 hetroaryl is a bicyclic heteroaryl having at least one aromatic ring, at least two nitrogen atoms, and at least one additional heteroatom selected from N, O, or S. In certain other embodiments, R 4 heteroaryl is selected from purinyi, pyrimidinyl, thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl , pvrrolopyrimidinyl, furopyrimidinyi, or imidazotriazinyl.

(0072) In any of the foregoing formulae, R 4 is heteroaryl optionally substituted with one, two or three members independently selected from halo, cyano, -NHjt, C1 alkyl, and Ci* haloalkyl, wherein the R 4 heteroaryl is selected from the group consisting of

[0073] In certain other embodiments, R 4 is selected from purinyi, pyrimidinyl,

ftiazolopyrimidinyi, pyridopyrimidinyl, thienopyrimidinyl, pynolopyrimidinyl, furopyrimidinyi, and imidazotriazinyl, each of which is optionally substituted with one, two, or three members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and -NH 2 . In other embodiments, R 4 is selected from purinyi, pyrimidinyl, thiazolopyrimidinyl,

pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrimidinyi, and

imidazotriazinyl, each of which is optionally substituted with one, two, or three members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, fluoromethyl, difluormethyl, trifluoromethyl, fraoroethyl, difluoroethyl, trifluoroethyl, fluoropropy], difluoropropy!, trifluoropropyl, and -NH 2 . In certain other embodiments, R 4 is selected from thiazolopyrirnidinyl, pyridopyrimidinyl, tlnenopyrimidinyl, pyirolopyrimidinyl, ruropyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with two members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, and -NH 2 . In other embodiments, R 4 is selected from thiazolopyrimidinyl, pyridopyrimidinyl, thienopyrimidinyl, pyrrolopyrimidinyl, furopyrirxddinyi, and irmdazotriazinyl, each of which is optionally substituted with independently one member of cbJoro, fluoro, bromo, iodo, and -NH 2 . In other embodiments, R 4 is selected from thiazolopyrimidinyl, pyridopyrimidinyl,

thienopyrimidinyl, pyrrolopyrimidinyl, fcropyrimidinyl, and imidazotriazinyl, each of which is optionally substituted with one or two members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, and -NH 2 . In some other embodiments, R 4 is pyrimidinyi or pyrazinyl, each of pyrimidinyi or pyrazinyl is substituted with at least one -NH 2 . In certain other embodiments, R 4 is pyrimidinyi or pyrazinyl, each of which is substituted with two or three members independently selected from chloro, fluoro, bromo, iodo, methyl, ethyl, propyl, cyano, and -NH 2 . In certain embodiments, R 4 is pyrimidinyi or pyrazinyl, each of which is substituted with at least two or three members, each of the members is independently selected from -NH 2 . fluoromethyl, difluormethyl, trifluoromethyl, fluoroethyl, difluoroeethyl, trifluoroethyl, fluoropropyl, diluoropropyl, and trifluoropyl. In other embodiment, R 4 is selected from

[0074) In one embodiment, p is 0. In some embodiments, p is 1 or 2. In the embodiment where p is 1 , R 4 * moiety may be located on any position of the pyriinidinyl ring. When p is 2, both R 4 * substituents or moieties may be the same or different. In the embodiment where p is 2, both R 4 * substituents or moieties may be the same or different; each of the two R 4 * moieties may be located on any position of the pyrimidinyi ring. In the embodiment where p is 3, all R 4 * substituents may be the same or different, or two R 4 * may be the same and different from the third R 4 ".

(0075] In the present application, each R 4 " is independently selected from halo, cyano, optionally substituted alkyl, and -NH 2 . In one embodiment, each R 4 * is independently halo, cyano, C alkyl, and -NH 2 . In some embodiments, each R 4 * is independently selected from bromo, chloro, fluoro, iodo, cyano, methyl, ethyl, propyl, and -NH 2 . In additional embodiments, each R is independently selected from bromo, chloro, fluoro, iodo, cyano. methyl, ethyl, propyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, fluoropropyl, difluoropropyl, trifluoropropyl, and -NH 2 . In other embodiments, each R 4 * is independently selected fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, fluoropropyl, difluoropropyl, trifluoropropyl, and -NH 2 . Each and every variation of p and R 4 * may be combined with each and every variation of n, m, R l , R 2 , R 3 , and R 5 as described above.

[0076] In the present application, R la , R lb , R tc , and R ,d may be the same or different Each R , R tb , R lc , and R ,d is independently selected from hydrogen, fluoro, chloro, bromo, and iodo. Also, R 2 * and R a may be the same or different. Each R 2 " and R ¾ is independently selected from hydrogen, methyl, ethyl, and propyl. In one embodiment, R is chloro, and R lb R ,c , R ,d , R 2 *, and R 2b are hydrogen. In one other embodiment, R and R w are chloro, and R ,b , R ,c , R ,d , R 2 ", and R 2b are hydrogen. In some other embodiments, R lb is chloro, R 14 is fluoro, and R lb , R lc , R 2 *, and R ¾ are hydrogen. In additional embodiments, R 2 ' and R a are hydrogen. In some additional embodiments, each R la , R lb , R lc , and R ,d is independently selected from hydrogen, chloro, fluoro, bromo, iodo, and methyl.

[0077] In certain embodiments, W is CH or N. In certain other embodiments, W is CH. In yet other embodiments, W is N. Each and every variation of W may be combined with each and every variation of n, m, R l , R 2 , R 3 , R 4 , and R J as described above.

[0078] The compounds of the present application may bear one or more chiral centers. The compounds bearing me chiral center have the same molecular formula and the same chemical name with different stereoisomer designations. For example, the below 3-(5-aminopyrazin-2- yl)-5-chloro-2-(l-((2,6-diamino-5-chloropyrimidin-4-yl)a-^

one bearing one chiral center can be resolved into the (S) and (R) enantiomers, (S)-3-(5- animopyrazin-2-yl)-5-c oro-2-(l-((2,6-dainin^

fluoroquinazolin-4(3H)-one and ((R)-3-(5-amiiu>pyrazm-^^

chloropyrimimn^yl)animo)em^

{0079] Representative compounds of the present application are listed in Table 1 below. Additional representative compounds are listed in Table la below. The compounds may be named using the nomenclature systems and symbols mat are commonly recognized in the art of chemistry including, for example, ChemBioDraw Ultra 12.0, Chemical Abstract Service (CAS), and International Union of Pure and Applied Chemistry (IUPAC). For example, compound 2 in table 1 may be named as 2 ) 4-o^-raimo-6-[[(l S)-l -[3-(5-amiiK pvrazin-2-yl)-5 >hloro-8-fl oxc-^umazolin-2-yl]e l]ammo]py^ or (S)-2,4-diamino-6-((l-(3-(5- aminopyraz -2-yl)-5-chloro-8-fluoro

pyrimidme-5-carbonitrile using IUPAC or ChemBioDraw Ultra 12.0, respectively.

Table 1. Representative Compounds

[0080) The present application provides pharmaceutically acceptable salts, hydrates, solvates, isomers, tautomers, stereoisomers, enantiomers, racemates, atropisomers, polymorphs, prodrugs, or a mixture thereof, of the compounds described herein. In addition, the present application provides the compounds in which from 1 to n hydrogen atoms attached to a carbon atom may be replaced by a deuterium atom or D, in which n is the number of hydrogen atoms in the molecule. It is known mat the deuterium atom is a non-radioactive isotope of the hydrogen atom. Such compounds may increase resistance to metabolism, and thus may be useful for increasing the half-life of the compounds of any of the formulae described herein or

pharmaceutically acceptable salts, isomers, prodrugs, or solvates thereof, when administered to a mammal. See, e.g., Foster, "Deuterium Isotope Effects in Studies of Drug Metabolism", Trends Pharmacol. Sri., 5(12):52 -527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogen atoms have been replaced by deuterium.

{0081] The terms "a compound of the present application," "a compound described herein," "a compound of any of the formulae described herein, * ' or variant thereof refer to a compound having the structure of any of the formulae (J), (I), (la), and (lb). In some embodiments, compounds of the present application are Compounds 1-21 as described herein.

[0082] "Pharmaceutically acceptable" or physiologically acceptable' * refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use.

"Pharmaceutically acceptable salts" or "physiologically acceptable salts" refer to salts of pharmaceutical compounds that retain the biological effectiveness and properties of the underlying compound, and which are not biologically or otherwise undesirable. There are acid addition salts and base addition salts. Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Acids and bases useful for reaction with an underlying compound to form pharmaceutically acceptable salts (acid addition or base addition salts respectively) are known to one of skill in the art. Similarly, methods of preparing pharmaceutically acceptable salts from an underlying compound (upon disclosure) are known to one of skill in the art and are disclosed in for example, Berge, at al. Journal of Pharmaceutical Science, Jan. 1977 vol.66, No.1 , and other sources. If the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.

(0083] ' somers" refers to compounds that have the same molecular formula. As used herein, the term isomers include double bond isomers, racemates, stereoisomers, enantiomers, diastereomers, and atropisomers. Single isomers, such as enantiomers or diastereomers, can be obtained by asymmetric synthesis or by resolution of a mixture of isomers. Resolution of a mixture of isomers (e.g. racemates) maybe accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral high pressure liquid chromatography (HPLC) column. "Double bond isomers" refer to Z- and E- forms (or cis- and trans- forms) of the compounds with carbon-carbon double bonds.

10084] "Atropisomers" refers to conformational stereoisomers which occur when rotation about a single bond in the molecule is prevented, or greatly hindered, as a result of steric interactions with other parts of the molecule and the substituents at both ends of the single bond are asymmetrical, i.e., they do not require a stereocenter. Where the rotational barrier about the single bond is high enough, and interconversion between conformations is slow enough, separation and isolation of the isomeric species may be permitted. Atropisomers may be separated by the methods well known in the art Unless otherwise indicated, the description is intended to include individual atropisomers as well as mixtures. Also, as understood by those skilled in the art, the atropisomers may be represented by the same chemical name with different atropisomer designations.

|0085] "Racemates' * refers to a mixture of enantiomers.

{0086] "Stereoisomers" or "stereoisomeric forms refer to compounds that differ in the chirality of one or more stereocenters. Stereoisomers include enantiomers and diastereomers. The compounds may exist in stereoisomeric form if they possess one or more asymmetric centers or a double bond with asymmetric substitution and, therefore, can be produced as individual stereoisomers or as mixtures. Unless otherwise indicated, the description is intended to include individual stereoisomers as well as mixtures. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art (see, e.g., Chapter 4 of Advanced Organic Chemistry, 4th ed., J. March, John Wiley and Sons, New York, 1992).

[0087] 'Tautomers" or "tautomeric formers" refer to alternate forms of a compound that differ in the position of a proton, such as enol-keto and irmne-enamine tautomers, or heteroaryls such as pyrazoles, imidazoles, benzimidazoles, triazoles, and tetrazoles.

(0088] A "solvate" is formed by the interaction of a solvent and a compound. Solvates of salts of the compounds of any of the formulae described herein are also provided. Hydrates of the compounds of any of the formulae are also provided.

J 0089] A "prodrug" is defined in the pharmaceutical field as a biologically inactive derivative of a drug that upon administration to the human body is converted to the biologically active parent drug according to some chemical or enzymatic pathway.

[0090] In any one of the foregoing embodiments, the compound described herein or a pharmaceutically acceptable salt thereof is an (S)-enantiomer. In any one of the foregoing embodiments, the compound described herein or a pharmaceutically acceptable salt thereof is an (R)-enantiomer. In any one of the foregoing embodiments, the compound described herein or a pharmaceutically acceptable salt thereof is an atropisomer.

[0091] The application also provides a composition containing a mixture of enantiomers of the compound or a pharmaceutically acceptable salt thereof In one embodiment, the mixture is a racemic mixture. In other embodiments, the composition comprises the (S)-enantiomer of a compound in excess of over the corresponding the (R)-enantiomer of the compound, hi some embodiments, the composition contains the (S)-enantiomer of the compound and is substantially free of its corresponding (R)-enantiomer. In certain embodiments, a composition substantially free of the (R)-enantiomer has !ess than or about 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5%, 1%, 0.05%, or 0.01% of the (R)-enantiomer. In other embodiments, the composition containing the (S)-enantiomer of a compound or a pharmaceutically acceptable salt thereof, predominates over its corresponding (RVenantiomer by a molar ratio of at least or about 9: 1 , at least or about 1 : 1 , at least or about 40: 1 , at least or about 80: 1 , at least or about 160: 1 , or at least or about 320:1.

|0092] The composition containing a compound according to any of the formulae described herein or a pharmaceutically acceptable salt thereof, may also contain the compound in enantiomeric excess (e.e.). By way of example, a compound with 95% (S)-isomer and 5% (R)- isomer will have an e.e. of 90%. In some embodiments, the compound has an e.e. of at least or about 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%.

(0093] In any one of the foregoing embodiments, the compound or a pharmaceutically acceptable salt thereof, is an atropisomer. Another embodiment provides the composition containing a mixture of atropisomers of the compound or a pharmaceutically acceptable salt thereof. By way of example, a compound with 95% of one atropisomer and 5% of the other atropisomers. In some embodiments, a compound with about 90, 80, 70, 0, 50, 40, 30, 20, or 10% of one atropisomer and 10, 20, 30, 40, 50, 60, 70, 80, or 90%, respectively, of the other atropisomers.

10094] The application also provides the free base forms of the compounds described herein. In certain embodiments, provided herein are the enantiomers, ( ) or (S), of the compounds of the formulae described herein. In other embodiments, provided herein are the atropisomers of the compounds of the formulae described herein.

[0095] The application further provides compositions comprising the compounds described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof. The composition may include racemic mixtures, mixtures containing an enantiomeric excess of one enantiomer or single diastereomers or diastereomeric mixtures. All such isomeric forms of these compounds are expressly included herein, the same as if each and every isomeric form were specifically and individually listed. [0096J In certain embodiments, provided herein are also pol morphs, such as crystalline and amorphous forms, of the compounds described herein. In some embodiments, provided are also chelates, non-covaient complexes, and mixtures thereof, of the compounds of the formula described herein or pharmaceutically acceptable salts, prodrugs, or solvates thereof. A "chelate" is formed by the coordination of a compound to a metal ion at two (or more) points. A "non- covalent complex" is formed by the interaction of a compound and another molecule wherein a covalent bond is not formed between the compound and the molecule. For example, complexation can occur through van dear Waals interactions, hydrogen bonding, and electrostatic interactions (also called ionic bonding).

Therapeutic Uses of the Compounds

{0697) The compounds of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof may be used for the treatment of diseases and/or conditions mediated by PI3K isoforms. In addition, the application provides the compounds for use in therapy. Also, provided herein are methods for inhibiting one or more PI3K. isoforms. In one embodiment, provided are methods for inhibiting PI3 activity using the compound described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof. In other embodiment, provided are methods for inhibiting PI3KS and/or ΡΙ3 β activities using the compound or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof. The application further provides methods for use in such methods. The PI3K isoforms may be selectively or specifically inhibited. Additionally, the compounds may be used to inhibit ΡΓ3Κ activity therapeutically or prophylactically, such as ΡΙ3Κδ and/or ΡΙ3 β.

[00981 The compounds according to the present application may be used in combination with one or more additional therapeutic agents. The therapeutic agents may be in the forms of compounds, antibodies, polypeptides, or polynucleotides. The therapeutic agent includes, but is not limited to, a chemotherapeutic agent, an irnmunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. In one embodiment, the application provides a product comprising a compound described herein and an additional therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy, e.g. a method of treating a disease, disorder, or condition that is mediated by PI3K isoforms. (0099] Also, the therapeutic agents may be those that inhibit or modulate the activities of Bruton's tyrosine kinase, spleen tyrosine kinase, apoptosis signal-regulating kinase, Janus kinase, lysyl oxidase, lysyl oxidase-Iike proteins, matrix metallopeptidase, bromodomain- containing protein, adenosine A2B receptor, isocitrate dehydrogenase, serine/threonine kinase TPL2, discoidin domain receptor, serine/thxeonine-protein kinases, IKK, MEK, EGFR, hi stone deacetylase, protein kinase C, or any combination thereof. In certain embodiments, the therapeutic agent may be selected from a PI3K (including ΡΙ3Κγ, ΡΒΚδ, ΡΙ3Κβ, ΡΟΚα, and/or pan-PI3K) inhibitor, a JAK (Janus kinase, including JAK1 ,JAK2, and/or JAK3) inhibitor, a SYK (spleen tyrosine kinase) inhibitor, a BTK (Bruton's tyrosine kinase) inhibitor, an A2B (adenosine A2B receptor) inhibitor, an ACK (activated CDC kinase, including ACK1) inhibitor, an ASK (apoptosis signal-regulating kinase, including ASK1) inhibitor, Auroa kinase, a BRD (bromodomain-containing protein, including BRIM) inhibitor, a Bel (B-cell CLL lymphoma, including Bcl-1 and/or Bcl-2) inhibitor, a CAK (CDK-activating kinase) inhibitor, a CaMK (calmodulin-dependent protein kinases) inhibitor, a CDK (cyclin-dependent kinases, including CDKl, 2, 3, 4, and or 6) inhibitor, a CK (casein kinase, including CK1 and/or CK2) inhibitor, a DDR (discoidin domain receptor, including DDR1 and/or DDR2) inhibitor, a EGFR inhibitor, a FXR (farnesoid x receptor) inhibitor, a FAK (focal adhesion kinase) inhibitor, a GSK (glycogen synthase kinase) inhibitor, a HDAC (histone deacetylase) inhibitor, an IDO (indoleamine 2,3- dioxygenasc) inhibitor, an IDH (isocitrate dehydrogenase, including IDH1) inhibitor, an TKK (I- Kappa-B kinase) inhibitor, a KDM5 (lysine demethylase) inhibitor, a LCK (lymphocyte-specific protein tyrosine kinase) inhibitor, a LOX (lysyl oxidase) inhibitor, a LOXL (lysyl oxidase like protein, including LOXL1, LOXL2, LOXL3, LOXL4, and/or LOXL5) inhibitor, a MTH (mut T homolog) inhibitor, a MEK (mitogen-activated protein kinase kinase) inhibitor, a matrix metalloprotease (MMP, including MMP2 and or MMP9) inhibitor, a mitogen-activated protein kinases (MAPK) inhibitor, a PD-1 (programmed cell death protein 1) inhibitor, a PD-L1 (programmed death-ligand 1) inhibitor, a PDGF (platelet-derived growth factor) inhibitor, a phosphorylase kinase (PK) inhibitor, a PLK (polo-like kinase, including PLK1, 2, 3) inhibitor, a protein kinase (PK, including protein kinase A, B, C) inhibitor, a STK (serine threonine kinase) inhibitor, a STAT (signal transduction and transcription) inhibitor, a serine/thiwnine-protein kinase inhibitor, a TBK (tank-binding kinase) inhibitor, a TLR (toll-like receptor modulators, including TLR-1, TLR-2, TLR-3, TLR-4, TLR-5, TLR-6, TLR-7, TLR-8, TLR-9, TLR-10, TLR- 11, TLR- 12, and/or TLR- 13) inhibitor, a TK (tyrosine kinase) inhibitor, a TPL2

(serine threonine kinase) inhibitor, a NEK9 inhibitor, an Abl inhibitor, a p38 kinase inhibitor, a PYK inhibitor, a c-Kit inhibitor, a NP -ALK inhibitor, a Flt-3 inhibitor, a c-Met inhibitor, a DR inhibitor, a TIE-2 inhibitor, a VEGFR inhibitor, a SRC inhibitor, a HCK inhibitor, a LYN inhibitor, a FYN inhibitor, a YES inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti- proiiferation agent, an anti-fibrotic agent, an antt-angiogenic agent, a therapeutic antibody, or any combination thereof. In some embodiments, the JAK inhibitor is N-(cyanomethyl)-4-[2-{4- rao holinoanilino) yrimicUn^yl]benza-I^de as named by ChemDraw (may also be referred to as CYT0387 or momelotinib) and may be synthesized by the methods described in U.S. Patent No.8,486,941. In certain embodiment, the Sy inhibitor is 6-(lH-indazol-6-yi)-N-(4- mo holino henyI)iπlidazo[ 1 ,2-a]pyrazin-8-amine as named by ChemDraw (may also be referred to as 6^1H ndazol^yl)-N-[4-(m^to

and may be synthesized by the methods described in U.S. Patent No. 8,450,321. In other embodiments, the BT inhibitor is (S)-6^armnc-9-(l^ut-2-ynoyl)pyrToUdin-3-yI)-7-(4- phenoxyphenyl)-7H-pimn-8(9H)-one as named by ChemDraw (may also be 6-amino-9-((3R)-l - (2-butynoyl)-3-pyrroUdinyl]-7-(4-phenoxyphenyl)-7 ,9-dihydro-8H -purin-8-one) and may be synthesized by the methods in U.S. Pat. No. 8,557,803.

[00100] Chemotherapeutic agents may be categorized by their mechanism of action into, for example, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (floxuridine, capecitabine, and cytarabine); purine analogs, folate antagonists and related inhibitors, antiproliferative antimitotic agents including natural products such as vinca alkaloid (vinblastine, vincristine) and microtubule such as taxane (paclitaxel, docetaxel), vinblastin, nocodazole, epothilones and navelbine, epidipodophyliotoxins (etoposide, teniposide); DNA damaging agents (actinomycin, amsacrine, busulfan, carboplatin, chlorambucil, cisplatin, cyclophosphamide, Cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin,

ipbospharnide, melphalan, merchlorehtamine, mitomycin, mitoxantrone, nitrosourea, procarbazine, taxol, taxotere, teniposide, etoposide, triethyienethiophosphoramide); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (^asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative antimitotic alkylating agents such as nitrogen mustards cyclophosphamide and analogs, melphalan, chlorambucil), and (hexamethylmelam ine and thiotepa), alkyl nitrosoureas (BCNU) and analogs, streptozoctn), trazenes-dacarbazinine (DΉC); annprolifmtive/antimitouc antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, oxiloplatinim, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidinc, clopidogrel; antimigratory agents; antisecretory agents (breveldin); immunosuppressives tacrolimus sirolimus azathioprine, mycophenolate; compounds (TNP-470, genistein) and growth factor inhibitors (vascular endothelial growth factor inhibitors, fibroblast growth factor inhibitors); angiotensin receptor blocker, nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab, rituximab); cell cycle inhibitors and differentiation inducers (tretinoin); inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin, irinotecan and mitoxantrone, topotecan, irinotecan, camptothesin), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone); growth factor signal transduction kinase inhibitors; dysfunction inducers, toxins such as Cholera toxin, ricin, Pseudomonas exotoxin, Bordetelia pertussis adenylate cyclase toxin, or diphtheria toxin, and caspase activators; and chromatin.

[00101 ] As used herein (he term "chemotherapeutic agent" or "chemotherapeutic" (or "chemotherapy," in the case of treatment with a chemotherapeutic agent) is meant to encompass any non-proteinaceous (i.e, non-peptidic) chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and

cyclophosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; emylerumines and memylamelamines including alfretamine, triemylenemelamine, triemylenephosphoramide, triethylenethiophosphoramide and trimemylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (articularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, W-2189 and CBI-TMI); eleutherobin pancratistatin; a sarcodicryin; spongi statin; nitrogen mustards such as chlorambucil, chloniaphazine, cholophosphamide, estramustine, ifbsfamide, mechlorethamine, mechloremamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fbremustine, lomustine, nimustine, ranimus ine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammail and caUcheamicin phill, see, e.g., Agnew, Chem. Intl. Ed. Engl, 33:183-186 (1994); dvnemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycm, carabicin, camunomycin, carzinophilin, chromomycins, dactinomycin, daimorubicin, detorubicin, 6-diazo- 5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino- doxorubicin, 2-pyrrolino-doxon-bicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin,

olivomycins, peplomycin, potfiromycm, puromycin, quelamycin, rodorubicin, streptonigrin, stieptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as demopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogues such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replinisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; hestrabucil; bisantrene; edatraxate; defo&mine; demecolcine; diaziquone; elformthine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; leucovorin; lonidamine;

maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; losoxantrone; fluoropyrimidine; folinic acid; podophyUinic acid; 2-ethylhydrazide; procarbazine; PS (r); razoxane; rhizoxin; sizofiran spirogermanium; tenuazonic acid; triaziquone; 2,2 2"-tricUorotriemylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethane; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C");

cyclophosphamide; thiopeta taxoids, e.g., pactitaxel (TAXOL(r) and docetaxel

(TAXOTERE(r)); chlorambucil; gemcitabine (Gemzar(r)); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum;

etoposide (VP-16); ifosfamide; mitroxantrone; vancristine; vinorelbine (Navelbine(r));

novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeoloda; ibandronate; CPT-11 ; topoisomerase inhibitor RFS 2000; difluoromethylorni thine (D FO); retinoids such as retinoic acid; capecitabine; FOLFIRI (fluorouracil, leucovorin, and irinotecan) and pharmaceutically acceptable salts, acids or derivatives of any of the above. One or more chemotherapeutic agent are used or included in the present application.

[00102| Also included in the definition of "chemotherapeutic agent" are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including

Nolvadex™), raloxifene, droloxtfene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene ( areston(r»; inhibitors of the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles,

aminoglutethimide, megestrol acetate (Megace(r)), exemestane, fbrmestane, fadrozoie, vorozole (Rivisor(r)), letrozole (Femara(r)), and anastrozole (Arimidex(r).); and anti-androgens such as flutamide, mlutamide, bicalutamide, leuprohde, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.

[00103] The anti-angiogenic agents include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATIN(r), ENDOSTATIN(r), suramin, squalamine, tissue inhibitor of metalloproteinase-l, tissue inhibitor of metalloprotemase-2, plasminogen activator inhibitor- 1, plasminogen activator inbibitor-2, cartilage-derived inhibitor, paclitaxel (nab-paclitaxel), platelet factor 4, protamine sulphate (clupeine), sulphated chitin derivatives (prepared from queen crab shells), sulphated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-azetidine-2-carboxylic acid (LACA), cishydroxyproline, d,I-3,4-dehydroproline, thiaproline, .alpha.-dipyridyl, beta-aminopropionitxile fumarate, 4-pix)pyl-5-(4-pyridinyl)-2(3h)-oxazolone methotrexate, mitoxantrone, heparin, interferons, 2 macroglobulin-serum, chimp-3, chymostatin, beta-cyclodextrin tetradecasulfate, eponemycin; fumagillin, gold sodium thiomalate, d- peniciUamine (CDPT), beta-l-anticollagenase-serum, alpba-2-annplasrnin, bisantrene, lobenzarit disodium, n-2-c»rboxyphenyl-4-chloroantl x)niHc acid disodium or "CCA", thalidomide;

angiostatic steroid, cargboxynaminolmidazole; metalloproteinase inhibitors such as BB 4. Other anti-angiogenesis agents include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: beta-FGF, alpha-FGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-l/Ang-2. See Ferrara N. and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.

|00104] The anti-fibrotic agents include, but are not limited to, the compounds such as beta- aminoproprionitrile (BAPN), as well as the compounds disclosed in U.S. Pat No. 4,965,288 to Palfteyman, et al., issued Oct.23, 1990, entitled "Inhibitors of lysyl oxidase," relating to inhibitors of lysyl oxidase and their use in the treatment of diseases and conditions associated with the abnormal deposition of collagen; U.S. Pat. No.4,997,854 to Kagan, et al., issued Mar. 5, 1991, entitled "Anti-fibrotic agents and methods for inhibiting the activity of lysyl oxidase in situ using adjacently positioned diamine analogue substrate," relating to compounds which inhibit LOX for the treatment of various pathological fibrotic states, which are herein

incorporated by reference. Further exemplary inhibitors are described in U.S. Pat. No.4,943,593 to Palfreyman, et al., issued Jul.24, 1990, entitled "Inhibitors of lysyl oxidase," relating to compounds such as 2-isobutyl-3-fluoro-, chloro-, or bromo-allylamine; as well as, e.g., U.S. Pat. No. 5,021,456; U.S. Pat. No. 5,5059,714; U.S. Pat. No. 5,120,764; U.S. Pat. No. 5,182,297; U.S. Pat. No. 5,252,608 (relating to 2-(l-naphthyloxymem>i)-3-fl\.oroallylamine); and U.S. Patent Application No.2004/024887 , which are herein incorporated by reference. Exemplary anti- fibrotic agents also include the primary amines reacting with the carbonyl group of the active site of the lysyl oxidases, and more particularly those which produce, after binding with the carbonyl, a product stabilized by resonance, such as the following primary amines:

emyienemamine, hydrazine, phenymydrazine, and their derivatives, semicarbazide, and urea derivatives, aminonitriles, such as beta-aminopropionitrile (BAPN), or 2-nitroethylamine, unsaturated or saturated haloamines, such as 2-bromo-ethylamine, 2-chloroethylamine, 2- trifluoroethylamine, 3-bromopropylamine, p-halobenzyiamines, selenohomocysteine lactone. Also, the anti-fibrotic agents are copper chelating agents, penetrating or not penetrating the cells. Exemplary compounds include indirect inhibitors such compounds blocking the aldehyde derivatives originating from the oxidative deamination of the lysyl and hydroxylysyl residues by the lysyl oxidases, such as the tMolamines, in particular D-penicillamine, or its analogues such as 2-amino-5-mercapto-5-methylhexanoic acid, D-2-amino-3-methyl-3-((2- acetamidoethyl)dithio)buumoic acid, p-2-ammo-3-memyl-3-((2-amtt^

acid, sodium^((r Hlimerayl~2-am sulphurate, 2- acetanudoemyl-2-acetamidoemanethiol sulphanate, sodium^mercaptobutanesulphinate trihydrate.

[00105] The immunotherapeutic agents include and are not limited to therapeutic antibodies suitable for treating patients; such as abagovomab, adecatumumab, afutuzumab, alemtuzumab, altumomab. amatuximab, anatumomab, arcitumomab, bavituximab, bectumomab, bevacizumab, bivatuzumab, blinatumomab, brentuximab, cantuzumab, catumaxomab, cetuximab, citatuzumab, cixutumumab, clivatuzumab, conatumumab, daratumumab, drozitumab, duligotumab, dusigitumab, detumomab, dacetuzumab, dalotuzumab, ecromeximab, elotazumab, ensituximab, ertumaxomab, etaracizuraab, farietuzurnab, ficlatuzuniab, figitumumab, flanvotumab, futuximab, ganitumab, gemtuzumab, girentuximab, glembatumumab, ibritumomab, igovomab, imgatuzumab, indatuxirnab, inotuzumab, inteturaumab, ipilimumab, iratumumab, labetuzurnab, lexaturauraab, lintuzumab, lorvotuzuraab, lucatumumab, mapa umumab, maruzumab, milatuzumab, minreturaomab, mitumomab, moxetumomab, naraatumab, naptumomab, necitumumab, nimotuzumab, nofetumomabn, ocaratuzumab, ofatumumab, olaratumab, onartuzumab, oportuzumab, oregovomab, panitumumab, parsatuzumab, patritumab,

pemtumomab, pertuzumab, pintumomab, priturnumab, racotumomab, radretumab, iilotumttmab, rituximab, robaturauraab, sarumomab, sibrotuzumab, siltuximab, simtuzumab, solitomab, tacatuzumab, taplitumomab, tenaturaomab, teprorumurnab, tigatuzumab, tositumomab, trastuzumab, tucotuzumab, ublituximab, veltuzumab, voreetuzumab, votumumab, zalutumumab, obinutuzumab, CC49 and 3F8. The exemplified therapeutic antibodies may be further labeled or combined with a radioisotope particle, such as indium In 111 , yttrium Y 90, iodine 1-131.

[00106] The application also provides method for treating a subject who is undergoing one or more standard therapies, such as chemotherapy, radiotherapy, immunotherapy, surgery, or combination thereof. Accordingly, one or more therapeutic agent or inhibitors may be administered before, during, or after administration of chemotherapy, radiotherapy,

immunotherapy, surgery or combination thereof.

1001071 Other examples of chemotherapy treatments (including standard or experimental chemotherapies) are described below. In addition, treatment of certain lymphomas is reviewed in Cheson, B.D., Leonard, J.P., "Monoclonal Antibody Therapy for B-Cell Non-Hodgkin's Lymphoma" The New England Journal of Medicine 2008, 359(6), p. 613-626; and Wierda, W.G., "Current and Investigational Therapies for Patients with CLL" Hematology 2006, p. 285- 294. Lymphoma incidence patterns in the United States is profiled in Morton, L.M., et al.

"Lymphoma Incidence Patterns by WHO Subtype in the United States, 1992-2001" Blood 2006, 107(1), p. 265-276.

(00108] Examples of immunotherapeutic agents include, but are not limited to, rituximab (such as Rituxan), alemtuzumab (such as Campath, MabCampath), anti-CD 19 antibodies, anti- CD20 antibodies, anti-MN-14 antibodies, anti-TRAIL, Anti-TRAIL DR4 and DR5 antibodies, anti-CD74 antibodies, apolizumab, bevacizumab, CHlR-12.12, epratuzumab (hLL2- a ti-CD22 humanized antibody), galiximab, ha20, ibritumomab tiuxetan, lumiliximab, milatuzumab, ofetumumab, PRO131921, SGN-40, WT-l analog peptide vaccine, WT1 126-134 peptide vaccine, tositumomab, autologous human tumor-derived HSPPC-96, and veltuzumab.

Additional immunotherapy agents includes using cancer vaccines based upon the genetic makeup of an individual patient's tumor, such as lymphoma vaccine example is GTOP-99 ( yVax*).

(00109] Examples of chemotherapy agents include aldesleukin, alvocidib, antineoplaston AS2-1, antineoplaston A 10, anti-thymocyte globulin, amifostine trihydrate, aminocamptothecin, arsenic trioxide, beta alethine, Bcl-2 family protein inhibitor ABT-263, ABT-199, BMS-345541 , bortezomib (Velcade ® ), bryostatin I, busulfan, carboplatin, campath-lH, CC-S103, carmustine, casporungin acetate, clofarabine, cisplatin, Cladribine (Leustarin), Chlorambucil (Leukeran), Curcumin, cyclosporine, Cyclophosphamide (Cytoxan, Endoxan, Endoxana, Cyclostin), cytarabine, denileukin diftitox, dexamethasone, DT PACE, docetaxel, dofastatin 10,

Doxorubicin (Adriamycin ® , Adriblastine), doxorubicin hydrochloride, enzastaurin, epoetin alfa, etoposide, Everolimus (RAD001), fenretinide, filgrastim, melphalan, mesna, Flavopiridol, Fludarabine (Fludara), Geldanamycin (17- A AG), ifosfamide, irinotecan hydrochloride, ixabepilone, Lenalidomide (Revlimid ® , CC-S013), lymphokine-activated killer cells, melphalan, methotrexate, mitoxantrone hydrochloride, motexafin gadolinium, mycophenolate mofetil, nelarabine, oblimersen (Genasense) Obatoclax (GX15-070), oblimersen, octreotide acetate, omega-3 fetty acids, oxaliplatin, paclitaxel, PD0332991, PEGylated liposomal doxorubicin hydrochloride, pegfilgrastim, Pentstatin (Nipent), perifosine, Prednisolone, Prednisone, R- roscovitine (Selicilib, CYC202), recombinant interferon alfa, recombinant interleukin-12, recombinant interleukin-11, recombinant flt3 ligand, recombinant human thrombopoietin, rituximab, sargramostim, sildenafil citrate, simvastatin, sirolimus, Styryl sulphones, tacrolimus, tanespimycin, Temsirolimus (CCl-779), Thalidomide, therapeutic allogeneic lymphocytes, thiotepa, tipifarnib, Velcade* (bortezomib or PS-341), Vincristine (Oncovin), vincristine sulfate, vinorelbine ditartrate, Vorinostat (SAH A), vorinostat, and FR (fludarabine, rituximab), CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), CVP (cyclophosphamide, vincristine and prednisone), FCM (fludarabine, cyclophosphamide, mitoxantrone), FCR (fludarabine, cyclophosphamide, rituximab), hyperCVAD (hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate, cytarabine), ICE .phosphatide, carboplatin and etoposide), MCP (mitoxantrone, chlorambucil, and prednisolone), R-CHOP (rituximab plus CHOP), R-CVP (rituximab plus CVP), R-FCM (rituximab plus FCM), R-ICE (rituximab-lCE), and R-MCP (R-MCP). (00110) The therapeutic treatments can be supplemented or combined with any of the abovementioned therapies with stem cell transplantation or treatment. One example of modified approach is radioimmunotherapy, wherein a monoclonal antibody is combined with a radioisotope particle, such as indium In 111, yttrium Y 90, iodine 1- 31. Examples of combination therapies include, but are not limited to, Iodine-131 tositumomab (Bexxar*), Yttrium-90 ibritumomab tiuxetan (Zevalin*), Bexxar* with CHOP.

(00111] Other therapeutic procedures include peripheral blood stem cell transplantation, autologous hematopoietic stem cell transplantation, autologous bone marrow transplantation, antibody therapy, biological therapy, enzyme inhibitor therapy, total body irradiation, infusion of stem cells, bone marrow ablation with stem cell support, in vftro-treated peripheral blood stem cell transplantation, umbilical cord blood transplantation, immunoenzyme technique, pharmacological study, low-LET cobalt-60 gamma ray therapy, bleomycin, conventional surgery, radiation therapy, and nonm yeloablative allogeneic hematopoietic stem cell

transplantation.

[00112] In some embodiments, the methods include administering a compound of the formula described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, in a therapeutically effective amount to a human in need thereof. The method can be employed to treat a patient who has or is believed to have a disease or condition whose symptoms or pathology is mediated by expression or activity of ΡΙ3Κβ and or PI3 6. The patient may be a mammal or a human. In certain embodiment, the patient may be a human.

(00113] 'Treatment" or ''treating" is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired clinical results may include one or more of the following: a) inhibiting the disease or condition (e.g., decreasing one or more symptoms resulting from the disease or condition, and/or diininishing the extent of the disease or condition); b) slowing or arresting the development of one or more clinical symptoms associated with the disease or condition (e.g., stabilizing the disease or condition, preventing or delaying the worsening or progression of the disease or condition, and/or preventing or delaying the spread (e.g., metastasis) of the disease or condition); and/or c) relieving the disease, that is, causing die regression of clinical symptoms (e.g., ameliorating die disease state, providing partial or total remission of the disease or condition, enhancing the effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival. [00114] "Prevention" or "preventing" mean any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. Compounds may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition.

[00115) "Subject" or "patient" refer to an animal, such as a mammal (including a human), that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in human therapy and/or veterinary applications, hi some embodiments, the subject is a mammal. In one embodiment, the subject is a human. "Human in need thereof refers to a human who may have or is suspect to have diseases, or disorders, or conditions that would benefit from certain treatment; for example, being treated with the PI3 inhibitor of the compounds according to the present application. In certain embodiments, the subject may be a human who (i) has not received any treatment including chemotherapy treatment, (ii) is substantially refractory to at least one chemotherapy treatment, (iii) is in relapse after treatment with chemotherapy, or both (i) and (ii). In some of embodiments, the subject is refractory to at least one, at least two, at least three, or at least four chemotherapy treatments (including standard or experimental chemotherapies).

[00116} The terms 'therapeutically effective amount" or "effective amount" of a compound of the present application or a pharmaceutically acceptable salt, isomers, prodrug, or solvate thereof, mean an amount sufficient to effect treatment when administered to a subject, to provide a therapeutic benefit such as amelioration of symptoms or slowing of disease progression. For example, a therapeutically effective amount may be an amount sufficient to decrease a symptom of a disease or condition responsive to inhibition of PI3K6 and ΡΙ3 β activity. The

therapeutically effective amount may vary depending on the subject, and disease or condition being treated, the weight and age of the subject, the severity of the disease or condition, and the manner of administering, which can readily be determined by one or ordinary skill in the art.

[00117] In addition to the therapeutic uses, the compounds described herein have the selectivity or selective inhibition to certain PI3K isoforms. In one embodiment, the compounds have selectivity to ΡΙ3Κβ. In some embodiments, the compounds have selectivity to PI3 6. In yet other embodiments, the compounds have selectivity to Ρ13 β and PI3K& The selectivity to PI3 isoforms may be determined by measuring the compound's activity in inhibiting certain PI3K isoforms using the assay described in the example below or the methods commonly used. It is understood that the conditions (e.g. the reagent concentration or the incubation temperature) may be varied and the results of the assay may vary. In some instances, the value may vary within a range of one to three-folds.

[001181 The term "inhibition " " indicates a decrease in the baseline activity of a biological activity or process. The term "inhibition of activity of PI3 isofonns" or variants thereof refer to a decrease in activity in any PI3K. isoform (e.g., alpha, beta, gamma, or delta) as a direct or indirect response to the presence of a compound of any of the formula described herein relative to the activity of PI3K isoform in the absence of such compound. "Inhibition of PI3K5 and/or ΡΙ3 β activities" or variants thereof refer to a decrease in PI3K6 and or ΡΙ3 β activities as a direct or indirect response to the presence of the compounds described herein, relative to the activities of PI3K5 and/or ΡΙ3Κβ in the absence of such compound. In some embodiments, the inhibition of PI3K isoform activities may be compared in the same subject prior to treatment, or other subjects not receiving the treatment.

[00 19) Without being bound to any theory, the decrease in the activity of PI3K may be due to the direct interaction of the compound with PI3 , or due to the interaction of the compounds described herein with one or more other factors that affect PT3K activity. For example, the presence of the compounds may decrease the activities of PI3K5 and/or Ρί3Κβ by directly binding to PBK5 and/or PI3KJ3, by causing (directly or indirectly) another factor to decrease PI3K6 and or ΡΙ3Κβ activities, or by (directly or indirectly) decreasing the amount of PT3 5 and/or ΡΙ3Κβ present in the cell or organism.

[001201 The term "PI3K inhibitor" or variant thereof refers to a compound that inhibits the activity of PI3K. The term "PI3K isoform selective inhibitor" or variant thereof refers to a compound mat inhibits the activity of one or more PI3K isoforms more effectively than the other remaining PI3K isoforms. By way of example, the term Μ ΡΙ3 β selective inhibitor" generally refers to a compound that inhibits the activity of the ΡΙ3Κβ isoform more effectively than other isoforms of the PI3K family, and the term "PI3K δ selective inhibitor' * generally refers to a compound that inhibits the activity of the PI3K5 isoform more effectively than other isoforms of the PI3K. family. The term "dual ΡΙ3 ό7β selective inhibitor" generally refers to a compound that inhibits the activity of both PI3K5 and ΡΙ3Κβ isoforms more effectively titan other isoforms of the PI3K family (t?.g„ PI3 a or γ).

[00121] 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. In one embodiment, the efficacy of a compound as an inhibitor of one or more PI3K isoforms can be measured by the compound concentration that inhibits 50% of the activity in a biochemical assay, i.e., the 50% inhibitory concentration or "ICso" The determination of ICso values can be accomplished using conventional techniques known in the art, including the techniques described in the Examples below. In general, an ICjo can be determined by measuring the activity of a given enzyme in the presence of a range of concentrations of the compound under the study. The experimentally obtained values of enzyme activity may then be plotted against the compound 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 ICso value. Analogously, other inhibitory concentrations can be defined through appropriate determinations of activity. For example, in some settings it may be desirable to establish a 90% inhibitory concentration, i.e., ICOQ.

{00122] According to the present application, a ΡΒΚβ selective inhibitor is a compound that exhibits a 50% inhibitory concentration (ICso) with respect to ΡΙ3 β that is at least 10-fold, at least 20-fold, at least 30-fold, at least 50-fold, at least 100-fold, at least 200-fold, or at least 500- fold lower than the IC50 with respect to either PI3Ka or ΡΙ3Κγ or both Ka and ΡΙ3Κγ. In addition, a ΡΙ3Κ67β selective inhibitor is a compound that exhibits a 50% inhibitory

concentration (ICso) with respect to Ρ13 β and PI3 5 that is at least 10-fold, at least 20-fold, at least 30-fold, at least 50-fold, at least 75-fold, at least 100-fold, at least 200-fold, and at least 500-fold lower than the ICso with respect to either PD a or ΡΙ3Κγ. The dual Ρ13 δ/β selective inhibitor may have the same or similar ICso to both PI3 5 and ΡΙ3 β or may have different ICso to either PI3K5 or ΡΙ3 β. As used herein, the term "potency," "potent,*' or variants thereof refer to the compound exhibiting an IC$o value that is less than 100 nM. When comparing two compounds, the compound that exhibits a lower ICso value is referred to as a more potent inhibitor.

(00123) The compounds of the present application exhibit unexpected selectivity to ΡΙ3Κβ. As shown in the example, certain compounds in Table 1 exhibit low ICso values (e.g. 1 to 100 nM) to both ΡΙ3Κβ and ΡΙ3 δ. Certain compounds in Table la also exhibited such selectivity to PI3K. isoforms. Also, certain compounds of formula (1) exhibited at least between 10-fold to 400-fold lower IC50 values for ΡΙ3Κβ than Ρ13 γ, suggesting the compounds exhibit more selectivity to ΡΙ3Κβ compared to Ρ13 γ (i.e., inhibits the activity of the ΡΪ3Κβ isoform more

42 effectively than die ΡΙ3Κγ isoform as shown by the ΡΙ3Κγ/ΡΒ β ratio). Moreover, the compounds described herein exhibit selectivity to both ΡΙ3Κβ and PI3 6. The compound (S)- 2,4-diamin0-6-((l-(5-chloro-4H3xo-3-pheny

yl)etbyl)ammo)pyrimi<iine-5-carbonitrile, described in US Provisional Application No.

61/745,437, exhibited less selectivity to ΡΙ3Κγ (e.g. the PDKy/ΡΒΚβ ratio is less than 1-fold). The results of the present application suggest that the compounds described herein are dual selective inhibitors of PI3 6 and ΡΙ3 β and exhibit more selectivity to ΡΙ3Κβ compared to ΡΙ3Κγ. Each of the patents and the patent applications described in the present application are incorporated herein by the entirety.

{00124] The methods described herein may be applied to cell populations in vivo or ex vivo. "/« vivo" means within a living individual, as within an animal or human, in this context, the methods described herein may be used therapeutically in an individual. "£x vivo" means outside of a living individual. Examples of ex vivo cell populations include in vitro cell cultures and biological samples including 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, urine, and saliva. Exemplary tissue samples include tumors and biopsies thereof. In this context, the compounds may be used for a variety of purposes, including therapeutic and experimental purposes. For example, it may be used ex vivo to determine the optimal schedule and or dosing of administration of a PI3 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. The compounds of the formula described herein or a pharmaceutically acceptable salt, prodrug, or solvate thereof, may be further characterized to examine the safety or tolerance dosage in human or non-human subjects. Such properties may be examined using commonly known methods to those skilled in the art

(00125) Compared to other P13K isoforras, PI3K6 is generally expressed in hematopoietic cells. Also, ΡΙ3Κβ is generally mis-regulated in certain cancer cells. Aberrant proliferation of cells often interferes with normal tissue function, which may result in abnormal cellular response such as immunity, inflammation, and/or apoptosis. The selective inhibitors to PI3K5 and/or ΡΙ3Κβ are useful in treating, inhibiting, or preventing aberrant proliferation of cancerous and or hematopoietic cells and ameliorating die symptoms and secondary conditions.

43 (00126) The compounds described herein may be used to treat subjects having various disease states, disorders, and conditions (also collectively referred to as "indications") associated with PI3K isoforms or their activities. As used herein, the terms "diseases," "disorders," "conditions" are used interchangeably. Such indications may include, for example, cancer, including hematologic malignancies (e.g. leukemias and lymphomas, myeloproliferative disorders, myelodysplastic syndromes, plasma cell neoplasms) and solid tumors, inflammation, fibrosis, allergic conditions (including hypersensitivity), cardiovascular diseases,

neurodegenerative diseases, renal disorders, viral infections, obesity, and autoimmune diseases.

(00127] In other embodiments, the compounds described herein may be used to treat cancers mat are mediated by, dependent on, or associated with PDK activity. In certain embodiments, the disease or condition is an autoimmune disease, an inflammatory disease, or a cancer. In some embodiments, the disease or condition is chosen from rheumatoid arthritis, osteoarthritis, atherosclerosis, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, asthma, chronic obstructive airways disease, pneumonitis, dermatitis, alopecia, nephritis, vasculitis, atherosclerosis, Alzheimer's disease, hepatitis, primary biliary cirrhosis, sclerosing cholangitis, diabetes (including type I diabetes), acute rejection of transplanted organs, lymphomas, multiple myelomas, leukemias, neoplasms and solid tumors.

(00128) In other embodiments, the disease is a solid tumor. By way of examples, the solid tumor includes but is not limited to pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, rectum cancer, liver cancer, kidney cancer, stomach cancer, skin cancer, gastric cancer, esophageal cancer, head and neck cancer, melanoma, neuroendocrine cancers, CNS cancers (e.g., neuroblastoma), brain tumors (e.g., glioma, anaplastic oligodendroglioma, adult glioblastoma multiforme, and adult anaplastic astrocytoma), bone cancer, or soft tissue sarcoma. In some embodiments, the solid tumor is non-small cell lung cancer, small-cell lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, pancreatic cancer, prostate cancer, or breast cancer.

[00129] The present application also provides a method for treating a human in need thereof, who has or is suspected of having a disease or condition responsive or believed to be responsive to the inhibition of PI3K6 and/or ΡΪ3Κβ activity by administering to the subject a compound of the formulae described herein or a pharmaceutically acceptable salt, enantiomer, atropisomer, tautomer, prodrug, or solvate thereof.

44 {00130] Additionally, the application provides a method of inhibiting kinase activity of a PI3K6 and/or ΡΙ3Κβ polypeptides by contacting the polypeptides with a compound of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof.

[00131 ) Moreover, the application provides a method of decreasing cell viability, increasing cell death or apoptosis, increasing interference with P13K signaling pathways (including AKT, S6RP, ERK phosphorylation), and/or reduction in chemokine production with an effective amount of a compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof.

[00132] The application further provides a method of disrupting leukocyte function comprising contacting the leukocytes with an effective amount of a compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof, in a human in need thereof.

(00133) Provided is also a method of inhibiting growth or proliferation of cancer cells comprising contacting the cancer cells with an effective amount of a compound of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, solvate, or a mixture thereof.

Kits

[00134] Provided herein are also kits that include a compound of the formulae of the present application or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, and suitable packaging. In one embodiment, a kit further includes instructions for use. In one aspect, a kit includes a compound of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, and a label and or instructions for use of the compounds in the treatment of the indications, including the diseases or conditions, described herein.

[00135] Provided herein are also articles of manufacture that include a compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, in a suitable container. The container may be a vial, jar, ampoule, preloaded syringe, and intravenous bag.

45 Pharmaceutical Compositions and Modes of Administration

[00136] Compounds provided herein are usually administered in the form of pharmaceutical compositions. Thus, provides herein are also pharmaceutical compositions that contain one or more of the compounds of any of the formulae disclosed herein or a pharmaceutically acceptable salt, isomers, prodrug, or solvate thereof, and one or more pharmaceutically acceptable vehicles selected from carriers, adjuvants and excipients. Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants. Such compositions are prepared in a manner well known in the pharmaceutical art. See, e.g.,

Remington's Pharmaceutical Sciences, Mace Publishing Co., Philadelphia, Pa. 17th fid. (19SS); and Modern Pharmaceutics, Marcel Dekker, Inc.3rd Ed. (G.S. Banker & C.T. Rhodes, Eds.).

[00137] The pharmaceutical compositions may be administered in either single or multiple doses. The pharmaceutical composition may be administered by various methods including, for example, rectal, buccal, intranasal, and transdermal routes. In certain embodiments, the pharmaceutical composition may be administered by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant In some embodiments, the pharmaceutical composition is administered orally.

[00138] One mode for administration is parenteral, for example, by injection. The forms in which the pharmaceutical compositions described herein may be incorporated for administration by injection include, for example, aqueous or oil suspensions, or emulsions, with sesame oil, com oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.

[00139] Oral administration may be another route for administration of the compounds described herein. Administration may be via, for example, capsule or enteric coated tablets. In making the pharmaceutical compositions that include at least one compound of any of the formulae described herein or a pharmaceutically acceptable salt, prodrug, or solvate thereof, the active ingredient is usually diluted by an excipient and/or enclosed within such a carrier mat can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups,

46 aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 0% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders. In certain embodiments, the pharmaceutical composition is in the form of tablets.

[00140} Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents;

preserving agents such as methyl and propylhydroxy-benzoates; sweetening agents; and flavoring agents.

{00141] The compositions that include at least one compound of any of the formulae described herein or a pharmaceutically acceptable salt, isomer, prodrug, or solvate thereof, can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the subject by employing procedures known in the art Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Patent Nos.3,845,770; 4,326,525; 4,902,514; and 5,616,345. Another formulation for use in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds described herein in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, eg., U.S. Patent Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.

[00142] For preparing solid compositions such as tablets, the principal active ingredient may be mixed with a pharmaceutical excipient to form a solid prefbrmulation composition containing a homogeneous mixture of a compound of any of the above formulae or a pharmaceutically acceptable salt, prodrug, or solvate thereof. When referring to these preformulation compositions as homogeneous, the active ingredient may be dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.

47 [00143] The tablets or pi!ls of the compounds described herein may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action, or to protect from the acid conditions of the stomach. For example, the tablet or pill can include an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.

{00144] Compositions for inhalation or insufflation may include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. In other embodiments, compositions in

pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a facemask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.

Dosing

{00145] The specific dose level of a compound of the formulae described herein for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease in the subject undergoing therapy. For example, a dosage may be expressed as a number of milligrams of a compound of the formula per kilogram of the subject's body weight (mg/kg). Dosages of between about 0.01 and 200 mg/kg may be appropriate. In some embodiments, about 0.01 and 150 mg kg may be appropriate. In other embodiments a dosage of between 0.0S and 100 mg kg may be appropriate. Noimalizing according to the subject's body weight is particularly useful when adjusting dosages between subjects of widely disparate size, such as occurs when using the drug in both children and adult humans or when converting an effective dosage in a non-human subject such as dog to a dosage suitable for a human subject

48 100146] The daily dosage may also be described as a total amount of a compound of the formulae administered per dose or per day. Daily dosage of a compound may be between about 1 mg and 2,000 mg, between about 1,000 to 2,000 mg day, between about 1 to 1,000 mg/day, between about I to 500 mgday, between about 100 to 150 mgday, between about 1 to 100 mgday, between about between about 1 to 50 mg/day, between about 50 to 100 mgday, between about 100 to 125 mg/day, between about 100 to 150 mg/day, between about 00 to 175 mg/day, between about 100 to 200 mg day, between about 00 to 225 mg/day, between about 100 to 250 mg day, between about 100 to 350 mg/day, between about 100 to 400 mg day, between about 100 to 450 mg/day, or between about 100 to 500 mgday.

[00147] When administered orally, the total daily dosage for a human subject may be between 1 mg and 1,000 mg/day, between about 1 to 100 mg/day, between about 1 to 50 mg/day, between about 50 to 100 mg/day, between 100 to 200 mg/day, between about 200 to 300 mg/day, between about 300 to 400 mg day, between about 400 to 500 mg/day, between about 100 to 150 mg day, between about 50 to 200 mg day, between about 200 to 250 mg/day, between about 75 to 150 mgday, or between about 50 to 300 mg/day.

[00148] The compounds of the present application or the compositions thereof may be administered once, twice, three, or four times daily, using any suitable mode described above. Also, administration or treatment with the compounds according to any of the formulae described herein may be continued for a number of days; for example, commonly treatment would continue for at least 7 days, 14 days, or 28 days, for one cycle of treatment. In some treatment, the compound or the composition thereof is administered continuously, i.e. every day. Treatment cycles are well known in cancer chemotherapy, and are frequently alternated with resting periods of about 1 to 28 days, commonly about 7 days or about 14 days, between cycles. The treatment cycles, in other embodiments, may also be continuous.

[00149] In a particular embodiment, the method comprises administering to the subject an initial daily dose of about 1 to 500 mg of a compound of the above formula and increasing die dose by increments until clinical efficacy is achieved, increments of about 1 , 5, 10, 25, 50, 75, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.

49 Synthesis of the Compounds

(00150) The compounds of the present application may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent given the disclosure herein and methods well known in the art Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of typical compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aidrich or other chemical suppliers. In general, compounds described herein are typically stable and isolatable at room temperature and pressure.

General Synthesis

[00151] Typical embodiments of compounds described herein may be synthesized using the general reaction schemes described below. It will be apparent given the description herein that the general schemes may be altered by substitution of the starting materials with other materials having similar structures to result in products that are correspondingly different. Descriptions of syntheses follow to provide numerous examples of how the starting materials may vary to provide corresponding products. Given a desired product for which the substituent groups are defined, the necessary starting materials generally may be determined by inspection. Starting materials are typically obtained from commercial sources or synthesized using published methods. For synthesizing compounds which are embodiments described in the present disclosure, inspection of the structure of the compound to be synthesized will provide the identity of each substituent group. The identity of the final product will generally render apparent the identity of the necessary starting materials by a simple process of inspection, given the examples herein.

Synthetic Reaction Parameters

{00152} The terms "solvent", "inert organic solvent", or "inert solvent" refer to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran ("THF"), dimethylformamide ("DMF'), chloroform, methylene chloride (or dichloromethane), diethyl ether, methanol, and the like). Unless specified to the contrary, the solvents used in the reactions of the present invention are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen. (00153] The compounds of formula (J) may be prepared using the method shown in Reaction Scheme I. The compounds of formula (I) may be prepared using the method shown in Reaction Scheme 1, wherein R4 is optionally substituted pyrimidine.

Reaction Scheme I

o

NH 2

la

Step 1 ··· Preparation of a compound of formula (1 )

[00157} The compound of formula (1) can be made by combining compounds (A), (B) and (C) in the presence of a dehydrating agent. Compounds (A), (B) and (C) are commercially available or can be made by methods known in the art. With respect to compound (A), R 1 is as defined herein. With respect to compound (B), R 3 and R 5 is as de ined herein. With respect to compound (Q, R 2 is as defined herein. Compound (A) can be mixed with Compound (B) in the presence of a coupling agent such as diphenyl phosphite in a solvent such as pyridine. After stirring at a temperature between ambient and 100°C for between 1 and 5 hours, compound (C) is added. After further stirring at a temperature between ambient and 100°C for between 5 and

52 24 hows, the reaction mixture is allowed to cooi to room temperature. To extract the compound of formula (1), an organic solvent such as ethyl acetate (EtOAc) may be added, followed by washing with, mild acid, water, and brine. The organic phase can be concentrated to obtain the compound of formula (1 ). Alternatively, the residue may be purified directly without an aqueous work-up. The compound of formula (I) may be purified by any suitable methods known in the art, such as chromatography on silica gel. Alternatively, the compound of formula (1) may be used in the next step without purification.

Step 2— Preparation of a compound of formula (2)

(001581 The compound of formula (2) can be made by removing the protecting group(s) from the compound of formula (1). The compound of formula (1) is dissolved in a suitable solvent and treated with a suitable acid. Suitable solvents may include, for example, dichloromethane, dioxane, or other suitable solvents. Suitable acids may include, for example, trifluoroacetic acid, hydrochloric acid, or boron tribromide (BBrj). The reaction can be carried out at temperatures between -78°C to ambient temperature. On reaction completion, solvent is removed to obtain the compound of formula (2). In the case of a reaction using BBr 3 the reaction may first be treated with eOH before an aqueous work-up to obtain a compound of formula (2).

Step 4— Preparation of a compound of formula (3)

(00159) The compound of formula (3) can be made by treating S-substituted-2,4,6- trihalopyiirnidine with ammonium hydroxide in a suitable solvent such as dioxane, where the halo is either chloro or fluoro. The reaction is carried out at an elevated temperature between 30 and 80 °C for a suitable time, typically between 2 and 8 hours or when the reaction is complete. Upon completion, water is added to the cooled solution, and the precipitate is collected by filtration. The nitrile can be converted to the carboxamide under standard conditions.

Step 5— Preparation of a compound of formula (I)

[00160] The compound of formula (la) can generally be prepared by coupling compound of formula (3) and compound of formula (2) in the presence of a suitable base in a suitable solvent An example of a suitable base is diisopropylemylamine. An example of a suitable solvent is N- methylpyrrolidone (NMP). The reaction is typically performed at a temperature between 50°C to 150°C for about 30 minutes to 24 hours. Alternatively the reaction can be performed in a microwave at a temperature between i00°C to 150°C for about 30 minutes to 24 hours. Water

53 can be added to quench the reaction upon completion, and the precipitate may be filtered and then dissolved in an organic solvent such as dichloromethane (DCM). The product can be isolated by methods known in the art, for example by removal of solvent under reduced pressure. The product can be purified using any suitable methods known in the art, for example, chromatography of the residue on a silica column. Furthermore, compounds of formula (I) may be prepared by coupling compounds of formula (2) with appropriately substituted heterocycles of the general formula R ~X in a similar manner.

{00161] After synthesis, the compounds may be isolated in the form of a free base or a salt (which includes and is not limited to a hydrochloric acid salt form or a trifluoroacetic acid salt form) and characterized by NMR. Thus, the resulting compounds and their NMR

characterizations may be either the free base or salt The ratio of parent and corresponding salt is not determined.

Example 1. Preparation of a compound of formula (1)

A. Preparation of a compound of formula (1) in which n is 2, R 1 is chloro and fluoro, m is 0, R 5 is H, and R 3 is methyl

F

Boc

100162] A mixture of 2-amino-6-chloro-3-fluorobenzoic acid (1.43 g, 7.6 mmol) and Boc-L- alanine (1.7 g, 9.1 mmol) in pyridine (4.9 mL, 60.5 mmol) was warmed to 45°C until homogeneous then allowed to cool to room temperature, at which time diphenyl phosphite (S.OmL, 26 mmol) was added. The mixture was stirred for one hour at 45 °C, then treated with pyrazme-2,5-diamine bis HC1 (lg, 9.1 mmol) in a single portion. The mixture was stirred overnight at 55°C. After cooling to room temperature, the mixture was diluted with toluene (20 mL) and washed three times with 10 % aqueous hydrochloric acid solution, and concentrated to dryness under reduced pressure. The residue was chromatographed, using a 25 g SiliaSep flash column, eluting with hexanes to 65 % ethyl acetate. The combined fractions were concentrated under reduced pressure to give (S)-tert-butyl (l-(3-(5-arninopyrazin-2-yl)-5-chloro-8-fluoro-4- oxo-3,4-dmydroqumazolm«2<yl)emy1)carbamate. ES/MS 435.1 (M+H*).

54 B. Preparation of the below compounds of Formula (1) using the procedures described in Example 1 A and Reaction Scheme I:

(SMert4>utyI (l-(3 5-amiiK>p r^

yl)ethyl)carbamate;

(S)-tert-butyl (H3~(5-aminopyrazm-2-yl)-5-c^^

yl)ethyl)carbamate;

(S)-tert-butyl ((3^5-aminopyra2in-2-yl)-5-cWoK>-4^^

ylXcyclopropyl)methyl)carbamate;

(SHert-butyl (l-(3^5-aminopyrazk-2-yl^

yl)ethyl)carbamate;

(S)-tert-butyl (l-(3-(5-arainopyrazin-2-yl)-8-chloro-6-flu^

yl)ethyl)carbamate;

(S)-tert-butyl ((3-(5-aminopyrazin-2-yl)-5,8^ichloro-4^xo-3 > 4- lihydroquinazolin

ylXcyclopropyl)methyl)carbamate; and

(S)-tert-butyl ( 1 -(3-(5-annnopyrazin-2-yl)-5,8-dicMoro-4^

yi)ethyl)carbamate.

Example 2. Preparation of a compound of formula (2)

A. Preparation of a compound of formula (2) in which n is 2, R 1 is chloro and fluoro, m is 0, R 5 is H, and R J is methyl.

F NH 2

(00163] A solution of (give (S tert-butyi (l-(3-(5-anunc^yra2in-2-yl)-5^riioro-8-fluoro-4- oxo-3,4^uhydroqumazolin-2-yl)ewyl)c^amate (0.4g, 0.92 mmol) in dichloromemane (10 mL) was treated with trifluoroacetic acid (0.7 mL). After stirring 2h at room temperature, the mixture was concentrated to dryness under reduced pressure to give (S)-2-( 1 -aminoethyl)-3-(5-

55 amiiwpyrazin-2-yl)-5-cbloro-8-fluoroquinazolin^3H)-onc as a golden amorphous semi-solid, which was carried forward without farther purification. ES/MS 335.1 (M+H 4 ).

B. Preparation of the below compounds of Formula (2) using the procedure described in Example 2A and Reaction Scheme I:

(S>2-(l-aminoemyl)-3-(5-amirK>pyr^

(S)-2-(l-am oe yl)-3-(5-aminopyrazin-2-yl^

(S>-2-(amino(cyc!opropyl)methyl)-3-(5-aminopyrazin-2-yl)- 5-cWoroqui

(S>2^1~aminoe y_)-3^5-ammopyra^

(S)-2^1-amirK>emyl)-3 5-amm^^

(S)-2-<arnmo(cyclc^ropyl)meAyl)-3-(5-aminopyrazm-2^

and

(S)-2-( 1 -am oe&yl>3 5-aiimiopyr^^

Example 3. Preparation of a compound of formula (3)

A. Preparation of a compound of formula (3) in which R 4 is CN and X is CI (2,4-diamino-6- chIoropyrirmome-5-carDonitrile)

NH 2

(00164) Ammonium hydroxide (20 mL) was added to a solution of 2 ,4,6-tric&oropyrimidine- 5-carbonitrile (5.0 g, 24 mmol) in dioxane (20 mL) at room temperature. The solution was warmed to 50°C and stirred for 3 hrs. The reaction mixture was cooled to 10°C and water (50 mL) was added. The resulting solid was filtered, washed with water, and dried under high vacuum to afford the title compound as a white solid. (3 H NMR (100 MHz, DMSO) 164.8, 162.6, 161.9, 115.8, 77.6. ES/MS m/z - 169.9 (M+H) + .

B. Preparation of the below compounds of Formula (3) using the procedures described in Example 3A and Reaction Scheme I:

5-c oro-6^fluoropyrimidine-2,4-dianiine;

6-chloro-5<memylsulfonyl)pyritnidme-2,4^iamine;

56 6-chioro-5-(trifluoromemyl)pyrirm^ and

2,4^iamino-6-chloropyrimidm^

Example 4. Preparation of a compound of formula (I)

A. Preparation of a Compound of Formula (I) in which n is 2, R 1 is chloro and fluoro, m is 0 » R 5 is H, and R 3 is methyl, which is (S)-3-(5-ammopyra-dn-2-yl)-5-cWor^

chloropyrir a -4-y})ami^ (Compound 1).

{00165] 5-chloro^fluoropyrimidme-2, -diainine (0.11 g, 0.69 mmol) and DIEA (0.4 mL, 2.3 mmol) were added to a solution of (S 2-(l-aminoethyl)-3-(5-aminopyrazin-2-yl)-5-chlotx>- 8-fluoroquinazolin-4(3H)-one (0.15 g, 0.46 mmol) in IPA. The resulting mixture was heated to 120 °C for 4 h in a microwave then concentrated. HPLC purification of the residue afforded the title compound. Ή MR (400 MHz, DMSO-</ 6 ) δ 8.01 (s, 1 H), 7.85 - 7.71 (m, 3H), 7.63 (dd, J - 8.7, 4.5 Hz, 1H), 7.54 (s, 2H), 7.43 (s, 3H), 6.86 (s, 2H), 5.01 - 4.91 (m, 1H), 1.43 (d, J » 6.5 Hz, 3H). ES/MS 477.1 (M+H + ).

B. Preparation of the below compound of Formula (I), using the procedures described Example 4A and Reaction Scheme I:

(S)-2,4-diamino-6-((l-(3-(5-am opyraa^

2-yl)emyl)ammo)pyrimi<!ir^5^arbonitri (Compound 2). Ή NMR (400 MHz, DMSO-d*) δ 8.20 - 7.92 (m, 4H), 7.92 - 7.73 (m, 3H), 7.71 - 7.56 (m, 2H), 7.49 - 7.35 (m, 1H), 6.87 (s, 2H), 5.01 (q, J~ 6.8 Hz, 1H), 1.42 (d, J~ 6.7 Hz, 3H). ES MS 468.1 (M+H) +

(S)^aniino^-((l-(3-(5-arnmopyr^

yl)emyl)ammo)pyrimidme-5-carbonitrile (Compound 3). Ή NMR (400 MHz, DMSO-tfe) δ 8.54 (s, 1H), 8.09 - 8.02 (m, 2H), 7.94 (s, 1 H), 7.84 - 7.71 (m, 2H), 7.60 (dd, » 8.8, 4.3 Hz, 1H), 7.44 (s, 2H), 6.86 (s, 2H), 4.85 ~ 4.80 (m, 1H), 1.42 (d, J« 7.4 Hz, 3H). ES/MS 453.1 (M+H) + ;

(S>-2^ J -((3Η-Π ,2 ]triazolo[4,5^]pyrimidn-7-yl)amm

fluoroquina2olm-4(3H)-one (Compound 4). Ή NMR (400 MHz, DMSC ) δ 9.51 (s, 1H), 8.41 (m, 3H), 7.93 (t, J - 9.1 Hz, 1H), 7.74 - 7.64 (m, 1H), 7.53 (td, J - 8.3, 4.7 Hz, 1H), 7.46 - 7.17 (m, 4H), 4.86 -4.68 (m, 1H), 1.66 (d, J- 6.8 Hz, 3H). ES MS 420.1 (M+H) + ;

57 (S 3^5-aminop ) razin-2-yl ^

jdamino)eUiyl)quinazolin-4(3H)-one (Compound 5). l H NM (400 MHz, DMSC fc) δ 8.51 (s, IH), 8.17 - 8.10 (m, 2H), 8.03 (s, IH), 7.91 - 7.67 (m, 3H), 7.58 (m, IH), 7.25 (m, 2H), 7.08 (s, lK), 4.78 (ra, lH), 1.52 (d,./= 6.8 Hz, 3H). ES/MS 453.1 (M+H 4 );

(S)-2,4-diamino-6-((l-(3-(5-aminopyi^

yl)ethyl)amino)pyrimidinc-5-carix)nitrile (Compound 6). 1H NMR (400 MHz, DMSO-d6) δ 8.18 (s, 1 H), 8.03 (s, 3H), 7.82 (t,7= 8.0 Hz, 2H), 7.65 (ddd,7= 20.3, 8.0, 1.2 Hz, 2H), 7.49 (bra, 1H), 7.02 - 6.71 (bra, 3H), 4.99 (q, J~ 6.8 Hz, IH), 1.45 - 1.38 (m, 3H). ES MS 450.1 (M+H)*;

(S)-3^5-ammopyrazin-2-yl)-5<^

yl)anu∞)e&yl)quinazolin-4(3H)-one (Compound 7). 1 H NMR (400 MHz, DMSO-d6) δ 8.02 (bra, IH), 7.86- 7.77 (m, 3H), 7.69 (dd,/= 8.1, 1.2 Hz, IH), 7.62 (dd, 7= 7.9, .2 Hz, IH), 7.55 (brs, 2H), 7.44 (bra, 2H), .84 (bra, 2H), 4.94 (p, J- 6.8 Hz, IH), 1.42 (d, ~ 6. Hz, 3H). ES/MS 459.1 (M+HT);

(S)-3-(5-aiTimopyra2ia-2-yl)-5-ch1oro-2^cyclopropyl((2

yl)aniino)me4hyl)qumazolm-4(3H)-one (Compound 8). IH NMR (400 MHz, DMSO-d6) δ 7.88 - 7.78 (m, 2H), 7.78 - 7.66 (m, 3H), 7.63 (dd, J « 7.8, 1.2 Hz, IH), 7.56 (s, 2H), 7.41 (s, 2H), 6.82 - 6.73 (m, 2H), 4.74 ~ 4.47 (m, IH), 1.53 (s, IH), 0.57 (s, IH), 0.46 (dp, J « 12.4, 7.3, 6.1 Hz, 2H), 0.16 (dq, J * 9.6, 4.9 Hz, 1 H). ES MS 485.1 (M+H) + ;

(S)-2,4^1iamino-6-(((3-(5-ainmopyr^

yiXcyclopropyl)me hyl)ainin<>)pyrimidine-5-carbonitri (Compound 9). IH NMR (400 MHz, DMSO-d6) 88.14 (d,J = 19.0 Hz, 3H), 7.90 - 7.78 (m, 2H), 7.71 (dc = 8.2, 1.2 Hz, IH), 7.63 (dd, J = 7.8, 1.2 Hz, IH), 7.48 (bra, IH), 6.75 (m, 2H), 5.96 (bra, 2H), 4.69 (t, J= 8.3 Hz, IH), 1.54 (s, IH), 0.57 (s, IH), 0.51 - 0.39 (m, 2H), 0.17 (m, IH). ES/MS 476.1 (M+H) + ;

(S)-2,4-diammo^(((3^5~ammopyr^

ylXcyclopropyl)me&yl)ammo)pyritTU^ (Compound 10). IH NMR (400 MHz,

DMSO-d6) δ 8.07 - 7.97 (m, 5H), 7.91 ~ 7.82 (m, IH), 7.80 (s, IH), 7.63 (d, 7= 8.5 Hz, IH), 7.47 7.41 (m, 2H), 6.85 (s, 2H), 4.82 (t, J « 7.9 Hz, IH), 0.57 0.39 (m, 4H), 0.24 - 0.13 (m,

58 IH). ES/MS 510.1 (M+H ,

(S)-2,4-diannm)-6-((l-(3-(5-aminopyr^

yl)ethyl)amino)pyriraidine-5- arbonitrile (Compound 11). 1 H NMR (400 MHz, DMSO-d6) δ 8.11 - 7.90 (m, 5H), 7.83 (s, 2H), 7.62 (d, J - 8.5 Hz, 1H), 7.54 - 7.26 (m, 2H), 6.90 (s, 2H), 5.07 (p, 6.7 Hz, 1H), 1.43 (d, 7= 6.6 Hz, 3H). ES/MS 484.1 (M+H) + ;

(S)-3-(5-aminopyrazin-2-yl)-5,8- lichloro-2-{cyclopropyi((2,6-<iiami^

yl)ammo)memyl)<niiiu^^ (Compound 12). 1H NMR (400 MHz, DMSO-d6) $

8.52 (d, J= 1.5 Hz, 1H), 8.07 ··· 8.00 (m, 2H), 7.70 (d, 7= 1.5 Hz, IH), 7.63 (d, 7= 8.6 Hz, 1H), 7.49 (d, ~ 8.5 Hz, 1H), 6.89 - 6.81 (in, 3H), 6.45 (s, 2H), 6.15 (d,7 » 5.3 Hz, 1H), 4.81 - 4.76 (m, IH), 1.38 1.12 (m, 1H), 1.06 0.72 (m, 2H), 0.66 0.39 (ra, 2H). ES MS 519.1 (M+H) + ;

(S)-3-(5-aminopyrazin-2-yl)-5,8 iicM^

yl)amino)ethyl)quinazolin-4{3HVone (Compound 13). 1H NMR (400 MHz, DMSC 6) δ 8.02 (dd, 7= 8.6, 0.7 Hz, 2H), 7.84 (d, 7= 8.1 Hz, 2H), 7.67 - 7.56 (m, 3H), 7.56 - 7.46 (m, 2H), 6.95 - 6.87 (m, 3H), 5.01 (p,7= 6.7 Hz, 1H), 1.44 (d,7-- .6 Hz, 3H). ES/MS 493.0 (M+H ,

(S)-2-ammo-4-((l-(3-(5-arainopyrazin-2^

yl)ethyl)amino -6^dtfiuoromemyl)pyrinu 14). ! H NMR (400

MHz, DMSO) 6 8.16 (dd, J - 8.5, 2.9 Hz, 1H), 8.08 (br s, 1H), 7.86 - 7.79 (m, 3H), 7.54 (br a, 1 H), 7.35 (br s, IH), 6.87 (br s, 2H), 6.65 (t, J » 53.5 Hz, 1H), 5.10 - 5.00 (m, 1H), 1.45 (d, J - 6.5 Hz, 3H). ES MS 503.1 (M+lf);

(S)-2,4-Diamino-6-(( 1 -(3-(5-amii^yrazin-2-yl)-6,8-diflw^

yl)ethyl)amino)pyriraidine-5-caibomtrile (Compound 15). *H NMR (400 MHz, DMSO-4 δ 8.00 (s, 3H), 7.92 (ddd, 7 - 10.3, .9, 2.9 Hz, IH), 7.79 (s, 1H), 7.69 (ddd, 7= 8.2, 2.9, 1.3 Hz, 1H), 6.85 (s, 2H), 5.01 (p,7= 6.7 Hz, IH), 1.40 (d,7= 6.6 Hz, 3H). ES MS 468.1 (M+H );

(S>3-(5-AmiiH>pyrazm-2-yl)-2^H^

difluoroquinazolin-4(3H)-one (Compound 16). Ή NMR (400 MHz, DMSO-<4) 67.93 (ddd, 7= 10.4, 8.9, 2.9 Hz, 2H), 7.79 (d, 7 - 7.8 Hz, 2H), 7.69 (ddd, 7= 8.2, 2.9, 1.3 Hz, 1 H), 7.50 (s, 2H), 7.39 (s, 3H), 6.84 (s, 2H), 4.99 (p, 7= 6.7 Hz, IH), 1.41 (d, 7 - 6.6 Hz, 3H). ES/MS 461.9 (M+H * );

59 (S)-2,4-Diamino-6-(( 1 -(3 -{5-aminopyrazin-2-yl)-8-chloro-6-fluoro-4-oxo-3 ,4- dmydioquinazolin-2-yl)e&yl)^ (Compound 17). ! H NMR (400

MHz, DMS<_W 6 ) 68.17 (dd, J - 8.5, 2.9 Hz, 1H), 8.02 (s, 2H), 7.85 (tt, J 7.8, 3.6 Hz, 4H), 6.90 (s, 2H), 5.10 (p, 7- 6.6 Hz, 1H), 1.44 (d, J- .6 Hz, 3H). ES MS 468.1 (M+H + );

(S)-3^5-Aminopyrazm-2-yl 8^

6-fluoroquinazolin-4(3H)-one (Compound 18). Ή NMR (400 MHz, DMSO- ) & 8.19 (dd, J= 8.5, 2.9 Hz, 1 H), 7.98 (s, 2H), 7.86 (dd, 7 - 8.1, 2.9 Hz, 2H), 7.81 (s, 1H), 7.56 (s, 4H), 6.90 (s, 2H), 5.23 ··· 4.91 (m, 1H), 1.45 (d, J°° 6.6 Hz, 3H). ES/MS 477.1 (M+H*);

(S)-2-(l-((6-Amino-5-chloropyriim

chioroquinazolin-4(3H)-one (Compound 19). Ή NMR (400 MHz, DMSO-cfe) δ 8.73 (s, 1H), 8.18 (d,J « 1.4 Hz, lH), 7.92 (s, 1H), 7.89 (<_,./ « 1.5 Hz, 1H), 7.67 - 7.60 (m, 1H), 7.49 (dd,7

- 8.2, 1.2 Hz, 1H), 7.44 (dd,./ - 7.8, 1.2 Hz, 1H), 7.18 (s, 2H), 4.75 (s, 1H), 1.49 (d, J « 7.0 Hz, 3H). ES/MS 444.1 (M+fT);

(S>2-(((6-Araino-5^Moropyrimidm^yl)^

5,8-dichloroquinazolin-4(3H)-one (Compound 20). Ή NMR (400 MHz, DMSO-< 6 ) 58.01 (d, J

- 8.5 Hz, IH), 7.82 (s, 1H), 7.71 (s, 1H), 7.60 (d, 7= 8.5 Hz, 1H), 6.87 (s, 2H), 6.62 (d, 7= 17.2 Hz, 3H), 4.67 (s, 1H), 1.42 (s, 1H), 0.51 - 0.27 (m, 4H), 0.27 - 0.07 (m, 1H). ES MS 504.1 (M+H 4 ); and

(S>2^((6-Anuno-5-chloropyrimidin-4-yl)amino

5-cWotoquinazo1in-4(3H)-one (Compound 21). 5 H NMR (400 MHz, DMSC fc) δ 7.96 (s, IH), 7.81 (q, J- 7.9 Hz, 2H), 7.69 (d, 7 - 9.3 Hz, 3H), 7.60 (dd, 7= 7.9, 1.3 Hz, 2H), 6.81 (s, 3H), 6.61 (s, 4H), 4.37 (d,7 « 12.5 Hz, IH), 1.49 (d,7= 12.0 Hz, IH), 0.44 (s, 4H), 0.34 (q, 7 s 4.5 Hz, IH). ES/MS 470.1 (M+H ).

Biological Examples

(00166] The compounds of formula (I) were characterized for their enzymatic activity against the PI3K isoforms. The activities were measured using a time-resolved fluorescence resonance energy transfer (TR-FRET) assay. TR-FRET monitored the formation of 3,4,5-inositoI triphosphate molecule that competed with fluorescently labeled PIP3 for binding to the GRP-1 pleckstrin homology domain protein. An increase in phosphatidylinositide 3-phosphate product resulted in a decrease in TR-FRET signal as the labeled fluorophore was displaced from the GRP-1 protein binding site.

{00167) Class I PDK i so forms were expressed and purified as heterodimeric recombinant proteins. All assay reagents and buffers for the TR-FRET assay were purchased from Millipore. PDK isoforms were assayed under initial rate conditions in the presence of 25 mM Hepes (pH 7.4), and 2 Km ATP (75-500 uM), 2 uM PIP2, 5% glycerol, 5 mM Mg ¾, 50 mM NaCl, 0.05% (v/v) Chaps, 1 mM dithiothreitol, and 1% (v/v) DMSO at the following concentrations for each isoform: ΡΒΚα, ΡΠΚβ, and P13K6 between 25 and 50 M, and ΡΒΚγ at 2 nM. The compounds of Table 1 and Compound X ((S)-2,4^iarmno-6-((l-(5^hloro-4-oxc~3-(pyridin-3- yl)-3,4-dihydroquinazolin-2-yl)eftyl)anaino)pyrimi and Compound Y ((S>-

2,4-diammo-6-((l-(5-cUoro-4-oxo-3-(pyria¾-3-yi)-3,4-dmyd r^

yl)ethyl)ammo)pyrimidtne-5-caibonitrile) were added to the assay solution and incubated for 30 minutes at 25°C. The reactions were terminated with a final concentration of 10 mM EDTA, 10 nM )abeled-PIP3, and 35 nM Europium labeled GRP-1 detector protein before reading TR-FRET on an Envision plate reader (Ex: 340 nm; Em: 615/665 nm; 100 μβ delay and 500 μ$ read window).

[00168] The results were normalized based on positive ( 1 μΜ wortmanin) and negative (DMSO) controls, and the ICso values for PDK α, β, δ, and γ were calculated from the fit of the dose-response curves to a four-parameter equation. These assays generally produced results within 3-fold of the reported mean.

(00169] Table 2 summarizes the IC» (nM) values for PI3K isoforms β, δ, and γ. The results indicate that certain compounds of formula (1) inhibit bom PI3K5 and ΡΙ3Κβ. Also, Compound X exhibited PI3K6 IC» of 0.2 nM, ΡΙ3Κβ ICso of 11 nM, ΡΙ3Κγ ICso of 7 nM. The

ΡΙ3Κγ/ΡΙ3Κβ ratio for Compound X is 0.6. The results indicate mat certain compounds have greater selectivity for ΡΙ3Κβ over Ρ13Κγ compared to compound X. Compounds in Table 1 a were analyzed using the same assay, and the results are summarized in Table 2a.

Table 2. The ICso values (nM) for PI3K isoforms β, δ, and γ.

100170] All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description. From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the present application.

62