Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SPECIFIC AKT3 ACTIVATOR AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2019/051063
Kind Code:
A1
Abstract:
Compositions and methods of selectively activating Akt3 are provided.

Inventors:
KHLEIF SAMIR (US)
MKRTICHYAN MIKAYEL (US)
Application Number:
PCT/US2018/049715
Publication Date:
March 14, 2019
Filing Date:
September 06, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV RES INST INC AUGUSTA (US)
International Classes:
A61K31/47; A61K31/4709; A61K31/5377
Foreign References:
US3987102A1976-10-19
US20170202829A12017-07-20
Other References:
SASAKI ET AL.: "Design, Synthesis, and Biological Activity of Potent and Orally Available G Protein- Coupled Receptor 40 Agonists", JOURNAL OF MEDICINAL CHEMISTRY, vol. 54, 14 February 2011 (2011-02-14), pages 1365 - 1378, XP055168132, DOI: doi:10.1021/jm101405t
ALI ET AL.: "Synthesis and Structure-Activity Relationship Studies of HIV-1 Virion Infectivity Factor (Vif) Inhibitors that Block Viral Replication", CHEMMEDCHEM, vol. 7, 3 May 2012 (2012-05-03), pages 1217 - 1229, XP055581697
ATWELL ET AL.: "Potential antitumor agents. 13. Bisquaternary salts", JOURNAL OF MEDICINAL CHEMISTRY, vol. 16, June 1973 (1973-06-01), pages 673 - 678, XP002378754, DOI: doi:10.1021/jm00264a021
See also references of EP 3678666A4
Attorney, Agent or Firm:
VORNDRAN, Charles et al. (US)
Download PDF:
Claims:
We ciaira:

I , A co pound- according to For mu I ;

2, A compound according to Formula Ι.Ϊ ;

A compound accordin to Foimola 111:

Formula HI

or pharnraceutiealJy acceptable enantiomer, salt, or solvate thereof, wlierein: Ri is selected it om -(Ci~Cif>}-a{kyl, ~(Ci-C¾2)~cycioa!kyi; -(G-Cis)- heterocycloalkyi, -(Ce-C^i-aryk or -((¾-C2 )~l¾teroaryi groups optionally substituted by one or more sisbstiroeais selected from -(C -.C s2)- lk i, -(Q-C )- cyoioalkyi -(C3~Ct2)-hetefocycloaiky !, -0~(O-C J2>alk i, -0~(e}-Cr2}~alkyi- (Cs-C20}-aryl5 -0-fCs-C]2)-cycloalkyl5 «S-(Ci~Gi2}-alkyL -S-CCs-C^-cycloalk l, -COO~(C,!-C}2)~8lkyl:> -COO~CC^C¾)~eyeloalkyl; rON i Ci a)-ii1kyi, - CO H-iC5~Ci3)-eye;« -CO-fCs -Ci2)-cyclo l'kyl , . ~M-

(C:j-C:i2)-alky!, <C3-C2o)~¾e|e o yk -(C C2o}-heteri aryi-{Ci~Ci2)-afky!s C2a)- eteroaryI-0-{Ci-Cj2)-aIkyIi -COOH, -OH, -SH, SOiH, -CM, -Nft, or a halogen,

Y5 aiid Z are independently selected from -0? -HB, -S, -K-(C,i alkyl, or ~(Ci~C¾{t)-aryl

Rt is selected from -(C^a«)~a kyi, «0, -OH, -SO2, -SO, or -SOC.H3; and is selected from -(Ci-Ci2}-aikyl? -(Ci-Ci2)-cycloaSkyl, -(C -Cn)- beteroeyx!oaikyi, -0-(Ci-Ct2)-alkyi, ^^-(Ct-Ciil-aik i-CC^-Cso^a l, -0-( C¾2)-eycIoalkyk ~S-(Ci~Ci2)-alk.yk ~S-(C3~Ci2)-cycloaIkyk -COO~(Ci-Ci2)~ Ikyk -COO-(Cs~Ci2)-cycloalkyk -CONH-(Ci~Ci2)-alkyi; ~CONH~(C3-Ci 2)-cyc!fjaiky!;

heteroaryl, -(C3-C2f))-heteroaryl-(Ci-Ci )-aikyL "(C¾-C26)-heterQaryl-0-(Ci-Ci2 alkyl, -COOH, -OH, -SB, ~S ?B? ~CN, ~NH¾ or a Imloge

4. A compound according to Formula IV:

Formula IV or an enaniiomer, polymorph, or pharmaceutically acceptable salt thereof

5 , A method of increasing an immune suppressive response in a subject in: need thereof comprising administering to the subject any one of the compounds of claims 1-4 or an eraantioriier. polymorph, or pharmaceutically acceptable salt thereof that selectively activates A 3 by an amount effective to increase the immune suppressive response in the subject.

6. The method of claim 5 wherein the subject has an inflammatory disorder or disease.

7. A me thod of treating an Inflammatory disorder compr ising

administering to a subject in need thereof a eonrpositioti comprising any one f the compounds of claims 1 -4 or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof that selectively ac tivates Akt3 in an. amount effective to induce, increase, or promote an immune suppressive response in the subject.

8. The .method of any one of claims 5 -7 wherein, the inflammatory disorder or disease is .selected, from the group consisting of rhe umatoi arthritis, systemic htpus erythematosus, alopecia areata, anklosing spondylitis;, anii tospliolipld syndrome, autoimmune Addison's disease, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune

lympiioproliferaiive syndrome (ALPS), antoii m e thrombocytopenic purpura (ATP), Behcet's disease, bullous pemphigoid, cardiomyopathy^ celiac sprue- dermatids, chronic fatigue syndrome immune deficiency, syndrome (CF!DS), chronic inflammatory' demyelinating polyneuropathy, cicatricial pemphigoid, cold agglutinin disease. Crest syndrome, Crohn's disease, Dego's disease, dermatorayositi , dermatomyosi tis - juvenile, discoid Inpus, essential mixed cryoglobulinemia, fibromyalgia - irbroniyositis, Grave's disease, Gdllain- Barre, H¾shimoto%- thyroiditis* kiiopatfcic pulmonary fibrosis, idiopathic

ihro-mb.ocyiopenia purpura (ITP), Iga nephropathy, insulin dependent diabetes (Type I), juvenile arthritis, Meniere's disease, mixed connective- tissue disease, multiple sclerosis, myasthenia gravis, obesity } pemphigus vulgaris^ peruieioits anemia, polyarteritis nodosa, polychondritis, polygiancular syndromes, polymyalgia- rheiuttatiea, polymyositis and det atomyositis, primary agammaglobulinemia, primary biliary cirrhosis, psoriasis, Raynaud's phenomenon, Keller's syndrome, rhetsmatic fever, sarcoidosis, scleroderma, Sjogren's syndrome, s iff-man syndrome, Takayasu arteritis, temporal arteritis/giant cell arteritis, ulcerative colitis, uveitis, vasculitis, itiligo, and Wegener's granulomatosis..

9. A method of tr atin an a utoimrnurie disease comprising administering to a subject in need thereof a composition comprising any one of the compounds of claims 1-4 or an eiimitiomer, polymorph,, or pharmaceutically acceptable salt thereof that selecti vei inhibits A.kt3 in an amount effective to induce, promote, or enhance an immune suppressive response in the subject

10, The method of claim 9, wherein the autoimmune disease is selected from the group consisting of 'rheumatoid artlirips:, systemic lupus erythematosus, alopecia areata, autoimmune Addison's disease, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune

lympkoproliferative syndrome (ALPS), autoimmune thrombocytopenic purpura (ATP), Crohn's disease multiple sclerosis* and myasthenia gravis.

1 L The method of any one of claims' .5-10· wherein the immune suppressive response that is increased is .selected from, the group consisting of an immune suppressive function of natural Treg (nTr g and induction of conventional T cells into induced Treg (iTreg).

1.2. the metho o clai 11 wherein, the .hnmufte suppressive function of uTre is the secretion of one or more :ai)ii-iiiil as¾matory cytokines.

13. The method of claim 12 wherein -the ann-hvflamiiiatoty cytokine is JL 10, <5Fp, r a combination thereof.

14. The method o any of claims 5- 1,3, farther comprising administering to the subject a second kmnunosuppressive agent

15» The method of claim 14, wherein the second active agent is a compound selected from the group consisting of prednisone, budesonide, prednisolone, cyclosporin©, tacrolimus, sirotimiss, everoHmus, azathioprine, lefliraomide, ycophenolate, abaiacept, adaiimumah, anakisra, certoHzumab, etaaercept, goiiniuniab, infliximab, ixekizomab, nataiizumab, rituximab, secoMnomah, iociliztwnab, ustekin mab, vedolizumab, basil mab, daclizmmab, and inirromo!iab.

16, The method of anyone of claims 5-15, wherein any one of the componnds of claims 1-4 is administered m. an ame¾nt to increase FoxP3 expression on immune celis,

17. The method of claim 1.6S wherein the imrnime cells comprise iTregs.

18 , The method of any one claims: 5-1 Ί wher in any. o»e of the eornpoinKfo of clarms 1-4 is administered in an amount effecti ve to increase proliferation; of iTregs, 1 , A pharmaceutical composition comprising any one of the compounds of claims 1-4, or aw enantlomer, pol mor h, of plvarmaceuticajj v acceptable salt thereof and an exeipient.

20. The composition of claim 19, wherein auy one of the compounds of claims 1 - , or an enantlomer;* olym h;, or pliaonaoeuiicaliy acceptable salt thereof is ¾ an arftotint effective to increase a siippresstve Im une response when administered to a s«b ecf in need thereof

21 , A method of Iscreasmg an mmune stippressive response in subject m need there of comprising contacting imniim ceils ex vivo any one of the compounds of claims 1-4, or as enantiomer, polymorph, or pharmaceutically acceptable salt thereof! in an amount effective to increase expression' of FoxP3 on the immune cells, and administering the contacted immune cells to the subject.

22. The ethod of claim- 21, wherein the immune cells comprise autologous immune cells.

23. he method of claim 21 or 22, wherein immune : ce lis comprise T cells,

24, The method of claim 23, wherein the T cells comprise Tregs,

25i The method of claim 24, wherein the Tregs comprising iTregs,

26, A method for inhibiting or reducing transplant rejection in a su bject in need thereof b administering, aa effective amount of any one of the compounds- of claims 1-4 or an esaatiomer, polymorph, or pharmaceutically acceptable salt thereof to increase PoxP3 expressio on immune cells of the subject.

27. A method for ttealing Grail- versns-Bost disease in a subj ec m need. ihei¾offey-¾^it)!isteiin ,tjE> tfee subject an effective amount of y one of the com oun s of claims 1-4 or an enantio-mer, polymorph, or pham¾aceutica]iy acceptable salt thereof to increase FoxP3 expression on immune cell s of the subject

28. A method for treatin ehtrOnic infectio An a subject in need thereof by administering t the subject an. effective amount of any one of the compounds of claims 1-4 or an enantiomer, polymoipb, or pharmaceutically acceptable salt thereof to increase FoxP3 expression on immerse cells of the Sabject

29. A method for treating obesity in a subject in need thereof by

administering to the subjec t an effective amount of any one of the compounds o f claims 1-4 or an enamiomer, polymorph, or pharmaceutically acceptable salt thereof

Description:
SPECIFIC A T3 ACTIVATOR ΑΝΪ> USES THEREOF

CROSS-REFERENCE TO RELATED APPLICATIONS

This applieatios claims benefit o and priority to US. Provisional Application os. 62/555,14.1. filed on September 7, 2017, 62/657,345 filed on April 13. 2018, and 62/659,870 filed on April 19,.2018, which are incorporated by reference in their entirety..

EFER NCE TO S QUE CE LiSTiNG

The Secpenc listing su mitte o September 6, 201.8, as text file nam d "064466,070 sequence lisirog__ST25. txt M created on August 21, 2018, aod having a size of 10.7 kilobytes is hereby incorporated by reference pursuant to 37 CJF.it § l.,S2(e)(5),

FIELD OF BE i VE TiO

The invention is generally directed to compositiens and TOediods for se!ecti ve activation of Akt3 activity, and methods of use thereof for modulating regulator T cells,

B CKG OU D OF TOE INVENTION

Regulatory T cells (Tregs) are a. subset of CD4f T cells that suppress immune .responses -and are essential mediators of se lf- tolerance and immune aonieostasis (Sakaguchi, el al., C , 133, 775-78? (2008)). Depletion or inactlvation. of Tregs results trs. the development of severe antoi mnnity (Sakaguchi, et at,, J, Immunol, 155, 1151-1164 (199.5}), and their accumulation inhibits anti-tumor immunity (Daanull, et at* I ' hc Jwmioi of clinical

mvestig tmti, 115, 3623-3633 (2005)). Tregs are characterized by f¾xp3 expression, a transcription factor belonging to the f orkhead Box family of transcription factors. The Foxp3 is a master regulator of Tregs, as it is necessary for their development ami [ function (Hori, Science, 299, 1057-1061 (2003); Fontenot, et at, . M Immunol, 4(4);330~6 (2003). E iib 2003 Mar 3; Khatiti, et aU Nm Immtif l, 4(4)3Z7 ' 42 (2003). Epuh 2003 Mar 3}.}.

There are two major types of Tregs: tbyrnus-derived Tregs (of natural. Tregs (n Tregs)} that constitute 5-10% of the total peripheral€D4 T cells, and peripheral TGPP-iMdueed Tregs (iTregs). Bot types are shown to have iramunosoppmsive properties mediated via several processes that involve immunosuppressive soluble factors or ceil contact (Biuestone, et a!, s Nat ev Immunol, 3, 253-257 (2003); Glisic, et at. Cell and Tissue Research, 33 , 585- 595 (2010); Hon, Science, 299, 1057-1061 (2003); Sakaguehi C ll, .1.01, 455- 458 (2000); Sakagnski., et at. Curr. Top Microbiol Immunol, 305. 5.1-66 (2006); Sskagushi et at, Immunol, Rev.., 21.2, 8-27 (2006); (Schmidt,, et al,, Front ImnmmL 3:5! (2012);}. However, the molecular mecj^nls s by which iiTreg and iTreg develo arid then exhibit non-redundant roles to suppress the mmunity are not fully understood (Dipica, et at, Immunity, 35(1); 109- 122 (201 !)).

P!3R.-Akt signaling affects many processes and is central to many signaling pathways. Akt phosphorylation and kinase activity ar induced by P13 activation^ which is, in turn, induced, by several growth factor receptors, TCR, CD28, and IL-2:R, among many others (Parry, et al. Trends in

immunology, 28, 161-168 (2007)). in mammals, there are three Akt isoibrmsi namel Akt I , Akt2, and A.kt3, encoded by three independent genes, in vitro these isoforrns appear to have redundant functions, as different extracellular inputs can induce similar Akt signaling patterns (Franke, Science l, pe29~

(2008.)). However, isoforni-specsic ' knockouts show unique features and their involvement in diseases and physiological conditions indifferent (Boland, et al. American Journal of Human Genetics, 81, 292*303 (2007); DeBosch, et at, J, BioL Ckem, 281, 32841-32851 (2006); Erna iati, et t, Nat Genet, 36, 131-137 (2004); Garofaio, et al. The Journal of clinical investigation, 1. 12, 1 7-208 (2003); George, et at. Science, 304, 1325- 1.328 (2004); Nakaiani, et at , the Journal of ' Biological Chemistry, 274, 2 ί 528-21532 (1999); Ts hopp, et al. Development (Cambridge, England), 132, 2943-2954 (2005); Yang, etat,J BioL Chew * 278, 32124-3:2:1.31 (2003)).

Studies have shown that Akt 1 and Aki2 cm negatively regulate the transcriptional signature ofTteg, thereby selectively affecting Tteg lineage differentiatio (Saner, et at, Proceedings of the NationalA - cademy of Sciences, 105, 7797-7802 (2008a)}, Additionally, lthough ft was shown that inhibition of AJcti a«d Akf2 isoforros increase Foxp3 ' expression in TGFp induced iTregs (Saner, et ah, Proa. N iL Acad ScL USA, 105, 7797-7802 (2008b}), the

mechanism remained nnciear. Another finding. -shows that deletion of A 2 resulted in detective iThl 7 cell, differentiation but preserved fiTIii 7 eel! de lopment et al, Nai Immumoi ^ 14(ό):ό! 1.-8 (2013) Epnb 201.3 May 5)„ F rther Akl3 is also expressed m immune cells and the spinal cord of Akt3 knoekont mice have decreased numbers of Foxp3 regulatory T cells compared with. wild, type mice (Tsipersoii, et ai., lmmiif l^ l§©(4)*i528~39 (2013) Epob 20.13 Jan 18)). Th us, ahhoogh some studies have examined the rele vance of Aki isoform expression on T cell biology (Carson, et al , Annals of the New Ywk Amdemyof Sconc s, 1 103, 167-178 (2007) , Crell¾ et al , lood, 109, 2014- 2022 (2007a); Crellin, et at Journal of Immunological Methods, 324, 92-104 (2007b); Haxhmasfo, /. Exp. Med , 205, 565-574 (2008); Li, et af, Blood, 106, 3068-3073 (2005); Patten, et al, Biac em. Soc. Trans. * 35, 167-17! (2007); Patten, et al, J. Immunologic 177, 6598-6602 (2006); Saner, et al, Proc. HatL Acad. Scl USA, 105, 7797-7802 (2008b); Walsh, et al,, J, Qin. Invest, 1 16, 2521 -2531. (2006)), the roles that Akt isoforms play hi Treg function and induction was not clear.

Therefore, it is an object of the invention to provide compounds and compositions for selectively activating: Akt3 in i mune cells.

It is another object of the invention to provide methods of decreasing an immune response in a subject

Still another object of the invention, is to provide methods of increasing a suppressive mmune response in a subject.

SUMMARY OF THE INVENTION

•Compositions and methods of selectively activating Akt3 are provided. One embodiment provides a compound according to Formula I;

Formula ' ϊ

or a pharmaceutically ' acceptable enatiiionier, sal t * of sol vate thereof, wherein;

rings A, B ; and C are indepen entl slx-membered aiyl or -eoiJiainlng heteroaryl rrtono- or bicyciie .ring s stems eo aming zero or more M~aioms such as .phenyl, .pyridine, pyrirnidiae- pyridazioe, pyraziae, triazme. quino!am quinazoline, isOquinohne* naphthalene, . naphthyridine, indole, isoindo!e,

halogen.

Another embodiment provides a compound according to Formula II

Auotte emfeodimeai provides a com cvuud accordin to Formula IB;

Another embodimeiit provides compound according to ormula IVr

one or more anti -inflammatory cytokines. The anti-mflammatory cytokineis) can IL10, TGFfl or a combination thereof

in some embodiments, the subject has an autoimmune disease.

Therefore, methods of treating autoimmune diseases by administering to subject in need thereof an effective amount of a compound according to Formula 1, Formula II, Formula Ιίί, or Formula IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof thai induces or increases the

bioavailability or bioactivity of Ak*3. are also disclosed.

Combination therapies including modulators of Akt3 bioaeiivity and methods of use thereof are also provided.

S One embodiment provides a ..method of increasing an immune

suppressi ve response in a subject in need thereof by administering to the subject a composition containing an effective amount of a compound according to Formula I, Formula II, Formula HI, or Formula. IV, or an enantiorne , polymorph, or pharmaceutically acceptable salt thereof that selectivel activates Ak& b an amount effective to increase the immune suppressive response i the subject The subject can have mi inflammato y disorder or disease, for example an autoimmune disease .

Another embodiment provides a method of ' treating an infl mmat y disorder in a subject i need thereof b adnimiste-rmg a composition comprising a compound according to- ormula 1, Formula II, Formula IIS, or Formula IV, or an enaittiomer, polymorph., or lmrraaceuticaib' acceptable salt: thereof that selectively activates Akt3 in as amount effective to increase, induce, or promote an. immune suppressive response n the subject,

In some embodiments, the inflammatory disorder or disease is selected from the group consisting of rheumatoid arthritis, systemic lupus erythematosus, alopecia areata, ankylosing spondylitis, antip osphoiipid syndrome,

autoimmune Addison's disease, autoimmune hemolytic- anemia, autoimmune hepatitis, autoimmune inner eat disease, autoimmune K-TOphoprolifeiative syndrome (ALPS), autoimmune thrombocytopenic purpura (ATP), Behcet's disease, bullous pemphigoid, -cardiomyopathy, -celiac sprae-detffistiiis, chronic fatigue syndrome immune deficiency, syndrome (CFIDS), chronic inflammatory derayefinating polyneuropathy s cicatricial pemphigoid* cold agglutinin disease. Crest syndrome, Crohn's disease, Degp*s disease, dennatonwesitis,

dermatomyosttfs - juvenile, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia - tibromyositis, Grave's disease, Guillain-Barre, Hashimoto's- thyroiditis, idiopathic pulmonary fibrosis, idiopathic thrombocytopenia purpura. (FTP), . Iga .nephropathy. Insulin dependen diabetes (Type 1), juvenile arthritis, Meniere's disease, mixed connecti e tissue disease, multiple sclerosis, myasthenia gravis, obesity , pemphigus vulgaris, pernicious anemia,, polyarterit s nodosa, polychondritis, polygkncular syndromes, polymyalgia rheumatica,

rheumatoid arthritis, systemic lupus erythematosus, alopecia areata, autoim une Addison's disease, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune tymphoproKferative syndrome

(ALPS), autoimmune thrombocytopenic purpura (ATP), Crohn's disease multiple sclerosis, and myasthenia gravis,

in some embodiments, the immune suppressive response that is increased is selected from the group consisting of as. immune suppressive function of natural Treg (nTreg and induc tion of conventional T cells into induced Treg (iTreg , The immune .suppressive taction of nTreg can. include the secretion of one or more anti-inflammatory cytokines, for example It 10, TGFp, or combination thereof

Another embodiment provides a method of treating a subject in need thereof by administering m effective amount of a composition con ain ng a compound according to Formula L Formula II, Formula III, or Formula IV, or an enantiomer, polymorph, or -pharmaceutically acceptabl e salt thereof in combination or ahernali.on with a second imMnnosupptessive ag t Exemplary immunosuppressive agents include, but are no limited to prednisone, budesotride, prednisolone, cyclosporine, tacrolimus,. sitoHmus, everoiimus, azatMopriae, feflunomi.de., mycophenolaie, ahataeept, adalimuniab, anakinra. certolizurtid , eianereept, golininrnab, inilixlraab, ixekizumab, fiatali¾aniah\ rltuxi mb;, secukmumab, tocHiztimab* ustek!tturoab, vedoH¾unmh ? basiiiximab, dacliziirnab, niarorrionab, or combinations thereof.

In some embodiments, the compound according to Formul I, Formula H, Formula ΙΉ, or Formula IV, or an enanlioniet , pol morph, or

pharmaeeutica!l aceeptabie salt thereof is administered in an amount effecti ve to increase exP3 g es ion on m tm cells, for ex mple T cells, including but not limited to Tregs such as il regs and nTregs.

in other embodiments, the eonipotnid according to FPrmula 1, F r ula: II, Formula III, or Formula IV, or an enan&oraer, polymorph, or

pharmaceutically acceptable salt thereof is adfoa isiered in an amount effective to increase proliferation of iTregs and nTregs,

Still another embodiment provides a pharmaceutical composition containing a compound according to Formula I, Formula II, Formula 01, or Formula I V, or an eaantiomer. polymorph, or pharmaceutically acceptable salt thereof and an excipient The compound according to Formula L Formula II, Formula III, Formula IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof is typically in an amount effective to increase a suppressive immune response when administered to a subject in need thereof.

Anothe embodiment provides a method of increasing an immune suppressive response in subject in need there of b contactin immune cells ex vivo with the compound according to Formula I, Formula II, Formula 01. or Formula IV, ©ran enantiomer, polymorpli, or pharmaceutically acceptable salt thereof, i«. an amount effective to increase expression of FoxPS on the immune cells, and administering the contacted immune cells to the subject, one embodiment, the immune cells are autologous immune cells. The immune cells can include T cells including but not limited t nTregs and iTregs.

Another embodiment provides a method, .for inhibiting or reducing transplant rejection in a subject i need thereof by administeri ng to the subject an effective amount of a compound according to Fommla L Formula II, Formula ill, or Foonuia IV, or an enantionier, polymorph, or pharmaceutically aceeptabie salt thereof to increase Fo>tP3 expression on im une cells of the subject In some embodiments, the increase of FoxP3 on immune cells of the subjects induces, promotes or iiicreases a suppressive immune response in the subject

Another embodiment provides a method for treating Graft-versus-host disease in $&bjeet in .need thereof by administering an ef ective amount, of die compound according to Formula Ϊ, Formula Π, Formula OS, or Formula IV, or an enanfianier, polymorph, or pbaitiiaceftticatly acceptable salt thereof to increase Foxl?3 expression on immune cel l s of the subject .

Another em dim nt provides a method for treating chronic infection in sabject m need thereof by administering an effective amount of the compound according to Formula I, Formula II, Formula III, or Formula IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof to Increase FaxP3 expression on immune cells of the su ject. BRIEF .DESCRIFOON OF THE DRA I GS igure 1 shows the structure of the compound of Form ula I V (mJJ 6 A), Figures 2A-2F are histograms of FACS sorted i ' Tregs treated as indicated with niJ½4A. Figure 2G is a bar graph showing percent CD4- Foxp3+ T cells treated with the indicated amount of mjJ64A. Figure 2H is a bar graph of mean. fluorescence intensity ( Fi) CD4-fFdxp3 cells) treated as indicate with uiJjMA,

Figure 3 A is an mutoradiograph of a Western Blot showing Foxp3 expression in iTregs induction and treatment with the indicated amount of mJJ64A, β-act serves as the control Figure 3B is a repeat of the experi ment in Figure 3 A.

Figure 4 A is a bar graph representing the expression of thrse isoforrns o Akt (Akt I s Ate, and Akt3) in Tconv (gray bar) and Treg (Mack bar) cells. Figure 4B is a western blot showing the expression of Afatl, A ' k¾ and Akt in. Teonv and Treg cells, p-actin serves as a loading control, Figure 4C is a western blot showing pSer expression in an IP pulldown of Akt! , Akt2 :i or Akt3 in non-stimulated or stimulated- Teonv and rvTreg cells. Figure 41) is a bat graph showing suppressive activity of Tregs :t¾? WT (gray bar) and AM3 KO (Mack bar) mice. The X~axis represents Teonv to Treg ratio. The Y-axis represents norma! ized percent of proliferation. Figure 4E is bar .graph representing IL- 10 levels (pg ml) in Tregs from WT, Aktl. KO, Aki2 O, of Aki3 KO mice. Figure 4F is a bar graph representing TGFjJ levels (pg/ml) in. Tregs from WT ; , Aktl K s Ak£2 KQ Of Akt3 KO mice.

Figure 5 A is a line graph representing percent survival of RAG colitis mice treated with PBS ·(»), WT naive T cells + PBS (a), WT naive T cells + WT t Tregs ( A), or WT nai T cells + Akt3 nTregs (▼). Figure SB is a line graph representing tumor volume (mm 3 ) over time (days) in mice with adoptive transfer of EL4 (♦), PC61 ÷ EL4 (0), PC61 + EL4 + WT Treg (ml or PC6i + EL4 + Akt3 KO Treg (A).

Figure oA is a western blot showing expression of Aktl . ,. Akt2 f and Aki3 iti ' Th and iTreg cells. β-actin is used as a. loading control, .Figure 6 B is a bar graph representing NA expressio of Aktl , Akt2, and Akt3 in Th (gray bar) and iTreg (black bar) ceils. Figure 6C is a western btof showing pSer expression in Th or Treg cells after IP pulldown of Akt 1 , Akt2, or Akt3. Figure 6D is a Bar graph showing the percent of FoxPS cells withi CD4 T cell population in Th (gray bar) and iTreg (black bar) cells in Akt l KO mice, Akt2 KO mice and Akt3 KO■ mice. Figure 6E is a histogram showing FoxP3 induction in Teonvs i response to TGF|¾ in Th (WT) f GV (solid line , ITreg (WT) + C¥ (dotted line), and iTreg (WT) + Akt3 sfeRNA (dashed line).

Figure 7 A. is a western blot showing Akt3 and. FoxP3 expression i control and Akl3 knock-in Tregs, β-actin is used as a loading control. Figure 78 is a. histogram showing FoxPS expression. Figure 7C is a bar graph showing: MFI (CD4 + FoxP3) in control and Akt3 KI regs. Figure 7D-1 shows histograms representing expression of IL2 and actin in control and -Akt3 KI Tregs. Figure 7D and. Figure 7G show 1L2 expression in controls and Aki3 Kl, respectively. Figore ?B and. Figure 7H show m. expression in control arid. Akt3 Kl, respecti vely. Figure 7F and Figure 7 show the overlay o the expression peaks of IL2 and actin for control and Akt3 KI, respectively. Figure 8 is a bar graph show? tig the effect of MJ¾4A on the expression of AM p$ 73, AM2 pS474 s AM3 pS47¾ and Akt pan $473,474,472111 A2780 cells compared to control The Y axi represents integrated density value.

Figure 9A is a western blot showin the expression of F oxP3, pAktS, A.kf3, pAkt l , and Akt 1 in activated Tcotrv cells induced with TGF-β and treated with various concentrations of m3J64A, Figure 9B is a histogram showing the expression of ROR^t and foxP% in. activated iTregs: induced with TGF~p and treated with various concentrations of mJ J¾ A. Figure 9C is a western blot showing FoxP3 expression in activated tTregs ; induced with TGF-p * treated with various concentrations of nxlI64A.

Figure 10 A shows histograms representing proliferation of activated iTregs induced with TGF-β and treated witii various concentrations of i Ji A, Figure tOB is a bar graph showing percent proliferation of iTregs treated wit various concentrations of mJj64A, The X-axis represents concentration of niD64A, The Y-axis represents percent proliferation. Figure IOC is a bar graph showing the- percent of live cells in tTregs treated with various concentrations of raJJ6 ' 4A. The X axis represents treatment and the Y axis represents percentage of live cells. Figure S OD is a set of histograms showing proliferation of activated nTregs treated with various concentrations ofmJJ64A. Figure I0E is a bar graph showing percent proliferation of nTregs treated with various concentrations of niJj¾4A. The JC-asis represents treatment and the Y-axis represents percent proliferation. Figure 10F is a bar graph showing the percent of Hve cells ¾ nTregs treated with various concentrations of miJ 4A. The X- axis represents treatment and the Y-axis represents percentage of live cells. Figure 10G is a set of histograms showing proliferatio of CD4 T cells treated with various concentrations of mil 6 A. Figure ! GH is a bar graph showing percent proliferation of CD4 T cells treated with various concentrations of ffiJJ A, The X-axis represents concentration, of mJJ64-A.. The Y-axis represent percent proliferation. Figure ίΟΧ is a Bar graph showing tiie percent of live ceils in-CD4 T cells treated with various concentrations of mj J6 A. The X-axis represents treatment and the Y-axis represents percentag of Hve ceils. Figure 10J is a set: of his oeratns ho ks proliferation of CDS T cells tre ted with various concentrations of rnJJ64A\, Figure 10K is a bar gra h showing percent proliferation of CD8 T ceils treated with various concentrations of m J64A. The X-axis represents. concentration of mJJ64A, The Y-axis represents percent pfoiiieration. Figure WL is a bar graph showing the percent of l ve ceils in CDS T cells treated with various concentrations of niJJ64A. The X-axis represents ireatoett arid tile Y-axis represents pe¾e« ge of live ceils.

Figure 1 1 A is a set of histograms showing the suppressive ftmction of mous iTregs in untreated and mjfj$ A treated iTregs. The ratio of iTre to Tconv cells was 0:1 s 0.5; 1, 1 : 1, and 2:1. Figure -1 IB is bar graph, showing the percent proliferation in untreated (black bar) and niJi¾ A treated (gray bar) cells at TregiTconv ratio of 0,5; 1 , The X-axis represents the experimental group and the Y-axis represents percent proliferation. Figure 1 1 C is a bar graph showing the percent proliferation in untreated (black bar) and mjJ64A treated (gray bar) cells at TregiTeonv ratio of 1 L The X-axis represents the experimental group and the Y axis represents percent proliferation. Figure- Ί 1 D is a bar graph showing the percent proliferation in untreated (black bar) and mjJ64A treated (gray bar) cells at a Tre-grTconv ratio of 2: 1. The X-axis represents the experimental group and the Y~axis represents percent

proliferation- figure. 1 A. is a set of histograms showing the suppressive fenctio of untreated and mJJ64A treated nTregs, The ratio ofnTreg to Tconv cells is 0:1, 0.5;! , i s 2:1, and 3;L Figure 12B is a bar graph showing the percent

proliferating cells in untreated and mJ ' J64A treated nTregs in a mixture of nfreg and Tconv (0.5; 1), Figure 12C is a bar graph showing the percent proliferating cells in untreated and mJJ64A treated nTregs in a -mixture of nTreg and Tconv (1:1). Figure 12D is a bar graph showing the percent proliferating cells in untreated and mJJ64.A treated nTreg$ i mixture o Treg and Tconv (2: 1). Figure 12 E i a bar graplv showing the percent: proliferating cells i nntreated and ffiJJ64A treated «Tregs In a mixture of nTreg and Tcon v (3:1). Figure J.2F is a histogram showing proliferation of nTregs treated ' With, various concentrations of r»jJ64A. Figure I2G is a bar graph representing tire percent proliferation af nTregs ixeated with various concentrations of mJJ64A. The ~ axis .represents treatment group and the Y -axis represents percent pmiiferation. Figure 12H is a bar grap representing percent live cells in nTregs .treated wi h various co centratio s of nr 64A,. The X-a is represents treatment group and the Y-axis represents percent proliferation.

Figure 13 A. is a set of histograms showing FoxP3 and JT!D expressio in. n ' l reg cells treated with various concentrations of rn J J64A. Figure 13B is a bar graph representing the percent. ofiL-10 " FoxP3 ! cells n nTregs treated with various concentrations of mJJ64A.

Figure 14A is m illustration showing the experimental method and treatment schematic for TC-1 tumor ex erirnenis t Figure ! 4B is a line graph showing ftimor volurne (cur 5 ) over time (days) for untreated (A) and mJJ64A treated (*): TCI. tumor bearing mice.

Figure 15A is a bar graph representing the number of CDS '5 ceils per 10* live cells in tumors from untreated: (black bar) and mJJ 4A treated (gray bar) mice. Figure I SB is a bar graph representing the number of CD4 cells per 10* live cells in. tumors from untreated (black bar) and mJJ64A treated (gray bar) mice. Figure 15C is a bar graph representing the number of FoxP3 ;" ceils per W' CD4 *' cells in tumors from untreated (black bar) and m JI64A treated (gray bar) inice. Figure i SO is a bar graph representing the number .ofC&8 ' % Ρ χΡ3^ '' CD4 and Treg celts per 10 c> live ceils in the spleen of untreated (dark gray bar) or mJJ64A treated (light gray bar) tumor-bearing mice. Figure 15E is a bar graph representing the number of CD8\ FoxP3 SG CD4 ; ; and Treg cells per .10* live cells in the spleen of untreated (black bar) or mJJ64A treated, (light gray bar) tumor-free mice-.

Figure 16A is a schematic illustration of the experimental design of colitis ruodel ' Figure I6B is a line graph representing weight: (g) over time (days postinfection) for control .(#), iTreg (a), raJi 64 A + iTreg. ( AX and.

miJ64A ( ) treated colitis mice-, Figure 16C is a line graph representing aormalized weight over time (days post-injection for control (·), iTreg {*), «xJJ64A + iTreg (A), and rnJ M4A (o) treated colitis mice. Figure ! <SD is a line graph showing percent survival of untreated. *), iTreg (Mite cimle mJJ64A + iTreg (▼), mid mjJ64A (red: circle) treated coli tis mice. The X-axis represents time (days) and the Y-axis represents percent survival. Figure 16B-1 are representative photos of untreated (Figure I6E), iTreg treated (Figure .16F), JIa treated iTreg (Figure 16G), untreated (Figare 1 H), and mJJ64A treated (Figure 161) colitis mice. Lower image- shows- rectal prolapse in untr ated groups ,

Figure 1.7 ' A is a photo showing representative colons f om untreated (UT), iTreg, MJJ64A treated iTreg, mJJ64A:, and wild-type (WT) mice.. Figure Ϊ IB is a. bar graph representing length and weight of colons from normal, untreated fUT), iTreg, niJJ64A 5 mid mJJ64A - iTreg mice. The X-axis represents the treatment group and the Y-axis represents colon weight/length (mg/nin ). Figure 17C-J- show representative histology sections from colons from WT norma! colon (Figure 1.7C), Rag " * * colitis- untreated (Figure 17D), WT- normal colon (Figure 17B), Rag" " colitis- untreated (Figure 17F), Rag" " colitis- iTreg treated (Figure ' 1 G) ? ' Rag * ' " colitis- mIJ64A (iOnig/kg) treated (Figare 1.7H), Rag' ~ colitis- iTreg treated (Figure 171), and Rag " " colitis- mJJ64A

(JOmg kg) treated (Figare 1.7 ).

Figure ISA is a bar graph showing the number of CD4 ? T ceils per 10* live cells in the spleen of WT, UT, iTreg;, mTJ64A, and ηιΙΙ6 treated iTreg treated Rag ;~ mice. The X-axis .tepresetits t eaUTieni grou an ' d the Y-axis represents number of CD * cells per 10* live cells. Figure I SB is a bar graph showing the percent of FoxP3 cells per CD4 ÷ T cells in the spleen of WT, UT, i reg, m.J J64A, and mJ ' J6 A treated iTreg treated Rag" ' mice. The X-axis represents treatment group and the Y-axis represents number of Fox;P3 : cells per CD4 ; cells. Figure i 8C is a bar graph showing the percent of FoxF3 " ceils per CD * T cells in the spleen of WT, UT, iTreg, .r IJ64A, and .mJJ6 A treated ' iTreg treated Rag ":'" mice. The X-axis represents treatment group and th Y-axis represents number of FoxE? ' cells per C04 i ceils. Figure I SO is a bar graph showing the number of C.D4 * T cells per 10* li e cells in the lymph node ofWT, UT, iTreg. mJJ64A, and mJJ64A treated iTreg treated Rag " " " mice, The X«axis represents treatment g ^np and the Y-axis represents - number of CD4 '; cells per 10 s;i live cells * Figure 1 SE is a bar graph, showing the percent of FoxFT ; cells per €04'· ' T cells in the lynrph node of WT, OT, i reg, ni.JJ64A, .and J J64A treated iTreg treated Rag " ' * mice. The X-axis represents treatment group and the Y-axis represents mimber of FoxP3 ; cells per CD4 ; cells. Figure 18F is a bar graph showing the percent of FoxP3~ cells per C.D4 *' T cells in the l mph node of WT, OT, iTreg, m JJ64A, and f¾£J64A treated. iTreg treated Rag * * " mice. The X-axis epresents treatment grou and the Y-axis represents number of FoxP3' cells per CD4 ; cells.

Fi gure 19A i s a schematic illustration of induction of experimental autoimmune encephalomyel tis (EAE) model. Figure 19B is a chart showing the grading criterion for scoring- severity of EAE. Figure 1 C is a line graph showing EAE score over time (days post EAE induction) for control. (* iTreg (blue circle), and mX?64A-l 0 (▼) treated mice. The X-axis represents time (days) and the Y-axis represents EAE score. Figure 19D is a line graph showing pe cent survival over time (days) for untreated ( » ), iTreg treated (·), and mJ J64A-10 treated (Ψ ) mice. The X-axis represents time (days) and the Y-axis represents percent - survival . Figure 1 E is a line graph representing EAE score over time (days post EAE induction) for control. (»), iTreg (♦), mJ.T(S4A~3 (blue circle), ni06 A»6 (f), and :mJJ64A-10 (A) tr ated EAE mice. The .X-axis represents time (days post EAE induction) and the Y-axis represents EAE score. Figure 19F is line graph representing percent survival over time (days) for untreated {*), iTreg (bine circle), j¾jJ64A-3 (¥), mJJ64A-6 (A), and mlJ64A- 10 (♦) treated EAE mice. The X-axis represents time (days) and the Y-axis represents percent survival

Figure 20A-C are bar graphs showing the percent of FoxET cells per CD * T cells in the spleen (Figure 20A), blood (Figure 20B), and brai (Figure 20C) of UT, iTreg, mJJ A-3 s mIJ64A-6 and mJ 4A-IO treated EAE mice. The X-axis represents treatment group and the Y-axis represents number of FoxP3 * cells per CD * cells. Figure 20D-F are bar graphs showing the percent of Fox 3" cells per CD4 * T cell in the spleen (Figure 20D), blood (Figure 20E), and hihm (Figure 20F) of UT 5 ITreg, ntJJ A-i, ni 64A~6 and mJJ64A-!0 treated EAE mice. The X -axis represents ieatraent group and the Y-axis represents number of FoxP3 " ceils per CD4 * cells. Figure 20G are bar -graph showmg the percent of ROE ! cells per C 4 ; T cells in the spleea (Figure 20G) > blood (Figure 20H), and braia (Figure 201) of UT, ITreg, m! J64A-3, mJJ64A-6 and ffijJ64A-l 0 treated EAE mice. The X-axis represents treatment grou and trie Y-axis represents numb r of EQR ' cells per CD4 ÷ cells.

Figur .21 is -a bar grap showing the percent of live human iTregs in cells treated with various concentrations ' of mJJ64A., The X-axis represents treatment group and the Y~axis represents percent of live cells. Figure 21B is a bar graph showing the percent of FoxP3" CD4 ' cells in human iTregs treated wit various concentrations of mJJ64A. The X-axi represents treatment group and the Y-axis represents percent of FosP3 ;* C ' D4 + cells, DETAILE DESOWFTTON GIF THE INVENTION

I. Deli rntions

The term "stimulate expression * means To affect expression of, for example to induce expression or activity, or induce increased greater expression or activity relative to norma!, healthy controls.

The terms "imninne activating response", "acti vating immune response'V and "iiOTupe stimulating response" refer to a .response that initiates, induces, enhances, or increases die activation or efficiency of innate or adaptive immunity . Such immun responses include, for- example, the development of a beneficial humoral (antibody mediated) and/or a cellular (mediated by antigen- specific T cells or their secretion products) response directe against a peptide in recipient patient Such a response can be an active response induced by administration of immnnogen or a passive response induced by administration of antibody or rimed T-celk, A. cellular immun response is elicited by the presentation of polypeptide epitopes in association with Class 1 or Class II MKC molecules to activate antigen-specific CD4 'S' T helper ceils and/or DS ' cytotoxic T cells. The response ca also involve activation of monocytes.

An "immunogenic agent" or "immtmogen" is capabl e of inducing an imm nioiogical response agains itself on administration to a mammal, optionall in conjunction with an adjuvant.

The term "Imm e cell" refers to cells of the innate and acquired immune system including neutrophils, eosinophils, basophils, monocytes, macrophages, -dendritic cells* lymphocytes including B cells, T cells* and natural killer cells.

As used herein "eonvestional T cells'* are T lymphocytes that express an Cx T cell receptor (TC&) as well as a eo-reeepfer CD4 or CDS, Conventional T cells are present is the peripheral blood, lymph nodes, and tissues. See, Roberts and C rardi, "Conventional and Unconventional T Cells", Ckjiicai and Basic

The "bioactivity" of Akt3 refers to the biological function- of the. Akt3 polypeptide, Bioaetivity can he increased or reduced by increasing or reducing the activity of basal levels of pol ypeptide, increasi ng or reducing the avidity of5 basal levels of poly peptide, the quantity of the polypeptide, the ratio of Afct3 relative to one or more other isoforms of Akt (e.g.. Akt ! or Akt2) of the pol ypeptide, increasing or reducing the expression levels of the polypeptide (including fey increasing or -decreasing. -RN A expression of AkO), or combination thereof. For example, -b oav-ailabte Akf3 polypeptide is a

0 polypeptide that has kinase activity and can hind to and phosphorylate

substrate of Akt.3. Akt3 polypeptide that is not bioavailable includes Akt3

mixture of single-and double-stranded regions, single- and double- stranded UNA, and UNA that is mixture of single- and double-stranded regions, hybrid molecules comprising -DMA and NA that maybe single -stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. The term *¾ueletc acid" or "'nucleic acid sequence" also encompasses a polynucleotide as defined above.

In addition, polynucleotide a used herein refers to triple-stranded regions comprising RNA or DMA or both RNA and DMA. ' The strands in such regions may be from the same molecule or ' .from, different molecules. The regions may include all of one or more of the molecules., but more typically '

(Gin, Q), Glutamic Acid (Gin, E), Glycine ( ly, G), Sistidhae (His, H),

Isoleocine (lie,: I) s Leucine (Leu, L), Lysine (Ly$ t i¾ Kfemionine (Met M} ? Phenylalanine (Phe, F), Proline (Pro, P), Serine (Sex, S), Threonine (Thr, T), Tryptophan (Tip* W)„ Tyrosine (Tyr, Y), and Valine (Val* V).

•• Variant" refers to a polypeptide or polynucleotide that differs from a reference polypeptide or polynucleotide, but retains essentia} properties. A typical variant of a polypeptide differs in amin acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and tl¾e variant ate closely similar overall and, in. many regions, identical, A variant and reference polypeptide nray differ in amino acid sequence by one or more modifications ( g-., substitutions,

additions, and/or deletions), substituted or inserted amino acid residue may or n*ay not he one encoded by the genetic code:- A, variant of a polypeptide may be. naturally occurrin such as an allelic variant, or it may be a variant that is not: known to occur .naturally..

M odifications and changes can be made i n the stractore of the

polypeptides of the disclosure and still obtain a molecule having similar characteristics as the polypeptide (e.g., a conservative amino acid substitution). For ex l certain anihio acids can be substituted for o her amino acids in a se uence' without appreciable loss of activity.- Because it is the interactive capacity nd nature of a. polypeptide thai defines that polypeptide's biological fiinetional activity, certain, amino acid sequence substitutions can be made in a polypeptide sequence and nevertheless obtain a polypeptide with like properties, in making such changes,, the hydropathic i ndex of amino aeids can be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function, on a polypeptide is generally understood in the art. It is known that certain amino acids can be substituted for other amino acids having a similar hydropathic index or score and still result in a polypeptide with similar biological activity. Each amino acid has been assigned a hydropathic index on the basis of its hydro hobicity and charge characteristics. Those indices are: isoleueine (÷4,5): valine (+4.2); leucine (43,8); p en lal nine (+2.8); cysteine/cystine (4-2,5); methionine (HJ>); alanine -(+1.8); glycin (-0.4); threonine (-0.7): serine ( h$); uyptophan {¾$ ; tyrosine (- ί .3); proline {- 1.6); histidine (-3.2); glutarnate (-3.5); glutaraine (-3,5); aspartate (-3.5); asparagine (- 3.5); lysine (-3,9); and arg!nine (~4.3), :

I t is believed that the relati ve hydropathic character of the amino acid determines the secondary structure of the resultan polypeptide, whic in tor defines the interaction of the polypeptide with other molecules, such as enzymes, substrates, receptors, antibodies, antigens, and cefaclors, it is known in. the art that an amino acid can. be suhsdttned b another amino acid having a similar hydropathic index and still obtain a functionally equivalent polypeptide:, i n such changes, the substitution of amino acids whose hydropathic indices at e within ± 2 is preferred, those wit-h ^ 1 are particularly preferred, and those within ± 0.5 ' are even more particularly preferred.

Substitution of like amino acids can also be made on. the basis of hydrop licity, particularly where, the biological functional e uivalent polypeptide or peptide thereby created is intended for use in immunological embodimeivis. The following bydrop lieity values have been assigned to amino acid residues: ar nhre (+3.0); lysine (+3.0); aspartate 1); glutaniate

(+3.0 1); serine (+03); asparagine (+0.2); gluiarauine (+0.2); glycine (Q); proline (-0:5 + t ); threonine (~0 ÷ 4); alanine (-0.:5); hlsridine (-0.5); cysteine (-. 1.0); .methionine (- 1.3); valine (-1.5); leucine (-1.8); i.soleucine (> < tyrosine (- 23); phenylalanine (-2.5); tryptophan (-3.4). it is understood that an amino acid can be substituted for another haying a similar hydtophlSicIty value and still obtain a biologically equivalent, and in particular, an iraraunologicaily equi valent polypeptide. In such changes, the substitution of amino acids whose hydrophllieity values are within ± 2 is preferred, those within ± I are particularly preferred, and those within +. 0,5 are even more particularly preferred.

As- utlined, above, amino acid substitutions are generally based on the relative similarity of the amino acid side-ehain. substitueats, .for example, their hydrophobicity, faydrophJIieity, charge, size, and the like. Exemplary substitutions -thai take · various - foregoing - characteristics Into consideration are well known to those of skill in the art and include (original residue: exemplary substitution): (Ala; Gly, Ser), (Arg; Lys), (Asa: Gin, His), (Asp; Glu, Cys, Set)* (Gin: Asn), (Glu: Asp). (Gly: Ala), (ffis; Asn, Gin), (lie; Leu, Val), (Leu; lie, Val), (Lys: Arg), (Met; Leu, Tyr), (Sen Thr), (Thr. Set), (Trp: Tyr), (Tyr: Trp, I%e). and (Val; e, Leu), Embodiments of this disclosure thus contemplate functional or biological equivalents of a polypeptide as set forth above. In articular embodiments of the polypeptides can include variants having about 50%, 60%. 70%. W%, f ø%, 95%, 96%, 97%, 99%, or more sequence identity to the polypeptide of interest. The term ^percent {%) .sequence identity" Is defined as th percentage of nucleotides or amino acids in a candidate sequence that are identical with, the nucleotides or amino acids in a reference nucleic acid sequence, after aligning the sequences and introducing gaps, if necessar , to achieve the maxi mum percent sequence identity. Alignment for purposes of detsTimning percent sequence identity can be achieved i various ways that are within the skill in the art s tor instance,, usn g publicl available computer software such as BLAST, BLAS ' f-2, ALIGN, ALIG -2 or Jvlegalign (DM ASTAiR) software, Appropriate parameters for measur ing alignment; including any algorithms needed to achieve maxima! alignment over the foil-length of the sequences being compared can be determined b known .methods *

For purposes herein, the %. sequence identity of a given nucleotides or amino acids sequence C to, with, or against a given nucleic acid, sequence D (which can alternativel be phrased as a given sequence C that has or comprises a certain % sequence identity to, with, or against a given sequence D) is calculated as follows:

} 00 times the fraction W.¾

where W is the number of nucleotides or amino acids- scored as identical matches b the sequence alignment program in that program's alignment of C and Ό, and where Z is the total nntn er of nucleotides or amino acids in O, i will he appreciated that where the length of sequence C is not equal to the length of sequence D, the % sequence identity of C to D will not equal the % sequence identity of D to C.

The term "carrier" refers to an organic or inorganic ingredient;, natural or synthetic, with which the active ingredient is combined t facilitate the application.

The term "phamiaceirticall acceptable" means a non-toxic material that does not interfere with the efiectiveness of the biological activity of the active ingredients. The teffti "pfaaimaeeiiticaliy-acce iablfi carrier" means one or more compatible solid or liquid fitters, diiutants or encapsulating substances which are suitable for administratio to a Imwao or other vertebrate animal

The term "effective amount or "therapeutically effective amount" mean a dosage sufficient to provide treatment a disorder, disease, or condition being treated, or to otherwise provide a desired pharmacologic and/or physiologic effect. ' The precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., age, imtmine system health, etc.), the disease, arid the treatment being effected.

The terms ' ndividual ' "subject," and "patient" are used

interchangeably herein, and refer to a mammal, including, but not limited to, humans, rodents, such as mice and rats, and other laboratory animals,

! Com positions for Activating Akt3

Com ositions and methods of their rise lor selectively activating Akt3 are provided herein.

One embodiment provides a compound of Formula 1:

Forrnnia I

or a pharmaceuttcal!y acceptable enaatiomer, salt, or solvate thereof, wherein:

C2o)-heteroaryl-O Ct-C»)-aikyl, -COOH, -OIL -SH, -SQ¾ ~C -Nft, or hatosen.

Another embodiment rovides a compoand of Formul 111:

Formula 111

or a pharmaceutically acceptable enantromer, salt, or solvate thereof,

Formula i V

or a p amiaceirtkaliy acceptable enantiomer, salt, or solvate thereof. The compound of Formula XV, also referred to as raj J64A, and anantioffiers, olymor s, pharmaceuticall acceptable s¾¾ and derivatives, thereof can be used t indiice, promote, or increase Aki3 bioactivify i imrnuae cells.

In some embodiments, the Atk3 activator is a derivative of Formula 1, Formula II, Formula ill or Formula IV. The term: "derivative" or "derivatteed" as used herein includes one or more chemical modifications of Formula I, Formula II, Fonmila III., or Formula IV, or euantiomef, polymorph, or pharmaceutically acceptable salt thereof Thai is, a "derivative 1 - may be a functional equivalent of Formula I, Formula. ¾ Formula III o formula IV which is capable of inducing: the improved phannaeological functional activity and/or behavioral response i a given subject. Illustrative of such chemical modifications would be replacement of hydrogen by a halo group, an alkyi group, an aeyl group or an amino group.

The chemical modification of Formula 1, Formula 11, Formul 111, or Formula IV, an enantiomer, polymorph, or pharmaceutica !y -acceptable salt thereof may either enhance or reduce hydrogen bonding interaction, charge

AA €Ti¾T¾GC CCTSX ^^

GiA&S¾¾ ! Γ T TAG iCTC AGACS&TίAGA¾ AAC ACGAC CT GAAAASΐ A ; rGATG AGGSTGG1' ¾TGGAC^GG;&iG ¾CA&T¾

GG¾C A ^¾T¾A XCXGT£¾CA^

fG&X CCIGG&C^

CCCA ¾0-V-A CCAAGC:GTCGGG^

C¾CGC¾&«mCACTCCAGTTTTTGXXTXTS^^

^ C QTfGCTGCS¾C GXG T ¾G¾¾T * EA

(S¾iDMO:l). Amino acid sequences are also known m. the art. See, Jbr example, UHiProiK Swiss-Frot ccessiosj no. Q9Y243 (Akt3 . HUMAN), which is specifically incorporated by reference in its entirety and provides the ammo acid sequence:

F F FQVTS E ί D RY FDE E ? T AQT X 'JI PS¾' J^ DCMDi¾BB.RPHE £¾ S Y S¾Si¾E (SEQ ID NO:2).

The domain structure of Akt3 is reviewed in Romano, Sei tifiea, Volume 2013 (2013):. Article ID 317186, 12 pages, and includes an. N-termmal pieckstrm homology domain. (PH), followed, by a catalytic kisase domain (KD), and the C~termmal regulatory ' hydrophobic region. The catalytic and regulatory domains are both important for the biological actions mediated hy Akt protein kinases and exhibit the maximum degree of homo logy mong the three Akt isofomis. The FH dom in binds lipid suhstfates, such as phospha idyHnositol (3,4) diphosphate (PiF2) and phosphatid linos ito! (3*4,5) triphosphate (HP3). The A TP binding site is situated approximately in. the middle of the catalytic kinase domain, which has a substantial degree of homology with the other components of the AGCkinases family, such as p70 S6 kinase (S6K) and p90 ribosomal S6 kinase (RSK), protei n kinase A (P A) and protein kinase S

(PKB). The hydrophobic regulatory moiety is a typical feature of the AGC kinases family. With . reference to SEQ I ' D NO:2 Akt 3 is generall considered to have the following molecule processing and -dortta sinic¾iriB · ^Hned ¼ w».

Mo!ec k Processing:

Meatere fcev Po$it¾oa(-0 Len h Pescriiiiiun

initiator methionine 1 1 Removed

Chain - 419 478 Akt3 Domain 5- 107 103 PH

Domain 148 -405 258 Protein kkmse

Domafe 406 - 479 74 AGC -kinase C-tennmal Nucleotide binding 154-162 9 ATP

Sues:

preferably of 0,O§1 o 500 mg kg, more preferabl 0.01 to 50 mgi ' kg of body weight daily are administered to mammals- ^

1. Deli ver Vehi cles

Compounds of Formulas I, II, ill, and IV can be actennistefed to a subject, preferably a human subject, where it is taken. -up nto the cel ls of a subject with or without the aid of delivery vehicle. Appropriate delivery vehicles fbr the disclosed active agents are known in the art and cm be. selected to spit the particular active agent For example, ½ some embedments, the compound is incorporated Into or encapsulated by a ftopar&cte* tnicrojjarticle, micelle, synthetic lipoprotein particle, or carbon nanotube. ί-or example, the compositions can be incorporated into a vehic le such as polymeric

microparticles which provide controlled release of the acti ve agentCs), In some embodiments, release of the drug(s) is controlled by diffusion of the active agent(s) out of the microparticles and/or degradation of the polymeric particles by hydrolysis and/or enzymatic degradation. Suitable polymers include ethylcellufose and other natural or synthetic cellulose derivatives. Polymers which are slowly soluble and form a gel in an aqueous environment, such as hydroxypropyl methyl-cellulose or polyethylene oxide may also be suitable as materials for drag containing microparticles. Other polymers include* but ate not limited, to., polyanhydrides, poly (ester anhydrides), polyhydxoxy acids, such as polyiactide (PLA), pofyglyeolide (PGA), ppIy(iactide-co-§iycolide} (PLGA), poly-3-hydroxybut rate (PHB) and copolymers thereof, poly*4 iydrQxybutyrate (P4HB) and copolymers thereof, polycaprolactone and copolymers thereof* and. combinations thereof;

In some embodiments, compounds according to -Formula I, Formula II,

Formula til, or Formula iV ? or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof and a second therapeutic agent are incorporated into the same particles an are formulated for release at different times and/or over different time periods, f or example, in some embodiments, one of th agents-is released entirely from the particles before release of the second agent begins. In other embodi ments, release of the firs t agent begins followed by release of the second ageafc before th al of the first agent is r el eased, in still other embodiments, both agents are released at the same time over the same period of time or over different per iods of time.

The compounds according to Formula 1, Formate II, Formula ill or Formula IV, or an enantiotner, polymorph, or pharmaceutically acceptable salt thereof can foe incorporated into a delivery vehicle prepared from materials whic are insoluble in aqueoirs solution, or slowly soluble in aqaeoas solution, fo l are capabl of degrading within the G I tract by means ind ding enzyrnaflc degradation, surt ctant action of bile acids, and/or mechanical erosion. As used herein, the term "slowly soluble in water" refers to materials that are not dissolved in water within a period of 30 minutes. Preferred examples include fats, fatty substances, waxes, wax-like substances and mixtures thereof " Suitable fats and fatty substances include fatty alcohols (such as lauryl, rayristyl stearyl, cetyl or cetostearyl alcohol), fatty acids and derivatives, including, but. not limited to, fatty acid esters, fatty acid glycerides (mono-, di- and tri-glyeerides), and hydrogenated fats. Specific examples include, but are not limited to hydrogenated vegetable oil, hydrogenaied cottonseed oil, hydrogenated castor oil, hydrogenated. oils available under the trade name Sterotex®, stearic acid, cocoa butter, and stearyl alcohol. Suitable waxes and wax-like materials iiiclnde natural or synthetic waxes, hydrocarbons, and normal waxes.

Specific examples of waxes include beeswax, glyeowax, castor wax, camauba wax, paraffins and candelilla wax. As used herein, a axrlike material is defined as any material which is normally solid at room temperature and has a melting point of .from about 30 to 3Q0 ¾ C. The release point and/or period of release can be varied as discussed above.

2. Fharmacettiical Compositions

Pharmaceuticai compositions including compounds according. to

Formula I, Formula 11, Formula 1H or Formula IV with or without a delivery vehicle are provided. Plmnnaceatkal eomposiiions can be formulated for administration by parenteral, (intramuscular, intraperitoneal, Intravenous (IV) or subcutaneous injection), enteral, traasmucosal (nasal, vaginal, rectal or

' 3 >' suMitigtial),. or trmisderftt¾l (either passively or -using iontophoresis or eieeiroporaiion) routes of administration or using bioerodible inserts and can be formulated in dosage forms appropriate .for each route of administration.

In certain embodiments, the compositions are administered locally, fo example by injection directly into a site to be treated (e..g,, : into a rumor). In. some embodiments, the ' compositions are injected or otherwise administered directly into he vasculature onto vascular tissue at or adjacent to the intended site of treatment (e. g, , adj acent to a tumor). Ty i cally, local: administration catises an. increased localized concentration of the composition which: is greater tha that which can be achieved by systemic admini strati on.

a. Formulations for Parenteral Administration Compounds according to Formula l f Formula 11, Formula 111, or Formula IV, or an enaniionier, polymorph, or pharmaceutically acceptable salt thereof and pharmaceutical compositions thereof can. be administered in m aqneons solution, by parenteral injectio , The formulation may also be in the form of a suspension or emulsion. In general, phan mceimcai compositions are provided including effective amounts of the active agent(s) and optionall include pharmaceutically acceptable diluents, preservatives, solnbilizers, emnlsiflers, adjuvants and/or carriers. Such compositions include diluents sterile water, birff bred sal ine f various buffer content (e,g. , Tris-HCl, acetate, phosphate), pH and ionic . stren t ; and optionally, additives such as detergents and solabilixing agents (e.g., ' TWEEN® 20, TWEEN® 80 also referred to as polysorbate 20 or 80), anti-o idants (e. ., ascorbic acid* sodium metabisulfite), and preservatives (e.g., Thiffiersot benzyl alcohol) and bulking substances (e.g., lactose, mannitol). Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and com oil, gelatin, and injectable organic esters such as ethyl oieate. The foniiulations may be lyop iized -a»d r^issoi ed riesus ^nded ' irn sediaiely before use. The formnlstion ma be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. Compounds according to Formula i, Formula H, Formula ill, or Formula IV » or an enartt m.er, ol morph,. or pharmaceutically acceptable salt thereof can formulated for enteral .administration:. Suitable oral dosage forms -of compounds ' of Formula i, Formula ¾. Formula III, or Formul IV, or .an eaaatiomer, polymorph, or pharmaceutically acceptable, salt thereof include tablets, capsules, solutions, suspensions,, s rup , and lozenges * Tablets can be. made usiag compression or molding techniques well known in the art. Gelatin or non-gelafm capsules can prepared as hard or soft capsule shells, which can encapsulate liquid, solid, and semi-solid fill materials, usin techniques well known in the art.

Foj »iilatioiis raay he prepared using a pharmaceutically acceptable carrier. As generally used herein "carrier" includes, but is not limited to, diluents, preservatives, binders, lubricants, disintegrators, swelling agents, fillers, stabilizers, and combinations thereof.

Carrier also includes all components of the coating composition, which may include plasticizers, pigments, colorants, stabilising agents, and gfidaats. Delayed release dosage formulations m y be prepared as described in standard references. These references provide information oa carriers, materials, equipment and process for preparing tablets and capsules arid delayed release dosage forms of tablets, capsules, and granules.

Examples of suitable coating materials include, but are not limited to, cellulose polymers such as cellulose acetate phthalais, hydroxypropyl cellulose, hydroxypropyl niethyleellnlose, hydroxypropyl methylcelhilose phihalate and hydroxypropyl niethyieelhilose acetate succinate; pol vinyl acetate phihalate, acrylic acid polymers and copolymers, and methacryiie resins that are commercially available under the trade name Eudragit® (Roth Pharraa, Westerstsdt, .Ger an ' },; zeln, sheSlae, and polysaccharides.

Additionally, the coating material may contain conventional carriers such as plastici¾ers, pigments, colorants, glidaats, stabilization agents, pore formers and surfactants. Optiona! pfaaimaceiiticaliy acceptable exeipisnts Include, ' but te not limited to, diluents., binders, lubricants, disintegrams, colorants, stabilizers, and surfactants,. Diluents, also referred to as "fillets," are typically necessary to increase the bulk of a solid dosage form so that a practical size is provided for com r ssion of tablets or formation of beads and granules. Suitable diluents include, but are not limited to, diealciurn phosphate ds hydrate, calcium. sulfate, lactose, sucrose,, tMintnt l, sorbitol, cellulose, raieroet sialiine cellulose, kaolin, sodium chloride, d y starch, hydroteed starches, ptegelatiniaed starch, .silicone dioxide, tttai«uf» oxide, magnesium altfmimim. silicate and powdered, sugar.

Binders are used to Impart cohesive qualities to solid dosage

formulation, and thus ensure that a tablet or bead, or-gra ile remains intact after the formation of the dosage forms. Suitable binder materials include, but are net limited to, starch, pregelattnized starch, gelatin, -sugars ' (including sucrose, glucose, dextrose, lactose and sorbitol), polyethylene glycol, axes, natural and synthetic gums such as acacia, tragaeanth, sodium alginate, cellulose, including hydroxypropyiri etiiylceiiulose, hydroxypropylcefluiose, eihyleellulose, and veegu o, and syndetic- polymers such as acry lic acid andraethacrylic acid copolymers, methaerylic acid copolymers, methyl methaerylate copolymers, aminoalkyl ethacrylate copolymers, polyactylic acid polymethacryllc acid and pQlyvinylpynolidone,

Luhiicarits are used to tacllitats tablet mauoi¾et«te. Examples of suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, glycerol hehenate. polyethylene glycol, talc, and. mineral oil.

Dislntegrants are used to facilitate dosage form dismtegration or

"breakup" after administration, and generally include, but are not limited to, starch, sodium starch glycate, sodium earboxyraethyl starch, sodium carboxymethyiceiitilose, hydro&ypropyl cellulose, pregeiatini¾ed starch, clays, cellulose, alginate, gums or cross linked polymers, such as cross-linked PVP (Pol lasdone© XL from GAF Chemical Corp). Stabilizers are used id -inhibit or retard drug decomposition, reactions, which include, by way of example, oxidati ve reactions. Suitable stabilizers Include, but are not limited to, antioxidants, bittyiated hydroxytohtene (Bl-IT); ascorbic acid, its salts and esters; Vitamin E. tocopherol and its salts; sulfites such as sodium metabisulphite; cysteine and its deri vatives; citric acid; propyl galkte, and butylaied hydroxyanisoie (B ' HA) *

Oral dosage t rus, such as capsules, tablets,, solutions, and. suspensions, can for formulated for controlled release. For example, the one or more compounds and optional one or more additional active agents can be formulated into nanopariicles, microparticies, and combinations thereof, and encapsulated i n a soft or hard gel ate or non-gelatin capsule or dispersed i a dispersing medium to form an oral suspension or syrup. The particles can be formed of the drug and a controlled release polymer or matrix. Alternatively, the drug particles can be coated with one or more controlled release coatings prior to incorporation in to the finished, dosage form.

in another embodiment, the one or more compounds and. optional one or more additional active agents are disper sed in a -matrix mater ial , which gels or emulsifies upon contact with an aqueous medium, such as physiological fluids, in the case of gels, die matrix swells entrapping the active agents, which are released slowly over time b diffusion and/or degradation of the matrix m terial Such matrices ' can. be fonm la ted as tablets or as fill materiais for hard a id soft capsules.

In still ano her embodiment, the one or more compounds, and optional one or more additional active agents are formulated into sold oral dosage form.. such as a tablet or capsule* and the solid dosage form is coated, with one or mor controlled release coatings, such as a delayed release coatings or extended release coatings. The coating or coatings may also contain the compounds and dr additional active agents..

Extended release das ge jt ns

The extended release formulations are generally prepared as diffusion or osmotic systems, which are known, in the art, A diffusion, system typically consists of two types of devices, a reservoir and a matrix,, and Is well biown and described in the art. The matrix devices are generally prepared by compressing the drug with, a slowly dissol ving polymer- carrier into a tablet form. The three major ty es of materials used in the prepafatioa of matrix devices are insoluble plastics, hydr<op ilic polymers, and fatty compounds. Plastic matrices include, but are not limited to, methyl acryiate-methyl methacry late, polyvinyl chloride, and polyethylene. HydrophiMo polymers include, but are not limited to * ce ulosic polymers such as methyl and ethyl cellulose, hydroxyalky !celhioses such as hydroxypropyl-cellulose, bydroxypropyteedi lcellrilose, sodium cat oxyraetbyk-eiltilose * and Carbopol® 3 , polyethylene oxides and mixtures thereof. Fatty compounds include, but are not limited to, various waxes such as carnauba wax and glyceryl tristearate and wax-type substances including hydrogenated castor oil or hydrogenated vegetable oil, or mixtures thereof.

In certain preferred embodiments, the plastic materia! is a

pharmaceutically acceptable acr lic polymer, including but not 3 imited to, acrylic acid and me acrylic acid copolymers, methyl methacrylate, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethy! methacrylate, aminoalkyl methacrylaie copolymer, poIyCacrylic acid), poly(methaciy!ic acid), raethacrylic acid aikylaniine copolymer poJy(met yl memaerylate),

poiy(mediacryiie acid)(a»hydr:ide;) ! polymethacrylaie,. polyacryla ide, poiy(meihacr iic acid anhydride); and giycid l methacrylate copolymers.

In certain preferred embodiments, the acrylic polymer is comprised of one or more amrnonio methacfyjate copolymers.. Ammonio methacrylate copolymers are well known in the art, and are described in NF XVII as fully polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternar ammonium groups.

In one preferred embodiment, the acrylic polymer is an acrylic resin lacquer such as that which is commercially available from Rohm Phatma unite!: the trade name Eudragit®, In further .preferred embodiments, the acrylic polymer comprises a mixture of two acrylic resin lacquers commercially available from Rohm Pharma under the trade names Eudragiti -RL30D and Eudragit S RS30D, respectively. Eudragiti) RL30D and Endragit® RS30D are copolymers of acrylic d rnefhaerylic esters with a low content of quaternar ainrftoit tt n groups, the molar ratio of ammonium .groups to the remaining neutral (meth)aerylic esters being 1:20 in Bisdragit® RI30D and 1 40 m

Eudragit® RS30D. The mean molecular weight is about 150,000. EdragifcS $- 1 0 and Eudragit® L-100 are also preferred. The code designations RL (high peTTOeafcility) and RS (lo permeability;) refer to the permeability properties of these agents, Eudragit® RL RS mixtures are insoluble in water and digestive fluids. However, multiparticulate systems forme to include the same are sweliable and permeable in aqueous solutions- and digestive fluids.

The polymers described above such as EudragitS RL RS may be mixed together in any desired ratio in order to ultimatel obtain a sustained-release fonmdatiofi having a desirable dissolution profile. Desirable sustained-release

multiparticulate systems m y be obtained, for instance, from 100%

EudragitSRL, 50% Eudragit® RL and 50% EudragitDRS, and 10%

Eudrag¾®RL and 90% Eudragit© RS. One skilled in the art will recognize that other acrylic polymers may also " be used, such as, for example, Eudragii®L,

Alternatively, extended release formulations can be prepared using osmotic systems or by applying a semi-permeable eoating to the dosage form. in the latter case, th desired drag release profile can he achieved by combining low peoneabfe arid high pertneah!s coating materials in suitable proportion.

The devices with different drug release mechanisms described above can be combined in a final dosage form comprising single or multiple units *

Examples of .multiple units include, but are no limited to, multilayer tablets and capsules containing tablets, beads, or granules, etc. An immediate release portion can be added to the extended release system by means of either applying an immediate release layer m top of the extended release core using a coating or compression process or in a multiple unit system, uch as a capsule containing extended and immediate release beads .

Extended release tablets containing liydropliilie polymers are 'prepared by techniques commonly known in the art such as direct compression., wet granulation, or dry gra rulation processes. Their fonMdations usually incorporate polymers* <fi1ue»ts* binders, and lubricants as- we l as the active pharmaceutical ingredient The asual diluents include inert powdered

substances sac ' h as starches, powdered cellulose, especially crystalline and. fflicrociysiailirie cellulose, sugars such as fructose, rnannUol and sucrose, grain flours and similar edible powders. Typical diluents include, for example, various types of starch, lactose, mannitoL. kaolin, calcium phosphate or sulfate, inorganic salts such as sodiinn chloride and powdered sugar. Powdered cellulose derivatives are also iiseliil Typical tablet binders ' include substances such as starch, gelatin and sugars s«c as lactose, fructose, and glucose. Natural and synthetic gums, racketing acacia, alginates, methvlcetlulose, and

polyvinylpyrrolidone can. also be used. Polyethylene glycol hydrophilie polymers, etbylceihilose and waxes can also serve as binders. A lubricant is necessary in a tablet fbrniulation to prevent the tablet and punches from sticking i the die. The lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.

Extended release tablets containing wa materials are generally -prepared using methods known in the art such as a. direc blend method, a congealing method, and an aqueous dispersion method, inthe congealing method, the drug is mixed with a wax material and either spray- congealed or congealed and screened and processed.

Delayed release osage forms

Delayed release formulations can be created by coating a solid dosage form with a polymer Sim, which is insoluble in the acidic environment of the stomach, and soluble in the neutral environment of the small intestine.

The delayed release dosage units can be prepared, for example, by coating a, drug or a drug^ontaming composition with a selected coating material. Th drug-containing composition, may be, e.g., a tablet for

incorporation into a capsule, a tablet for use as an inner core in a. "coated core" dosag form, or a plurality of drug-containing beads, particles o grannies, for incorporation int either a tablet or capsule. Preferred coating materials include bioerodible., gradually lwdtolyzahle s graduall water-soluble, id or

enxymatieally degradable polymers, and may he conventional "enteric" polymers. Enteric polymers, as will be app e ia ed by those skilled in the art, become soluble it the higher pH -environment of the lower gastrointestinal tract of slowly erode as the dosage -form passes through the gastrointestinal tract, while en^yniatically degradable polymers are degraded by bacterial enzymes present in the lower gas troiivtestiiial tract, particularly In the colon. Suitable coating materials for elleeiing delayed release include, bat are net limited to, cellulosic polyraers such as .hydroxypfopyl cellulose, Mydroxyemyl cellulose, hydroxymeiSiyl cellulose, nydroxypropyi methyl cellulose, bydroxypropyl methyl cellulose acetate succinate;, hydroxypropylmethyl cellulose phthalaie, methylcetl lose, ethyl cellulose, cellulose acetate, cellulose acetate p ihaiate, cellulose acetate t iraeOitate and carboxyraethylceHulose sodium;, acrylic acid polymers- a d copolymers, preferabl formed fro acrylic acid, niethaerylic acid, methyl acrylate, ethyl acry!ate, methyl methacr late and/or ethyl metlmcrylate, and other meihaor lic resins that are- commercially available under the tradename Budragit® (Roh ' Pharma* Westerstadt, Germany), including Endragit® L30D-55 and L 1,00-55 (soluble at pH 5.5 and above), E dragit® L- 100 (soluble at pH 6.0 and above); Eadragit© S (soluble at H 7,0 and above* a a result of a higher degree of esteriftca i on), and Eudragi tsig ) HE, RL and RS- (water-ifjsOloble polymers having if¾?eni degrees of permeabiliiy anil expandability); vinyl polymers and copolymers such as polyvinyl pyrrolidone, vinyl acetate s vinylacetate phthalaie, vinykcetate crotonie acid, c ol er, and ethylene-vinyl acetate copolymer; enzyroatically degradable polymers such as mo. polymers.^ pectin, chitosan, amylose and. guar gum; win and shellac.

Combinations of di fferen t coating materials may also be used. M ulti-lay er coatings usin different polymers may also be applied.

The preferred coatin weights for particular coating materials may be readily determined by those skilled in the art by evaluating individual release: profiles for tablets, beads ami granules ' prepared with different quantities of various coating materials. It is the combination of materials, method and formof application that produce the d sired release eliatacteristics, which DI ca determine only '' from the clinical studies.

The coating composi ion may include conventional dditives, such as plasiieizets, pigments, colorants, stabilizing ageats, glidants, etc. A plasticizer is ftormaliy present to reduce the fragility of the coating,, and will generally represent about 10 wt. % to 50 wt % relative t the dry weight of the polymer. Exam les of typical plastickers include polyethylene glycol, propyleue glycol, iriaeetift difflfiithyl plithalate, diethyl phthaJate, dihuty! phthalaie, dihutyl sebacaie, n ethyl citrate, tributyl citrate:, tfie ' fhyl acetyl citrate, castor oil and: acety!aied ffionogiyeerides, A stabilizing agent is preferably used to stabilize particles in the dispersion. Typical stabilizing ageats are nooionic eraulsitlers such as sorbitan esters, polysorbates and polyvinylpyrrolidone. Glidants are recommended, to reduce sticking effects during film formation and drying, and will generally represent approximately 25 wt . % to 100 ' wt. % of the polymer weight in the coating solution. One effective giidant is talc. Other glidants such as magnesium stearate and glycerol rnonosiearates may also be used. Pigments such as titanium dioxide may also be used. Small quantities of an anti-tbaming agent, such as a silicone (e.g.. simethicone), may also be added. to the coating composition,

e. Formulations for Pulmonary and Mucosal

Administration

Active agent(s) and compositions thereof can be applied formulated for pulmonary or mucosal administration,: The adraraistration can include delivery of the composition to the lungs, nasal, oral (sublingual, buccal}., vaginal or rectal mucosa.

In one embodiment the compound ate formulated for pulmonary delivery, such as intranasal administration or oral inhalation. The respiratory tract Is the structure involved In tbe exchange of gases between the atmosphere and the blood stream. The fcngs are branchin structures ultimately ending: with the alveoli where the exchange of gases occurs.: The alveolar surface area i the largest in the respiratory system and is where drug absorption occurs. The alveoli are covered by a thin epithelium without cilia or a mat us blanket and secrete surfactant phospholipids, The respiratory tract encompasses the upper airways, including the oropharynx and larynx, followed by the lower airways, which include the trachea followed by bifurcations into the bronchi and bronehioli The upper .and lower airways are called th conducting airways. The terminal bronehioli then di vide into respiratory ' bronchiole which then lead to the ultimate respirator zone, the alveoli, or deep lung. The deep lung, or alveoli,, is the primary target of inhaled therapeutic aerosols for systemic drag delivery.

Pi nronary administration of therapeutic compositions comprised of low molecular weight drugs has been observed, for example, beta-androgenic antagonists to treat asthma. Other therapeutic agents that are active in the lungs have been admin stered sysienueally and targeted via pulmonary absorption. Nasal deli very is considered to he a promising technique for administration of therapeutics for the following reasons: the nose has a large surface area available for drug absorption due to the coverage of the epithelial surface by numerous microvilli, the subepithelial layer is hi hly vascularized, the venous blood from the nose passes directly into the systemic circulation and therefore avoids the loss of drug by .first-pass metabolism in. the liver, it offers lower doses, more

suspension, for in anasat admimstration as drops or as a spray. Preferably, such solutions or suspensions are isotonic r lative to nasal secretions and of about the same ρΉ, ranging e.g., f om about pH 4,0 to out pH 1,4 or 4 from pH 6.0 to pH 7.0. Buffers should be physiologically compatible and include, simpl by way of example, phosphate buffers. For example, a representative nasal

decongestant is described as being buffered to a pH of about 6.2. One skilled in the art can readily dete m ne a su table saline content and pH for as. innocuous aqueous; solution for nasal and/or upper respiratory administration,.

Preferably^ the aqueous solution is water, physiologically acceptable aqueous solutions containing salts and or buffers, such as phosphate buffered saline (PBS), or any other aqueous solution acceptable for administration to an. anitnal or hntna . Suc -solutions are well known to a person ski lled in the art and include, but are not limited to, distilled water, de-ionized water, pure or uftrapure water, saline, phosphaie-bnr&ed saline (PBS). Other suitable aqueous vehicles include, but are not limited to, Ri ger's solution and isotonic sodium chloride. Aqueous suspensions may include- suspending agents such as cellulose derivatives, sodium alginate, polyviny!-pyrrolidone and gum -tragacanth, and a wetting agent such as lecithin. Suitable preservatives for aqueous suspensions include ethyl and tv ropyi p-hydro ybenzoaie.

In another -embodiment, sol vents that are low toxicity organic (ie, nonaqueous) class 3 residual solvents, such as ethanol, acetone., ethyl acetate, tetrali drof iran, ethyl ether, and propane! may be used for the- formulations. The solvent is selected based on its ability to readily aerosolize the formulation. The solvent should not detrimentally react with the compounds. An appropriate solvent should be used that dissolves the compounds or forms suspension of the compounds. The sol vent should be sufficiently volatile to enable formation of an aerosol of the solution or suspension. Additional solvent or aerosolizing agents , such as freons, can be added as desired to Increase the volatility of the solution o suspension.

In one embodiment compositions may contain minor amounts of polymers, surfactants, or other exeipienis well known to those of the art. In this context, "t iiior amounts* means no exeipients are present thai: night affect or mediate uptake of the compounds in due l ungs nd that the exeipients that are present are present in amount thai do not adversely affect uptake of compounds in the lungs,,

Dry li id powders can be directly disperse in ethauol because of their hydrophobic eharacter. For lipids stored in organic soiv is. such as chloromrrn, the desired, quantity of solution is placed in a vial, and. the chloroform is evaporated under a stream of nitroge to form a dry thin film on the surface of a glass vial. The film swell easily when reconstituted with ethanol To fuily disperse the lipid molecules in the organic solvent, the suspension is sonicated. Nonaqueous suspensions of lipids can also foe prepared to absolute ethanol using a reusable PARI LC Jet+ nebulize (PARI Respiratory ' Equipment, Monterey, CA).

■Dry powder formications ("DPFs") with large particle size have improved flo abiltty characteristics, such as less aggregation, easier

aerosolszat!on, and potentially less phagocytosis. Dry powder aerosols for inhalation tlierap are generally produced with mean diameters primarily in the range of less than 5 microns,, although a. preferred range is between one and ten microns in aerodynamic diameter. Large "earner" particles (containing no drug) have been co-delivered with therapeutic aerosols to aid in achieving efficient aerosolkation amon other possible benefits.

Polymeric particles ma be prepared using single and double emulsion solvent evaporation, spray drying* solvent extraction, solvent evaporation* phase separation, simple and complex coacervation,, interfkcial polymerization., and other method well known to those of ordinary skill, in the art. Particles may be made using methods for making microspheres or microcapsules; known- in the art. The preferred methods of manufacture are by spray drying and freeze drying, which, entails using a solution, containing the surlaciaat. spraying to form, droplets of the desired ske 5 and removing the solvent,

The particles may be fabricated with the appropriate material, surface roughness, diameter, and tap density for localized delivery to seiected regions of t!ie respiratory tract such as the deep lung or npper airways. For example, higher density o larger particles may be used for uppe airway delivery.

Similarly., a mixture of different -steed particles, provided with the same or different EGS .may be administered to -target different regions of the lung in one administration.

Formulations for pulmonary delivery include unilamellar phospholipid vesicles, liposomes, or lipoprotein particles. Formulations and methods of making: such foroiu!ations containing nucleic acid: are well known t one of ordinary skill in: the art. Liposomes are formed from commercially availabl phospholipids supplied by a v ariety of vendors including Avanti Polar lipids * inc. (Birmingham, Ala.), in one embodiment, the liposome can include a iigand molecule specific for a receptor on the surface of the target cell to direct the liposome to the target cell

tl Transdermal

Transdermal formulations may also he prepared. These will typically he

Ointments, lotions, sprays, or patches, all of which can be prepared using standard technology, Transdermal o ations can include penetratio enhancers.

ill* Methods of Selectively Activating A3kt3

The disclosed, compositions for selectively activating Afct3 can.be used, to modulate an imm ne response by decreasing:a ssp ressive function of nTregs, In some embodiments . , compounds according to Formula 1, Formula 11, Formula III, or Formula IV, or an. euantiorner, polymorph, or pharmaceutically acceptable salt thereof is administered systemieally. In other embodiments, compounds according to Formula I, Form la 11, Formula ill, or Formula * or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof is administered locally or regionally. For example, in some embodiments, compositions containing compounds according ?.o Formul I, Formula ίϊ,.

Formula ill, or Formula IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof -are delivered to or specifically target the tissue or organs in need -of modulation. Tregs can be modulated by targeting or deliverin the compositions to the !ynrph nodes, nTregs can be modulated by targeting or specifically delivering ' the compositions to the thymus or spleen. iTtegs can be modulated by targeting or specifically delivering the compositions to

conventional T cells outside the thymus.

In some in vim approaches, the compos Itions disclosed herein ¾re administered to a subject k. a therapeutically eilective amount As used herein the -ter "effective arao ' oat" or ' her eutically effective amount" e ns a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of the disorder being treated or to otherwise provide a desired pharinaco logic and/o physiologic effect. The precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, etc), the disease, and the treatment being effected. Exemplary symptoms, pharmacologic, and physiologic effects are discussed in more detail bel w.

In some- embodiments, the effect of the composition on a subject is compared to a control, for example, the effect of the composition on a particular symptom, pharmacologic, or physiologic indicator can be compared to an untreated subject or the condition of the subject prior to treatment. In some embodiments, the symptom, pharmacologic, or physiologic indicator is measured in a subject prior to t eatment, and again one o more times after treatment is initiated. In some embodiments, the control is a reference level, or average determined based measuring the symptom. pharmacologic, or physiologic indicator in one or more subjects that do not have the disease or condition to. be treated (e.g., healthy subjects). In some embodiments, the effect of the treatment is compared to a conventional treatment that is known the art. For example* if the disease to be treated is cancer* conventional treatment could a chemodierapeuiic agent.

In some embodiments, the immune modulating compositions disclosed herein ar administered in combination, with one or more additional active agents . The combination therapies can include administration of the acti ve agents together in. the same admixture, or in separate admixtures. Therefore, in some embodiments, the pharmaceutical composition includes two, three, or more acti ve agents, lie pharmaceutical compositions can be lorn iiiated as a pharmaceutical dosage- unit, referred, to as a unit dosage " form. Such

formulations typically include an effective amount of -one or more of the disclosed immune modulating compoimds. The different active agents can have the same, or different mechanisms of action,. In some embodiments, the combination results in. an additive effect on the treatment of the disease or disorder. In .some eWbodirrtettts, th combi ations result in a more than additive effect o tire treatment of the disease or disorder.

Preferably, the disclosed compounds and methods of use specifically acti vate the activity of Akt3 without increasing or decreasing the activity of AMI ., Akf2, or the combination thereof

A, Increasing Immune Suppressive Responses and Decreasing Immune Stimulatory Responses

h M ethods of Treatment

The disclosed compounds or an enanttomer, polymorph, or

pharmaceutically acceptable salt .thereof are useful as therapeutic agents.

Immune cells, preferabl T cells, can be contacted m wo or ex vim wit compounds according to Formula I, Formula II, Formula III. o -Formula IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereo f to decrease or inhibit immune responses including, b t not limited to inflammation. The X cells contacted wit compounds according to Formula i, Formula II, Formula III, or Formula IV . , or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof can be an immune ce ll that expresses Akt3 or has Akt3 activity and has the ability to become Foxp3+. Exemplary immune cells thai can be treated with the compounds according to Formula I, Formula If, Formula 111, or Formula IV, or an enaniioraer, polymorph, or pharmaceutically acceptable salt thereof include, but are not limited to regulatory cells such as TM, Td, Th:2S Tc2, Th3 s Th! 7, Th22: s Treg :i nTreg :? tTreg, and Ft ! cells and cells that secrete,: or cause other cells to secrete, iuflammatoiy m lec les* including^ but not limited to, 0,4 β, TNF-csi, TOF-beta, IFN-y s IL~ 17, IL-ό, IL-23., IL~ 22, 1L~ 21, and MMPs. The disclosed compounds or an enanttomer, polymorph., or phamtacemically accept ble salt thereof can also be used to increase or promot the activity or production of Tregs, increase the production: of cytokines such as IL-10 from Tregs, increase the differentiation of Tregs, increase the number, of Tregs, or increase the survival of Tregs.

The disclosed eompourtds or an. enantiomer, polymorph, or

pharmaceatically acceptabie salt thereof can be used to increase expression of FoxP3 on immune cells *

One embodiment provides a method of increasing an immune suppressive response in subject in need thereof by contacting ' immune cells ex vi vo wit the disclosed compounds, or an enantlomer ; , polymorph, or pharmaceutically acceptable salt thereof, in an amount effective to increase expression of FoxP3 on the immune cells, and administering the contacted immune cells to the subject. In one embodiment, the immune cells are autologous immune ceils. The immune cells can include T cells including but not limited to Tregs and iTregs.

in some embodiments, the disclosed compounds or an ena tiorner, polymorph, or pharmaceuti cally acceptable salt thereof are administered in combination with a second therapeutic. Combination therapies may be useful in immune .modulation, in some embodiments, the disclosed compounds o an enantiomer, polymorph, or pharmaceutically acceptable salt thereof can be used, o attenuate or reverse the acti vity of a pro-intla matory drug, and¾r limit the adverse effects of such drugs.

B, Methods of treating inflammatory responses

One embodiment provides methods for treating or alleviating one of more symptoms of .inflammation. * In a more preferred embodiment- the compositions according to Formula 1, Formula 11, Formula Hi or Formula IV and disclosed methods are useful for treating chrome and persistent

in lamm tion, intlammation in general can be treated using the compounds according to Formula I,. Formula II Formula III, or Formula IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof. Connsositions containing the disclosed compounds o -an enamionier, polymorph s or pharmaceutically acceptable salt thereof thai selecti vely increase A.kt3 activity or expression can be used to decrease art immune stimulatory response in. subject In some embodiments, the subjects have an inflammatory disease including but not limited to autoirmsune disease,

.Representative Inflammatory or autoimmune diseases and di sorders that ma he treated using disclosed compounds: or an enantiorner, polymorph-, or pharmaceutically acceptable salt thereof or compositions containing the disclosed compounds or a enantiorrier, polymorph, or phar: niaeetrtically acceptable salt thereof include, but are not limited, to, rheumatoid arthri tis, systemic lu us erythematosus., alopecia areata * ankylosing spondylitis, antiphospholipid syndrome, autoimmune Addison's disease, autoimmune hemolytic anemia, antoimiriune hepatitis, aotQinnmaie inner ear disease, autoimmune lyinphoprohferative syndrome (ALPS), autoimmune

thrombocytopenic purpura (ATP), Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac sprue-dermatitis, chronic fatigue syndrome immune deficiency, syndrome (CFiDS), chronic intlammatory dem eliiiahftg

polyneuropathy,, cicatricial pemphigoid, cold agglutinin disease * Crest syndrome, Crohn's disease, Dego's disease, derniatomyositis, dermatomyositis ~ j m mi k\ discoid 1 opus, essential niixed^ cryoglobulinemia, fibromyalgia. - fibrotnyositls. Grave's disease, ikdllairs- B r e^ Hashimoto's mytoiditis, idiopathic pulmonary fibrosis,, idiopathic thrombocytopenia purpura (ΪΤΡ), Iga nephropathy, insulin dependent diabetes (Type I), juvenile arthritis, Meniere's disease, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, obesity, pemphigus vol garis , -pernicious anemia, polyarteritis nodosa, polychondritis, polyglaocnlat syndromes, polymyalgia rheumatica, polymyositis and dennatomyositis, primary agammaglobulinemia, primary biliary cirrhosis, soriasis * yn ud's phenoinenon, Reher's s ndro e, rheumatic fever, sarcoidosis;, scleroderma,, Sjogren^ syndrome, stiff-man syndrome, Takayasu arteritis, temporal. arterttis giant cell arteritis, ' ulcerative colitis, uveitis, vasculitis, vitiligo, and Wegener's granulomatosis. 2. €¾Mfeinat¼n The apies

The disclosed com ounds or an enaniiotrjer, polymorph., or

pharmaceutically acceptable salt thereof and compositions thereof can be used alone or in combination with additional therapeutic agents. The disclosed com oun s can. be administered togedrer or in alternation with addi tlonai therapeutic agents. The additional therapeutic agents include * but are not limited to, imatimos ' upprea&ive agents (eg.,, antibodies against other- lymphocyte surface markers (e.g,, CD40, aIpha-4 kiegrln) or against cytokines), other fusion pro eins (e,g. s CTLA-4~Ig, ahatacept (C^rencia®^ TNF~av blockers such as

TNFlt-Ig, etanercept (Enhrel©)), infliximab Re.«iicade ),. cerioltzumab

(Cirnzia®) and adahmnmab (Hunura©), cyclophosphamide (CTX) (i.e.,

Endox&n®, Cytoxan®, Neosar©, Procytox®, Revinrm«ne TM ), methotrexate (MTX) (i.e.., heuraatrex®, Trexa l®), belimumab (i.e.. Benlysta©}, or other inrffiuttosuppressrve drugs (e.g, ; cyclosporin A, FK5064ike compounds, rapamycin compounds, or steroids), anti-proiiferatives, cytotoxic agents, or other compounds thai may assist m immunosuppression.

Additional immunosuppressive agents include, but. are not limited to prednisone, hudesottide ; prednisolone, cyclosporine, tacrolimus, sifoiifnns, everoliniirs, azaihioprine, leSunomide, niycophenolate, anakinra, golimuinab, ixekiznmab, natalizittmab, rituximab, secukinurnah, tocihzuniab . , ustekinuniah, V¾doiiz nmb s basi!ixlniab, daelizuniah, muromonab, of combinations thereof;

One embodiment provides an additional therapeutic agent that functions to inhibit or reduce T celt activation, through, a separate pathway. In on such embodiment, the additional therapeutic agent is a CTLA-4 fusion protein., such as CTLA-4-Ig (ahatacept). CTLA-4-Ig fusion proteins compete with the co- stimulatory receptor, CD28, on T cells for binding to CD8Q/CD86 (B?-i/B7-2) on antigen presenting cells, and thus function to inhibit T cell activation. In another embodi n , the .additional therapeutic agent is a CTLA-4- ig fusion, protein known as beiaiacept Beiaiacept contains tw amin acid substitutions (L.1.04E and A2 Y) drat -markedly increase its avidity to CD86 in vivo, in.

another embodiment, the additional therapeutic agent is-Maxy-4, hi another embodiment, the second therapeutic agent is

cyclophosphamide (CTJ Cy ophosphamide (the generic name for

Endoxan®, Cytoxan®, Neosar®, Proeyiox®, Revim une TM ), also known as cyrophosphane, is nitrogen mustard alkylating agent front the oxazophprines group, It Is used to treat various types of cancer and some autoimmune disorders < i a another embodiment, compounds of Formula I or an enantiomer, polymorph, or pharmaceutically acce ta le salt thereof arid. CDC are coadministered in effective amount to inhibit reduce, or treat a chronic

autoimm ne disease or disorder such as Systemic lupus erythematosus (SLE) hi another embodiment , the second therapeutic agent preferentially treats chronic inflammation, whereby the treatment regimen targets both acute and ch onic inflammation.

In another embodiment, th compositions according to Formula I, f ormula 11. Formula Kh of Formula IV, or an enanttoiner, polymorph, or pharmaceutically acceptable salt thereo f are used in combination, alternation, or succession with compounds that increase Treg activity or product on.

Exemplary Treg enhancing agents include but are not limited to glucocorticoid fluticasone, salmeteroal, antibodies to 1L- 12, IF -γ. and IL-4; vitamin B3, and dexamethasone, and combinations thereof.

Antibodies to other proiafJammatory xnpleoufes can also be used to combination or alternation with the disclosed compound according; to f¾tn¾nk I, Formula II, Formula III, or Formula IV, or an eaantiomer, polymorph, or pharmaceutically acceptable salt thereof, fusion proteins, or fragments thereof. Prefe d antibodies bind to 1L-6, IL-23, 1L~22 or lL-21.

Another embodiment provides a method for treating transplant rejection by administering to a subject in need thereof and effective amount of the disclosed compounds according to Formula I, Formula IT, Formula III, or Formula IV, o an enaniiom&^ polymorph, or pharmaceutically acceptable salt thereof to increase expression of foxF3 on. immune cells.

Another embodiment pro v ides a method of treating Graft-Versws-Host disease by administering to a subject in need thereof an effecti ve amount of the disclosed compounds according to Formula I, Formula IL Forrnula III, or Formula IV, or an enantiomer, po!ymotph* or pharmaceutically acceptable salt thereof to increase expression of ' FoxP3 on immnne cells.

Still another embodiment provides a method for inhibiting or reducing transplant rejection m a host in need thereof by administering to a subject in need thereof and effective amount of the disclosed compounds or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof to Increase e pnissioii of FoxP-3 os immune cells.

Another embodiment provides a method For treating chronic infection by administering to a subject i need thereof and effecti ve amount of tire disclosed compounds according to Formula 1, Formula II, Formula III, or Formula IV, or an enantioiner, polymorph, or plmotiaceuticaOy aeceptable salt thereof io increase expression of FoxP3 on immune cells.

One embodiment provides: a method ' for tre ting obesity by

administering to a subject in need thereof an effective amount of the disclosed compounds according to Formula I, Formula If Formula ill, orFormafa IV, or an enantiomer, polymorph, or pharmaceutically acceptable salt thereof to increase Akl3 activity.. Without: being bound by any one theory, it is believed that Akt3 regulates adipogenesis and that dysregulation of Akt3 signaling can lead to increased adipogenesis, obesity , and insulin resistance,

IV, . Efts- Medical kits are also disclosed. The medical kits can include, for example, a dosage supply of the disc losed compounds or an enantiomer, polymorph, o pharmaceutical ly acceptable salt thereof or compositions thereof. The disclosed compounds or an eaanfcmer, polymorph, or pharmaceutically acceptable salt thereof or compositions thereof can be supplied alone (e.g., iyophilized), or i a pharmaceutical omposition. The disclosed compounds or an. enantiomer, polymorph, or pharmaceutically acceptable sal thereof or compositions thereof can be is a unit dosage, or in a stock that shoul fee diluted prior to adminisffation. In some embodiments, the kit includes a supply of pharmaceutically acceptable carrier. The kit can also include devices for administration of the active agen†.(s) o.r ' cos¾p0s tioB($) 5 for example, syringes and .needles, The Ills can include printed instructions ' for administering the disclosed. compounds m a use as described above *

Examples

Example %i m$ 4A signit¼antry increases expression of FoxPS on iTregs, Results

Hie data .show twiiMA significantly increased expression of FoxP ' 3. on iTreg and slightl increased -proliferation of iTregs (Figures 2 A-2H). Example 2: nijJ64A increases axP3 expressie« when added darsng f reg induction,

Results

The data show that !BJJ64 iaeteased FoxP. ' j exp ession whew added daring iTre induction (Figures 3.A and 3B).

Example 3: Aki3 specifically regulates hoth types of Tregs, nT regis and iTregs,

Results

The data show that Afet3 is the key regulator of aXfegs (Figure 4A~ 4F), The suppressive activity of Tregs from Akt.3 KO mic was due to decreased levels of Inhibitory cytokines IL-IQ and TGFfi (Figure 4E and 4F). The data also show that In the absence of Akt3,.but not other - isoforms* the suppressive activity of Tregs was impaired in vivo (Figure 5A- SB). Tregs front Akt3 KO mice showed impaired suppressive activity in a RAG colitis model (Figure 5A). Additionally., the adoptive transfer of Tregs from Akt3 O tniee into Treg- depleted tumor-bearing mice show impaired suppression of ntit mor anmunity (Figure SB).

' The data also showed that Akt3 was the key regulator of iTregs (Figure 6A- 6E), Akt3 SNA, protein, and Akt3 phosphorylation were iipregu!ate in iTregs (Figure 6A- 6C). in Akt3 KO mice the conversion of Tconv cells into iTregs was significantly inhibited (Figure 6D). In addition, knocking down AtetS- froni WT Teonv cells abrogated FoxP induction in response to TGFp (Figure 6 F).

Figure TA-I.sfeo tlmt Akt teock-i as sufficient to induce Tregs as shown by FoxF3 activation.

E ample 4: mSM4A ittc eases AfctS pims &ery i ii s hitman ovarian arcliioiwa cells.

Results

The data, show that mi J64.A significaBti increase the phosphorylation of Akt3, but not Akt l or Akt2 in human ovarian carcinoma cells (Figure 8)

Example 5; mJJ en ances FoxP3 and Akt3 m Tce«v eels during fFreg induction.

Re snlts

T¾e data show that JJ A treatment increased the expression o FoxP3 and Ak†3 in Tconv cells during iXreg induction ( Figure 9 A- 9B),

Example 6: mJ¾ increases proliferation of iTreg* and nfregs

Result

lJMA treatment increased proliferation of iTregs (Figure 10A* IOC) and nTregS- (Figiiie 10D~101¾ hut not non-Treg CD4 i ut 10G~ Ι0Ϊ) and CDS (Figure I 1GL) T cells.

Exam le 7i «i<f J6 A increases su p essive .function* of jisouse iTregs and nTre ,

Results

Figure 11 A- 1 I D show thai mji¾4A treatment increased the suppressive fonetion of mouse !Treg cells in viim, rnJJ A treatmen t also increased the suppressive fenctjfon of nionse nTregs cells, m Wro and. increased nTreg proliferation without affecting their viability (Figure 12A~ 12H),. Exatn ie 8: «i3½4 e« bailees JL-I production fey uTreg,

Results

The data show feat m} ' J64A treanne¾t increased IL-10 prednctioiT y uTregs (Figiire 13 A- 13B).

Example 9; mJ.f64A iucreases T tu or growth and slg«i8cajttly ΐό crepes Tregs In tumors autf spleens of treated i e.

Results

The data, show that TC-1 tornor-bearmg mice treated with mJJ64A showed significantly increased tumor growth compared to imtreated controls (Figures 14A arid 14-B), mJJ64A also increased the number of Tregs in the tumors arid spleens of treated, mice compared to untreated controls (Figures J.5D and 1 SE). Tttnior-mfdtratioa of CDS r and FOXF3 ne CD4 T cel ls is not affected by iniJ64A ' treatment (Figures 15 A- ISC).

Example Mi IBJJ A p o ects against experimental colitis.

Results

The data, show that mJJ64A treateent protected against experimental, colitis Fisaire 16A- 161 and Figure 1?A~ I7J).. In addition, treating mice with iTre s that were treated with i«JJ64A ex viv also resulted in. protection against experimenta!. colitis (Figure 16A- 161 and Figure 17 A.- 171).

Example t i t «iJJ64A enhances the percent of Tregs In. Rag-/- mice.

Results

The data show that treating Rag-/ ' - mice with mJJ64A increased the percent of Tregs in the spleen and mesenteric lymph nodes when compared to imtreated Rag-/- mice (Figure 18A-F), Exat« jie 12: Efficacy of m.IJ64A .lit mouse EAE-nioiJeL

R suiis

« J ' J64Ated«ced disease progress* ors. a«d Increase survival rate in a mouse experimental a«*oimrautte eace l omye itrs (EAE) mode! {Figure IP A- I F), I». addition, JJ64A~iaduced iTregs also reduced disease progression and increased survival rate in the EAE model, compared to■ untreated controls (Figure 19),

Exampl 13: ifn ^£4A ' iiierea$^si ' tnd»ctto{). of ITregs wi lioirt affectiii cell viability.

Results

The data show that raiJ64A Judaead .b«ma iTtegs (Figure 2 I ) bat did mi affect, cell viability (Figure 2: 1 A).