Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION COMPOSITIONS FOR IMMUNOTHERAPY
Document Type and Number:
WIPO Patent Application WO/2016/196218
Kind Code:
A1
Abstract:
The present invention relates to therapeutic combinations comprising WNT inhibitors and methods for treating cancers using combination therapy.

Inventors:
QIN XIAOLI (US)
AN SONGZHU (US)
HUANG TAO (US)
Application Number:
PCT/US2016/034430
Publication Date:
December 08, 2016
Filing Date:
May 26, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CUREGENIX CORP (US)
International Classes:
A61K31/4375; A61K31/497; A61K39/395
Domestic Patent References:
WO2014128245A12014-08-28
WO2013185353A12013-12-19
WO2013181452A12013-12-05
WO2014128245A12014-08-28
WO2010101849A12010-09-10
WO2014165232A12014-10-09
WO2010101849A12010-09-10
WO1993016185A21993-08-19
WO1993001161A11993-01-21
WO2001014557A12001-03-01
WO2004056875A12004-07-08
WO2013079174A12013-06-06
WO2011066389A12011-06-03
WO1998008856A21998-03-05
WO2002100442A22002-12-19
WO2006117217A22006-11-09
Foreign References:
US20090217401A12009-08-27
US20140255426A12014-09-11
US20100025813W2010-03-01
US5571894A1996-11-05
US5587458A1996-12-24
US5869046A1999-02-09
EP0404097A21990-12-27
US6248516B12001-06-19
US5567610A1996-10-22
US5229275A1993-07-20
US8518404B22013-08-27
US8513390B22013-08-20
US20120237977A12012-09-20
US20120149061A12012-06-14
US20130150558A12013-06-13
US20070202100A12007-08-30
US20080311117A12008-12-18
US20090110667A12009-04-30
US6808710B12004-10-26
US7101550B22006-09-05
US7488802B22009-02-10
US7635757B22009-12-22
US7722868B22010-05-25
US8354509B22013-01-15
US8168757B22012-05-01
US8008449B22011-08-30
US20140044738A12014-02-13
US20080025980A12008-01-31
US20130022595A12013-01-24
US6803192B12004-10-12
US7105328B22006-09-12
US7943743B22011-05-17
US8779108B22014-07-15
US8552154B22013-10-08
US8246968B22012-08-21
US6403779B12002-06-11
US6399754B12002-06-04
US6225460B12001-05-01
US6127533A2000-10-03
US6031086A2000-02-29
US6005087A1999-12-21
US5977089A1999-11-02
US20030175950A12003-09-18
US20040192626A12004-09-30
US20040092470A12004-05-13
US20050020525A12005-01-27
US20050032733A12005-02-10
US6716583B22004-04-06
US6465189B12002-10-15
US6482594B22002-11-19
US6458543B12002-10-01
US6458539B12002-10-01
US6376190B12002-04-23
US6344318B12002-02-05
US6242246B12001-06-05
US6184364B12001-02-06
US6001577A1999-12-14
US5958691A1999-09-28
US5874218A1999-02-23
US5853984A1998-12-29
US5843732A1998-12-01
US5843653A1998-12-01
US5817785A1998-10-06
US5789163A1998-08-04
US5763177A1998-06-09
US5696249A1997-12-09
US5660985A1997-08-26
US5595877A1997-01-21
US5567588A1996-10-22
US5270163A1993-12-14
US20050069910A12005-03-31
US20040072234A12004-04-15
US20040043923A12004-03-04
US20030087301A12003-05-08
US20030054360A12003-03-20
US20020064780A12002-05-30
Other References:
ZHAO ET AL., JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 2, no. Suppl 3, 2014, pages 1 - 5, XP021202475
LIU ET AL.: "Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974", PNAS, vol. 110, no. 50, 2013, pages 20224 - 20229, XP055121007
ZHAO ET AL., JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 2, no. 3, 2014, pages 209
CHEN B ET AL., NATURE CHEM. BIOL., vol. 5, 2009, pages 100 - 107
MAULIK ET AL.: "Molecular Biotechnology: Therapeutic Applications and Strategies", 1997, WILEY-LISS, INC.
LILJEBLAD ET AL., GLYCO J, vol. 17, 2000, pages 323 - 329
HEELEY, ENDOCR RES, vol. 28, 2002, pages 217 - 229
HUDSON ET AL., NAT MED, vol. 9, 2003, pages 129 - 134
GREEN ET AL., NATURE GENET., vol. 113, 1994, pages 269 - 315
HOLLINGER ET AL., PROC NATL ACAD SCI USA, vol. 90, 1993, pages 6444 - 6448
MARTIN-OROZCO, N. ET AL., SEMIN. CANCER BIOL., vol. 17, no. 4, 2007, pages 288 - 298
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1983, U.S. DEPT. OF HEALTH AND HUMAN SERVICES
CHOTHIA ET AL., J MOL BIOL, vol. 196, 1987, pages 901 - 917
LONBERG ET AL., NATURE, vol. 368, 1994, pages 856
TAYLOR ET AL., INT. IMMUN., vol. 6, 1994, pages 579
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 553
JOHNSONCHISWELL, CURRENT OPINION IN STRUCTURAL BIOLOGY, vol. 3, 1993, pages 5564 - 571
ISHIDA, Y. ET AL., EMBO J., vol. 11, 1992, pages 3887 - 3895
AGATA, T. ET AL., INT. IMMUNOL., vol. 8, no. 5, 1996, pages 765 - 772
YAMAZAKI, T. ET AL., J. IMMUNOL., vol. 169, 2002, pages 5538 - 5545
NISHIMURA, H. ET AL., J. EXP. MED., vol. 192, 2000, pages 1027 - 898
BLANK, C. ET AL., IMMUNOL. IMMUNOTHER., vol. 56, no. 5, 29 December 2006 (2006-12-29), pages 739 - 745
TOPALIAN ET AL., N ENGL J MED, vol. 366, 2012, pages 2443 - 54
BRAHMER ET AL., N ENG J MED, vol. 366, 2012, pages 2455 - 65
BRAHMER ET AL., N ENG J MED, vol. 366, pages 2455 - 65
RADVANYI ET AL., CLIN CANCER RES, vol. 19, 2013, pages 5541 - 9
MENZIESLONG, THER ADV MED ONCOL, vol. 5, 2013, pages 278 - 85
BERGER ET AL., CLIN CANCER RES, vol. 14, 2008, pages 13044 - 51
BUTTE ET AL., IMMUNITY, vol. 27, 2007, pages 111
SUBUDHI ET AL., J. CLIN. INVEST., vol. 113, 2004, pages 694
TAMURA ET AL., BLOOD, vol. 97, 2001, pages 1809
XIA WLOW PS: "Late-targeted therapies for cancer", J MED CHEM., vol. 53, no. 19, 2010, pages 6811 - 24
PRIBBLE PEDELMAN MJ: "EC145: a novel targeted agent for adenocarcinoma of the lung", EXPERT OPIN. INVESTIG. DRUGS, vol. 21, 2012, pages 755 - 761
LIN ET AL., J. AM. CHEM. SOC., vol. 17, 2005, pages 4570
PELLEGRINO ET AL., SMALL, vol. 1, 2005, pages 48
MURRAY ET AL., ANN. REV. MAT. SCI., vol. 30, 2000, pages 545
TRINDADE ET AL., CHEM. MAT., vol. 13, 2001, pages 3843
"Microcapsules and Nanoparticles in Medicine and Pharmacy", 1992, CRC PRESS
MATHIOWITZ ET AL., J. CONTROL. RELEASE, vol. 5, 1987, pages 13
MATHIOWITZ ET AL., REACTIVE POLYMERS, 1987, pages 275
MATHIOWITZ ET AL., J. APPL. POLYMER SCI., vol. 35, 1988, pages 755
"Oligonucleotide synthesis: a practical approach", 1984, IRL PRESS
LIE DCCOLAMARINO SASONG HJDESIRE LMIRA HCONSIGLIO ALEIN ESJESSBERGER SLANSFORD HDEARIE AR: "WNT signalling regulates adult hippocampal neurogenesis", NATURE, vol. 437, no. 7063, 2005, pages 1370 - 5, XP055057841, DOI: 10.1038/nature04108
"Antisense Drug Technology: Principles, Strategies, and Applications", 2001, MARCEL DEKKER
AKIRI GCHERIAN MMVIJAYAKUMAR SLIU GBAFICO AAARONSON SA: "Wnt pathway aberrations including autocrine Wnt activation occur at high frequency in human non-small-cell lung carcinoma", ONCOGENE, vol. 28, no. 21, 28 May 2009 (2009-05-28), pages 2163 - 72, XP055071213, DOI: 10.1038/onc.2009.82
BAFICO ALIU GGOLDIN LHARRIS VAARONSON SA: "An autocrine mechanism for constitutive Wnt pathway activation in human cancer cells", CANCER CELL, vol. 6, no. 5, November 2004 (2004-11-01), pages 497 - 506, XP055098552, DOI: 10.1016/j.ccr.2004.09.032
BARKER NCLEVERS H: "Mining the Wnt pathway for cancer therapeutics", NAT REV DRUG DISCOV., vol. 5, no. 12, December 2006 (2006-12-01), pages 997 - 1014, XP002545380, DOI: 10.1038/NRD2154
BLOM ABVAN LENT PLVAN DER KRAAN PMVAN DEN BERG WB: "To seek shelter from the WNT in osteoarthritis? WNT-signaling as a target for osteoarthritis therapy", CURR DRUG TARGETS., vol. 11, no. 5, May 2010 (2010-05-01), pages 620 - 9
BOONEN RAVAN TIJN PZIVKOVIC D: "Wnt signaling in Alzheimer's disease: up or down, that is the question", AGEING RES REV., vol. 8, no. 2, April 2009 (2009-04-01), pages 71 - 82, XP026000080, DOI: 10.1016/j.arr.2008.11.003
CAMILLI TCWEERARATNA AT: "Striking the target in Wnt-y conditions: intervening in Wnt signaling during cancer progression", BIOCHEM PHARMACOL., vol. 80, no. 5, 1 September 2010 (2010-09-01), pages 702 - 11, XP027117556
CHAN SLCUI YVAN HASSELT ALI HSRIVASTAVA GJIN HNG KMWANG YLEE KYTSAO GS: "The tumor suppressor Wnt inhibitory factor 1 is frequently methylated in nasopharyngeal and esophageal carcinomas", LAB INVEST., vol. 87, no. 7, July 2007 (2007-07-01), pages 644 - 50
CHEN BDODGE METANG WLU JMA ZFAN CWWEI SHAO WKILGORE JWILLIAMS NS: "Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer", NAT CHEM BIOL., vol. 5, no. 2, February 2009 (2009-02-01), pages 100 - 7, XP055563246, DOI: 10.1038/nchembio.137
CHENG JHSHE HHAN YPWANG JXIONG SASAHINA KTSUKAMOTO H: "Wnt antagonism inhibits hepatic stellate cell activation and liver fibrosis", AM J PHYSIOL GASTROINTEST LIVER PHYSIOL., vol. 294, no. 1, 2008, pages 39 - 49, XP055492329, DOI: 10.1152/ajpgi.00263.2007
CHUN JSOH HYANG SPARK M: "Wnt signaling in cartilage development and degeneration", BMB REP., vol. 41, no. 7, 31 July 2008 (2008-07-31), pages 485 - 94
CHIEN AJMOON RT: "WNTS and WNT receptors as therapeutic tools and targets in human disease processes", FRONT BIOSCI., vol. 12, 1 January 2007 (2007-01-01), pages 448 - 57, XP009091014, DOI: 10.2741/2074
DEALMEIDA VIMIAO LERNST JAKOEPPEN HPOLAKIS PRUBINFELD B: "The soluble wnt receptor Frizzled-8CRD-hFc inhibits the growth of teratocarcinomas in vivo", CANCER RES., vol. 67, no. 11, 1 June 2007 (2007-06-01), pages 5371 - 9, XP002476060, DOI: 10.1158/0008-5472.CAN-07-0266
D'AMOUR KABANG AGELIAZER SKELLY OGAGULNICK ADSMART NGMOORMAN MAKROON ECARPENTER MKBAETGE EE: "Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells", NAT BIOTECHNOL., vol. 24, no. 11, November 2006 (2006-11-01), pages 1392 - 401
HERBST AKOLLIGS FT: "Wnt signaling as a therapeutic target for cancer", METHOD MOL BIOL., vol. 361, 2007, pages 63 - 91
HOEPPNER LHSECRETO FJWESTENDORF JJ: "Wnt signaling as a therapeutic target for bone diseases", EXPERT OPIN THER TARGETS., vol. 13, no. 4, April 2009 (2009-04-01), pages 485 - 96, XP055568858, DOI: 10.1517/14728220902841961
HWANG ISEO EYHA H: "Wnt/beta-catenin signaling: a novel target for therapeutic intervention of fibrotic kidney disease", ARCH PHARM RES., vol. 32, no. 12, December 2009 (2009-12-01), pages 1653 - 62
INESTROSA NCARENAS E: "Emerging roles of Wnts in the adult nervous system", NAT REV NEUROSCI., vol. 11, no. 2, February 2010 (2010-02-01), pages 77 - 86
KANSARA M ET AL.: "Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice", J CLIN INVEST., vol. 119, no. 4, April 2009 (2009-04-01), pages 837 - 51
MACDONALD BTTAMAI KHE X: "Wnt/beta-catenin signaling: components, mechanisms, and diseases", DEV CELL, vol. 17, no. l, July 2009 (2009-07-01), pages 9 - 26, XP009122315, DOI: 10.1016/j.devcel.2009.06.016
MIKELS AJNUSSE R: "Wnts as ligands: processing, secretion and reception", ONCOGENE, vol. 25, no. 57, 4 December 2006 (2006-12-04), pages 7461 - 8
MOON RT: "Wnt/beta-catenin pathway", SCI STKE., vol. 271, 2005
MORRISEY EE: "Wnt signaling and pulmonary fibrosis", AM J PATHOL., vol. 162, no. 5, May 2003 (2003-05-01), pages 1393 - 7
NUSSE R: "WNT signaling and stem cell control", CELL RES., vol. 18, no. 5, 2008, pages 523 - 7
OUCHI NHIGUCHI AOHASHI KOSHIMA YGOKCE NSHIBATA RAKASAKI YSHIMONO AWALSH K: "Sfrp5 is an anti-inflammatory adipokine that modulates metabolic dysfunction in obesity", SCIENCE, vol. 329, no. 5990, 23 July 2010 (2010-07-23), pages 454 - 7, XP055125719, DOI: 10.1126/science.1188280
REYA TCLEVERS H: "Wnt signalling in stem cells and cancer", NATURE, vol. 434, no. 7035, 14 April 2005 (2005-04-14), pages 843 - 50, XP055229280, DOI: 10.1038/nature03319
RHEE CSSEN MLU DWU CLEONI LRUBIN JCORR MCARSON DA: "Wnt and frizzled receptors as potential targets for immunotherapy in head and neck squamous cell carcinomas", ONCOGENE, vol. 21, no. 43, 26 September 2002 (2002-09-26), pages 6598 - 605, XP001191155, DOI: 10.1038/sj.onc.1205920
SULLIVAN GJ ET AL.: "Generation of functional human hepatic endoderm from human induced pluripotent stem cells", HEPATOLOGY, vol. 51, no. 1, January 2010 (2010-01-01), pages 329 - 35, XP008161307, DOI: 10.1002/hep.23335
TAKAHASHI-YANAGA FKAHN M: "Targeting Wnt signaling: can we safely eradicate cancer stem cells?", CLIN CANCER RES., vol. 16, no. 12, 15 June 2010 (2010-06-15), pages 3153 - 62, XP055183887, DOI: 10.1158/1078-0432.CCR-09-2943
TEN BERGE, D. ET AL.: "WNT signaling mediates self-organization and axis formation in embryoid bodies", CELL STEM CELL, vol. 3, 2008, pages 508 - 518, XP055297343, DOI: 10.1016/j.stem.2008.09.013
YANG LSOONPAA MHADLER EDROEPKE TKKATTMAN SJKENNEDY MHENCKAERTS EBONHAM KABBOTT GWLINDEN RM: "Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population", NATURE, vol. 453, no. 7194, 22 May 2008 (2008-05-22), pages 524 - 8, XP002561095, DOI: 10.1038/nature06894
See also references of EP 3302467A4
Attorney, Agent or Firm:
CULLMAN, Louis C. et al. (1 Park Plaza 12th Floo, Irvine California, US)
Download PDF:
Claims:
Claims

1. A combination, comprising:

(i) a therapeutically effective amount of an antagonist of Porcupine, and

(ii) a therapeutically effective amount of a PD-L/PD-1 Axis antagonist.

2. The combination of claim 1, wherein said Porcupine antagonist comprises a compound of Formula (I):

or a physiologically acceptable salt thereof, wherein

X1, X2, X3, X4, X5, X6, X7, X8 are independently CR4 or N;

Y1 is hydrogen or CR4; Y2, Y3 are independently hydrogen, halo or CR3; R1 is morpholinyl, piperazinyl, quinolinyl, , aryl, C1-6 heterocycle, 5 or 6 membered heteroaryl containing 1-2 heteroatoms selected from N, O and S; R2 is hydrogen, halo, morpholinyl, piperazinyl, quinolinyl, aryl, C1-6 heterocycle, 5 or 6

membered heteroaryl containing 1-2 heteroatoms selected from N, O and S;

R3 is hydrogen, halo, cyano, C1-6 alkyl, C1-6 alkoxy optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R4 is hydrogen, halo, C1-6alkoxy,–S(O)2R5,–C(O)OR5,–C(O)R5,–C(O)NR6R7, C1-6 alkyl, C2-6 alkenyl or C2-6 alkynyl, each of which can be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R5, R6 and R7 are independently hydrogen, C1-6 alkyl, C2-6 alkenyl or C2-6 alkynyl, each of which may be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano.

3. The combination of claim 2, wherein said 5 or 6 membered heteroaryl is selected from:

wherein,

R4 is hydrogen, halo, C1-6alkoxy,–S(O)2R5,–C(O)OR5,–C(O)R5,–C(O)NR6R7, C1-6 alkyl, C2-6 alkenyl or C2-6 alkynyl, each of which can be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R5, R6 and R7 are independently hydrogen, C1-6 alkyl, C2-6 alkenyl or C2-6 alkynyl, each of which may be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano; and

R8 is hydrogen or C1-6 alkyl.

4. The combination of claim 2 or 3, wherein R1 and R2 is independently substituted with 1 or 2 R4 groups.

5. The combination of any one of claims 2-4, wherein said compound is selected from 6-(2- methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine; 6-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)isoquinolin-1-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-1,6-naphthyridin-5-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-2-phenylpyrido[4,3-b]pyrazin-5-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-4-yl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-phenyl-2,7-naphthyridin-1-amine;

6-(3-chlorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(4-(2-(trifluoromethyl)pyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrimidin-5-yl)-2,7-naphthyridin-1-amine;

6-(5-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

6-(6-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

3-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin-3-yl)benzonitrile; 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin-3-yl)benzonitrile;

6-(4-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-m-tolyl-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-2-yl)-2,7-naphthyridin-1-amine;

6-(2-fluoropyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

6-(2-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-3-yl)-2,7-naphthyridin-1-amine;

N-(biphenyl-4-ylmethyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine;

6-(2-methylpyridin-4-yl)-N-((5-phenylpyridin-2-yl)methyl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-((2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)methyl)-2,7-naphthyridin-1-amine; N-(3-fluoro-4-(2-fluoropyridin-4-yl)benzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine; 6-(2-methylpyridin-4-yl)-N-((2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)methyl)-2,7-naphthyridin-1- amine;

N-((3-fluoro-2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin-4-yl)-2,7- naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine; N-((2'-fluoro-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine;

4-(5-(((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-yl)amino)methyl)pyridine-2-yl)thiomorpholine 1,1-dioxide;

6-(2-methylpyridin-4-yl)-N-(4-(pyridazin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridazin-4-yl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-morpholino-2,7-naphthyridin-1-amine;

6-(4-methylpiperazin-1-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

4-(8-((4-(2-methylpyridin-4-yl)benzyl)amino)-2,7-naphthyridin-3-yl)thiomorpholine 1,1-dioxide; N-(3-fluoro-4-(2-fluoropyridin-4-yl)benzyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1-amine;

N-((3-fluoro-2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)methyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1- amine;

N-((2'-fluoro-2,4'-bipyridin-5-yl)methyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

4-(5-(((6-(3-fluorophenyl)-2,7-naphthyridin-1-yl)amino)methyl)pyridine-2-yl)thiomorpholine 1,1- dioxide;

N-(4-chlorobenzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine;

N-(4-methylbenzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine;

6-(2-methylpyridin-4-yl)-N-(pyridin-3-ylmethyl)-2,7-naphthyridin-1-amine; N-benzyl-2-(3-fluorophenyl)-1,6-naphthyridin-5-amine;

2-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-1,6-naphthyridin-5-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyridin-4-yl)-1,6-naphthyridin-5-amine;

N-((6-(3-fluorophenyl)pyridin-3-yl)methyl)-2-(2-methylpyridin-4-yl)-1,6-naphthyridin-5-amine; N-(4-(2-fluoropyridin-4-yl)benzyl)-2-(2-methylpyridin-4-yl)-1,6-naphthyridin-5-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-(trifluoromethyl)pyridin-4-yl)benzyl)-1,6-naphthyridin-5-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyridin-4-yl)-1,6-naphthyridin-5-amine; N-(biphenyl-4-ylmethyl)-6-(3-fluorophenyl)isoquinolin-1-amine;

N-((2-fluorobiphenyl-4-yl)methyl)-6-(3-fluorophenyl)isoquinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-phenylisoquinolin-1-amine;

6-(3-chlorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-phenylisoquinolin-1-amine;

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-4-yl)isoquinolin-1-amine;

6-(6-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)isoquinolin-1-amine;

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-3-yl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridazin-4-yl)isoquinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyridin-2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(3-fluorophenyl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-methylpyridin-3-yl)isoquinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-phenylpyrido[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)pyrido[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)pyrido[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)pyrido[4,3-b]pyrazin-5-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-2-(3-fluorophenyl)pyrido[4,3-b]pyrazin-5-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)pyrido[4,3-b]pyrazin-5-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyridin-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-2-(2-methylpyridin-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-2-(2-methylpyridin-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-yl)-2,7-naphthyridin-1-amine;

6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

(S)-6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

(R)-6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine; 1-(4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin-3-yl)piperazin-1-yl)ethanone;

6-(1H-imidazol-1-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

6-(4-methyl-1H-imidazol-1-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(1H-tetrazol-5-yl)-2,7-naphthyridin-1-amine;

6-(5-methyl-1,3,4-oxadiazol-2-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine; 6-(1-methyl-1H-pyrazol-3-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(thiazol-5-yl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(oxazol-5-yl)-2,7-naphthyridin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-methylpyridin-3-yl)-2,7-naphthyridin-1-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine; N-((3-fluoro-2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1- amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-fluoropyridin-3-yl)-2,7-naphthyridin-1-amine; N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)-2,7-naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)-2,7-naphthyridin-1-amine;

methyl 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin-3-yl)piperazine-1-carboxylate; 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin-3-yl)piperazin-2-one;

2-(4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin-3-yl)piperazin-1-yl)acetonitrile; 2-methyl-4-(4-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-ylamino)methyl)phenyl)pyridine 1- oxide;

6-(2-chloropyridin-4-yl)-N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-2,7-naphthyridin-1-amine; 6-(2-chloropyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

2-(2-methylpyridin-4-yl)-5-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1- ylamino)methyl)benzonitrile;

N-(3-methoxy-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-amine; N-((3-chloro-2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1- amine;

2'-methyl-5-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-ylamino)methyl)-2,4'-bipyridine-3- carbonitrile; and N-(4-(2-(difluoromethyl)pyridin-4-yl)benzyl)-6-(2-methylpyridin-4-yl)-2,7- naphthyridin-1-amine, or a physiologically acceptable salt thereof.

6. The combination of claim 1, wherein said Porcupine antagonist comprises a compound of Formula (II):

or a physiologically acceptable salt thereof, wherein:

X1, X2, X3 and X4 is selected from N and CR7;

one of X5, X6, X7 and X8 is N and the others are CH;

X9 is selected from N and CH;

Z is selected from phenyl, pyrazinyl, pyridinyl, pyridazinyl and piperazinyl;

wherein each phenyl, pyrazinyl, pyridinyl, pyridazinyl or piperazinyl of Z is optionally substituted with an R6 group;

R1, R2 and R3 are hydrogen;

m is 1;

R4 is selected from hydrogen, halo, difluoromethyl, trifluoromethyl and methyl;

R6 is selected from hydrogen, halo and -C(O)Ri0; wherein R10 is methyl; and

R7 is selected from hydrogen, halo, cyano, methyl and trifluoromethyl.

7. The combination of claim 6, wherein said compound is selected from the group of:

N-[5-(3-fluorophenyl)pyridin-2-yl]-2-[5-methyl-6-(pyridazin-4-yl)pyridin-3- yl]acetamide;

2-[5-methyl-6-(2-methylpyridin-4-yl)pyridin-3-yl]-N-[5-(pyrazin-2-yl)pyridin-2- yl]acetamide (LGK974);

N-(2,3'-bipyridin-6'-yl)-2-(2',3-dimethyl-2,4'-bipyridin-5-yl)acetamide;

N-(5-(4-acetylpiperazin-1-yl)pyridin-2-yl)-2-(2'-methyl-3-(trifluoromethyl)-2,4'- bipyridin-5- yl)acetamide;

N-(5-(4-acetylpiperazin-1 -yl)pyridin-2-yl)-2-(2'-fluoro-3-methyl-2,4'-bipyridin-5- yl)acetamide; and 2-(2'-fluoro-3-methyl-2,4'-bipyridin-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide; or a pharmaceutically acceptable salt thereof.

8. The combination of f claim 6, wherein said compound is 2-[5-methyl-6-(2-methylpyridin-4- yl)pyridin-3-yl]-N-[5-(pyrazin-2-yl)pyridin-2-yl]acetamide.

9. The combination of any one of claims 1 - 8, wherein said PD-L/PD-1 Axis antagonist is selected from the group consisting of a PD-1 binding antagonist, a PD-Ll binding antagonist and a PD-L2 binding antagonist.

10. The combination of claim 9, wherein the PD-L/PD-1 Axis antagonist is a PD-1 binding antagonist.

11. The combination of claim 10, wherein the PD-1 binding antagonist inhibits the binding of PD-1 to its ligand binding partners.

12. The combination of claim 10, wherein the PD-1 binding antagonist inhibits the binding of PD-1 to PD-Ll.

13. The combination of claim 10, wherein the PD-1 binding antagonist inhibits the binding of PD-1 to PD-L2.

14. The combination of claim 10, wherein the PD-1 binding antagonist inhibits the binding of PD-1 to both PD-Ll and PD-L2.

15. The combination of claim 9, wherein the PD-1 binding antagonist is an antibody.

16. The combination of claim 15, wherein the PD-1 binding antagonist is MDX-1106, Merck 3745, CT-011, AMP-224 or AMP-514.

17. The combination of claim 9, wherein the PD-L/PD-1 Axis antagonist is a PD-L1 binding antagonist.

18. The combination of claim 17, wherein the PD-Ll binding antagonist inhibits the binding of PD-Ll to PD-1.

19. The combination of claim 17, wherein the PD-Ll binding antagonist inhibits the binding of PD-Ll to B7-l.

20. The combination of claim 17, wherein the PD-Ll binding antagonist inhibits the binding of PD-Ll to both PD-1 and B7-1.

21. The combination of claim 17, wherein the PD-L1 binding antagonist is an antibody.

22. The combination of claim 81, wherein the PD-L1 binding antagonist is selected from the group consisting of: YW243.55.S70, MPDL3280A, MDX-1105, MEDI-4736, and MSB0010718C. 23. The combination of claim 22, wherein the PD-L/PD-1 Axis antagonist is a PD-L2 binding antagonist.

24. The combination of claim 23, wherein the PD-L2 binding antagonist is an antibody.

25. The combination of claim 23, wherein the PD-L2 binding antagonist is an immunoadhesin. 26. A method for treating or delaying progression of cancer in an individual comprising administering to the individual the combination of any of claims 1-25.

27. The method of claim 26, wherein said cancer is colorectal cancer, gastric cancer, liver cancer, esophageal cancer, intestinal cancer, bile duct cancer, pancreatic cancer, endometrial cancer, or prostate cancer.

28. The method of claim 26, wherein said cancer is selected from the group consisting of: breast cancer, colorectal cancer, diffuse large B-cell lymphoma, endometrial cancer, follicular lymphoma, gastric cancer, glioblastoma, head and neck cancer, hepatocellular cancer, lung cancer, melanoma, multiple myeloma, ovarian cancer, pancreatic cancer, prostate cancer, and renal cell carcinoma.

Description:
COMBINATION COMPOSITIONS FOR IMMUNOTHERAPY

CROSS REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of, and priority to, U.S. Provisional Patent Application Serial No.: 62/168,904, filed on May 31, 2015, the entire disclosure of which is hereby incorporated by reference in its entirety.

FIELD OF THE INVENTION

[0002] The present invention relates to therapeutic combinations comprising WNT inhibitors and methods for treating cancers using combination therapy.

BACKGROUND OF THE INVENTION

[0003] WNT signaling is important to both embryogenesis and homeostasis in adult animals. The WNT pathway is comprised in general of a network of proteins that regulate the following processes: (1) the production and secretion of WNT proteins; (2) the binding of WNT with cellular receptors; and (3) the intracellular transduction of the biochemical responses triggered by the interaction (Mikels and Nusse, 2006; MacDonald, 2009; Moon, 2005).

[0004] [0001] The so-called canonical WNT pathway triggered by binding of WNT proteins to cell surface co-receptors Frizzled LRP5/6 results in a change in the amount of β-catenin that reaches the nucleus where it interacts with TCF/LEF family transcription factors to promote transcription of specific genes.

[0005] The non-canonical WNT pathway transduced by a different set of intracellular proteins controls planar cell polarity in insects and several processes such as gastrulation in vertebrates.

[0006] Diseases may arise from altered WNT pathway activity. For example, hyperactivation of the canonical WNT pathway can lead to aberrant cell growth (Reya and Clevers, 2005). Notably, 90% of colorectal cancers are initiated by the loss of the adenomatosis polyposis coli (APC) gene, a suppressor of the WNT/β-catenin pathway (Kinzler and Vogelstein, 1996). Increased expression of WNT proteins and loss of extracellular inhibitors that normally suppress WNT protein function may give rise to WNT-dependent tumors (Polakis, 2007). On the other hand, the non-canonical WNT pathway has also been shown to play a role in the progression of certain cancers (Camilli and Weeraratna, 2010). More recently, WNT signaling is also implicated in cancer stem cells (Takahashi-Yanaga and Kahn, 2010).

[0007] Evidence suggests that targeting the Wnt-mediated signal transduction pathway would be therapeutically useful in a broad range of diseases (Barker and Clevers, 2006). Mutations of APC, beta-catenin or axin-1 leading to constitutive activation of the canonical Wnt pathway are critical events in a variety of human cancers including colorectal cancer, melanoma, hepatocellular carcinoma, gastric cancer, ovarian cancer and others (Polakis, 2007). Blockade of the Wnt pathway in a variety of cancers using either genetic or chemical approaches has been shown to abrogate aberrant cell growth (Herbst and Kolligs, 2007). Furthermore, inhibition of this pathway may directly influence the cells that sustain cancer cell growth and enable metastasis, and that are thought to be resistant to traditional chemotherapeutic agents.

[0008] In addition to activation caused by mutations of gene products downstream of the receptors, aberrant Wnt pathway activity caused by other mechanisms have been associated with a broad range of cancers. These cancers include but not limited to: lung (small cell and non-small cell), breast, prostate, carcinoid, bladder, scarcinoma, esophageal, ovarian, cervical, endometrial, mesothelioma, melanoma, sarcoma, osteosarcoma, liposarcoma, thyroid, desmoids, chronic myelocytic leukemia (AML), and chronic myelocytic leukemia (CML). There are now multiple examples of cancer cells dependent upon upregulated autocrine or paracrine Wnt signaling, and cell lines from osteosarcoma, breast, head and neck and ovarian cancers have been shown to derive protection from apoptosis by autocrine or paracrine Wnt signaling (Kansara, 2009; Bafico, 2004; Akiri, 2009; DeAlmeida, 2007; Chan, 2007; Chen, 2009; Rhee, 2002).

[0009] Programmed death 1 (PD-1) is a member of the CD28 family of receptors, which includes CD28, CTLA-4, ICOS, PD-1, and BTLA. The initial members of the family, CD28 and ICOS, were discovered by functional effect on augmenting T cell proliferation following the addition of monoclonal antibodies. Two cell surface glycoprotein ligands for PD-1 have been identified, PD- L1 and PD-L2, and have been shown to downregulate T cell activation and cytokine secretion upon binding to PD-1. Both PD-L1 (B7-H1) and PD-L2 (B7-DC) are B7 homologs that bind to PD-1. PD- L1 also has an appreciable affinity for the costimulatory molecule B7-1. Upon IFN-γ stimulation, PD-L1 is expressed on T cells, NK cells, macrophages, myeloid DCs, B cells, epithelial cells, and vascular endothelial cells. The PD-L1 gene promoter region has a response element to IRF-1, the interferon regulatory factor. Type I interferons can also upregulate PD-L1 on murine hepatocytes, monocytes, DCs, and tumor cells.

[0010] PD-L1 expression has been found in several murine and human cancers, including human lung, ovarian and colon carcinoma and various myelomas. It appears that upregulation of PD-L1 may allow cancers to evade the host immune system. An analysis of tumor specimens from patients with renal cell carcinoma found that high tumor expression of PD-L1 was associated with increased tumor aggressiveness and a 4.5-fold increased risk of death. Ovarian cancer patients with higher expression of PD-L1 had a significantly poorer prognosis than those with lower expression. PD-L1 expression correlated inversely with intraepithelial CD8+ T-lymphocyte count, suggesting that PD-L1 on tumor cells may suppress antitumor CD8+ T cells. SUMMARY OF THE INVENTION

[0011] The present invention generally provides therapeutic combinations comprising WNT inhibitors and the use of such combinations for treatment of diseases, such as tumor or cancer.

[0012] In one aspect, the present invention provides a combination, comprising: (i) a therapeutically effective amount of an antagonist of Porcupine, and (ii) a therapeutically effective amount of a PD-L/PD-1 Axis antagonist.

[0013] In some embodiments, the Porcupine antagonist comprises a compound of Formula (I):

or a physiologically acceptable salt thereof, wherein

X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , X 8 are independently CR 4 or N;

Y 1 is hydrogen or CR 4 ; Y 2 , Y 3 are independently hydrogen, halo or CR 3 ; R 1 is morpholinyl, piperazinyl, quinolinyl, , aryl, C 1-6 heterocycle, 5 or 6 membered heteroaryl containing 1-2 heteroatoms selected from N, O and S; R 2 is hydrogen, halo, morpholinyl, piperazinyl, quinolinyl, aryl, C 1-6 heterocycle, 5 or 6

membered heteroaryl containing 1-2 heteroatoms selected from N, O and S;

R 3 is hydrogen, halo, cyano, C 1-6 alkyl, C 1-6 alkoxy optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R 4 is hydrogen, halo, C 1-6 alkoxy,–S(O) 2 R 5 ,–C(O)OR 5 ,–C(O)R 5 ,–C(O)NR 6 R 7 , C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl, each of which can be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R 5 , R 6 and R 7 are independently hydrogen, C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl, each of which may be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano.

[0014] In some embodiments, the 5 or 6 membered heteroaryl is selected from:

wherein,

R 4 is hydrogen, halo, C 1-6 alkoxy,–S(O) 2 R 5 ,–C(O)OR 5 ,–C(O)R 5 ,–C(O)NR 6 R 7 , C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl, each of which can be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R 5 , R 6 and R 7 are independently hydrogen, C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl, each of which may be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano; and

R 8 is hydrogen or C 1-6 alkyl.

[0015] In some embodiments, R 1 and R 2 is independently substituted with 1 or 2 R 4 groups.

[0016] In some embodiments, the compound is selected from 6-(2-methylpyridin-4-yl)-N-(4-(2- methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-amine;

N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpy ridin-4-yl)-2,7-naphthyridin-1-amine; 6-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl) isoquinolin-1-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-1,6-naphthyridin-5-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-2-phenylpyrido[4,3-b]p yrazin-5-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-4-yl)-2,7-n aphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-phenyl-2,7-naphthyri din-1-amine;

6-(3-chlorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

6-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(4-(2-(trifluoromethyl)pyridin-4-yl) benzyl)-2,7-naphthyridin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(3-fluoro phenyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrimidin-5-yl)-2,7 -naphthyridin-1-amine;

6-(5-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

6-(6-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

3-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyrid in-3-yl)benzonitrile;

4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyrid in-3-yl)benzonitrile; 6-(4-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-na phthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-m-tolyl-2,7-naphthyr idin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-ami ne;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-2-yl)-2,7-n aphthyridin-1-amine;

6-(2-fluoropyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

6-(2-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-3-yl)-2,7-n aphthyridin-1-amine;

N-(biphenyl-4-ylmethyl)-6-(2-methylpyridin-4-yl)-2,7-naph thyridin-1-amine;

6-(2-methylpyridin-4-yl)-N-((5-phenylpyridin-2-yl)methyl) -2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-((2'-(trifluoromethyl)-2,4'-bipyridi n-5-yl)methyl)-2,7-naphthyridin-1-amine; N-(3-fluoro-4-(2-fluoropyridin-4-yl)benzyl)-6-(2-methylpyrid in-4-yl)-2,7-naphthyridin-1-amine; 6-(2-methylpyridin-4-yl)-N-((2'-(trifluoromethyl)-2,4'-bipyr idin-5-yl)methyl)-2,7-naphthyridin-1- amine;

N-((3-fluoro-2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)met hyl)-6-(2-methylpyridin-4-yl)-2,7- naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpy ridin-4-yl)-2,7-naphthyridin-1-amine; N-((2'-fluoro-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin -4-yl)-2,7-naphthyridin-1-amine;

4-(5-(((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-yl)am ino)methyl)pyridine-2-yl)thiomorpholine 1,1-dioxide;

6-(2-methylpyridin-4-yl)-N-(4-(pyridazin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)-2,7-n aphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridazin-4-yl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-morpholino-2,7-napht hyridin-1-amine;

6-(4-methylpiperazin-1-yl)-N-(4-(2-methylpyridin-4-yl)ben zyl)-2,7-naphthyridin-1-amine;

4-(8-((4-(2-methylpyridin-4-yl)benzyl)amino)-2,7-naphthyr idin-3-yl)thiomorpholine 1,1-dioxide; N-(3-fluoro-4-(2-fluoropyridin-4-yl)benzyl)-6-(3-fluoropheny l)-2,7-naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(3-fluoroph enyl)-2,7-naphthyridin-1-amine;

N-((3-fluoro-2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)met hyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1- amine;

N-((2'-fluoro-2,4'-bipyridin-5-yl)methyl)-6-(3-fluorophen yl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(3-methyl-4-(2-methylpyridin-4-yl)be nzyl)-2,7-naphthyridin-1-amine;

4-(5-(((6-(3-fluorophenyl)-2,7-naphthyridin-1-yl)amino)me thyl)pyridine-2-yl)thiomorpholine 1,1- dioxide;

N-(4-chlorobenzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyri din-1-amine;

N-(4-methylbenzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyri din-1-amine;

6-(2-methylpyridin-4-yl)-N-(pyridin-3-ylmethyl)-2,7-napht hyridin-1-amine;

N-benzyl-2-(3-fluorophenyl)-1,6-naphthyridin-5-amine; 2-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl) -1,6-naphthyridin-5-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyri din-4-yl)-1,6-naphthyridin-5-amine;

N-((6-(3-fluorophenyl)pyridin-3-yl)methyl)-2-(2-methylpyr idin-4-yl)-1,6-naphthyridin-5-amine; N-(4-(2-fluoropyridin-4-yl)benzyl)-2-(2-methylpyridin-4-yl)- 1,6-naphthyridin-5-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-(trifluoromethyl)pyridin -4-yl)benzyl)-1,6-naphthyridin-5-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyr idin-4-yl)-1,6-naphthyridin-5-amine; N-(biphenyl-4-ylmethyl)-6-(3-fluorophenyl)isoquinolin-1-amin e;

N-((2-fluorobiphenyl-4-yl)methyl)-6-(3-fluorophenyl)isoqu inolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-phenylisoquin olin-1-amine;

6-(3-chlorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-phenylisoquinolin-1- amine;

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-4-yl)isoqui nolin-1-amine;

6-(6-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)isoquinolin-1-amine;

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-3-yl)isoqui nolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)isoqui nolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridazin-4-yl)isoq uinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-yl )isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin- 2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyridin- 2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(3-fluoro phenyl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-methyl pyridin-3-yl)isoquinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-phenylpyrido[ 4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)pyri do[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)pyrido[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-(3-methyl-4-(2-methylpyridin-4-yl)be nzyl)pyrido[4,3-b]pyrazin-5-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-2-(3-fluoropheny l)pyrido[4,3-b]pyrazin-5-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)pyrido[4,3-b]pyrazin-5-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyri din-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-2-(2-methylpyrid in-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-2-(2-methylpyrid in-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-y l)-2,7-naphthyridin-1-amine;

6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)benzyl) -2,7-naphthyridin-1-amine;

(S)-6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)ben zyl)-2,7-naphthyridin-1-amine;

(R)-6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)ben zyl)-2,7-naphthyridin-1-amine;

1-(4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthy ridin-3-yl)piperazin-1-yl)ethanone; 6-(1H-imidazol-1-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7- naphthyridin-1-amine;

6-(4-methyl-1H-imidazol-1-yl)-N-(4-(2-methylpyridin-4-yl) benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(1H-tetrazol-5-yl)-2 ,7-naphthyridin-1-amine;

6-(5-methyl-1,3,4-oxadiazol-2-yl)-N-(4-(2-methylpyridin-4 -yl)benzyl)-2,7-naphthyridin-1-amine; 6-(1-methyl-1H-pyrazol-3-yl)-N-(4-(2-methylpyridin-4-yl)benz yl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(thiazol-5-yl)-2,7-n aphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(oxazol-5-yl)-2,7-na phthyridin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-methyl pyridin-3-yl)-2,7-naphthyridin-1-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyr idin-4-yl)-2,7-naphthyridin-1-amine; N-((3-fluoro-2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(2-meth ylpyridin-4-yl)-2,7-naphthyridin-1- amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-fluoro pyridin-3-yl)-2,7-naphthyridin-1-amine; N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl) -2,7-naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2- yl)-2,7-naphthyridin-1-amine;

methyl 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin- 3-yl)piperazine-1-carboxylate; 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin- 3-yl)piperazin-2-one;

2-(4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthy ridin-3-yl)piperazin-1-yl)acetonitrile; 2-methyl-4-(4-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1- ylamino)methyl)phenyl)pyridine 1- oxide;

6-(2-chloropyridin-4-yl)-N-((2',3-dimethyl-2,4'-bipyridin -5-yl)methyl)-2,7-naphthyridin-1-amine; 6-(2-chloropyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)- 2,7-naphthyridin-1-amine;

2-(2-methylpyridin-4-yl)-5-((6-(2-methylpyridin-4-yl)-2,7 -naphthyridin-1- ylamino)methyl)benzonitrile;

N-(3-methoxy-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylp yridin-4-yl)-2,7-naphthyridin-1-amine; N-((3-chloro-2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(2-meth ylpyridin-4-yl)-2,7-naphthyridin-1- amine;

2'-methyl-5-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1 -ylamino)methyl)-2,4'-bipyridine-3- carbonitrile; and N-(4-(2-(difluoromethyl)pyridin-4-yl)benzyl)-6-(2-methylpyri din-4-yl)-2,7- naphthyridin-1-amine, or a physiologically acceptable salt thereof.

[0017] In some embodiments, the Porcupine antagonist comprises a compound of Formula (II):

or a physiologically acceptable salt thereof, wherein: X 1 , X 2 , X 3 and X 4 is selected from N and CR 7 ;

one of X 5 , X 6 , X 7 and X 8 is N and the others are CH;

X 9 is selected from N and CH;

Z is selected from phenyl, pyrazinyl, pyridinyl, pyridazinyl and piperazinyl;

wherein each phenyl, pyrazinyl, pyridinyl, pyridazinyl or piperazinyl of Z is optionally substituted with an R 6 group;

R 1 , R 2 and R 3 are hydrogen;

m is 1;

R 4 is selected from hydrogen, halo, difluoromethyl, trifluoromethyl and methyl;

R 6 is selected from hydrogen, halo and -C(O)R i0 ; wherein R 10 is methyl; and

R 7 is selected from hydrogen, halo, cyano, methyl and trifluoromethyl.

[0018] In some embodiments, the compound is selected from the group of:

N-[5-(3-fluorophenyl)pyridin-2-yl]-2-[5-methyl-6-(pyridaz in-4-yl)pyridin-3- yl]acetamide;

2-[5-methyl-6-(2-methylpyridin-4-yl)pyridin-3-yl]-N-[5-(p yrazin-2-yl)pyridin-2- yl]acetamide (LGK974);

N-(2,3'-bipyridin-6'-yl)-2-(2',3-dimethyl-2,4'-bipyridin- 5-yl)acetamide;

N-(5-(4-acetylpiperazin-1-yl)pyridin-2-yl)-2-(2'-methyl-3 -(trifluoromethyl)-2,4'- bipyridin-5- yl)acetamide;

N-(5-(4-acetylpiperazin-1 -yl)pyridin-2-yl)-2-(2'-fluoro-3-methyl-2,4'-bipyridin-5- yl)acetamide; and 2-(2'-fluoro-3-methyl-2,4'-bipyridin-5-yl)-N-(5-(pyrazin-2-y l)pyridin-2-yl)acetamide; or a pharmaceutically acceptable salt thereof.

[0019] In some embodiments, the compound is 2-[5-methyl-6-(2-methylpyridin-4- yl)pyridin-3- yl]-N-[5-(pyrazin-2-yl)pyridin-2-yl]acetamide.

[0020] In some embodiments, the PD-L/PD-1 Axis antagonist is selected from the group consisting of a PD-1 binding antagonist, a PD-Ll binding antagonist and a PD-L2 binding antagonist.

[0021] In some embodiments, the PD-L/PD-1 Axis antagonist is a PD-1 binding antagonist.

[0022] In some embodiments, the PD-1 binding antagonist inhibits the binding of PD-1 to its ligand binding partners.

[0023] In some embodiments, the PD-1 binding antagonist inhibits the binding of PD-1 to PD-Ll.

[0024] In some embodiments, the PD-1 binding antagonist inhibits the binding of PD-1 to PD-L2.

[0025] In some embodiments, the PD-1 binding antagonist inhibits the binding of PD-1 to both PD-Ll and PD-L2.

[0026] In some embodiments, the PD-1 binding antagonist is an antibody. [0027] In some embodiments, the PD-1 binding antagonist is MDX-1106, Merck 3745, CT-011, AMP-224 or AMP-514.

[0028] In some embodiments, the PD-L/PD-1 Axis antagonist is a PD-L1 binding antagonist.

[0029] In some embodiments, the PD-Ll binding antagonist inhibits the binding of PD-Ll to PD-1.

[0030] In some embodiments, the PD-Ll binding antagonist inhibits the binding of PD-Ll to B7-l.

[0031] In some embodiments, the PD-Ll binding antagonist inhibits the binding of PD-Ll to both PD-1 and B7-1.

[0032] In some embodiments, the PD-L1 binding antagonist is an antibody, such as one that is selected from the group consisting of: YW243.55.S70, MPDL3280A, MDX-1105, MEDI-4736, and MSB0010718C.

[0033] In some embodiments, the PD-L/PD-1 Axis antagonist is a PD-L2 binding antagonist.

[0034] In some embodiments, the PD-L2 binding antagonist is an antibody.

[0035] In some embodiments, the PD-L2 binding antagonist is an immunoadhesin.

[0036] In another aspect, the present invention provides a method for treating or delaying progression of cancer in an individual comprising administering to the individual the combination provided herein.

[0037] In some embodiments, the cancer is colorectal cancer, gastric cancer, liver cancer, esophageal cancer, intestinal cancer, bile duct cancer, pancreatic cancer, endometrial cancer, or prostate cancer.

[0038] In some embodiments, the cancer is selected from the group consisting of: breast cancer, colorectal cancer, diffuse large B-cell lymphoma, endometrial cancer, follicular lymphoma, gastric cancer, glioblastoma, head and neck cancer, hepatocellular cancer, lung cancer, melanoma, multiple myeloma, ovarian cancer, pancreatic cancer, prostate cancer, and renal cell carcinoma.

INCORPORATION BY REFERENCE

[0039] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

BRIEF DESCRIPTION OF THE DRAWINGS

[0040] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which: [0041] Figure 1 depicts tumor growth by the treatment of CGX1321, anti-PD-1 and combination of CGX1321/antiPD-1, 8 mice per group. TGI: tumor growth inhibition %.

[0042] Figure 2 depictsre 2 individual tumor growth curve of each treatment group, 8 mice per group: (A): No treatment, (B): CGX1321 treatment at 2mg/kg, (C) anti-PD-1 treatment at 10mg/kg, and (D) combination treatment of CGX1321 (2mg/kg) and anti-PD-1 antibody (10mg/kg). The fraction in each group represents the number of mice in the group showing tumor regression.

[0043] Figure 3 depicts flow cytometric analysis on T-cell population in spleen of each group at the end of treatment. Spleens were collected from four randomly selected mice per treatment group. *p<0.05 compared to vehicle group. (A): CD8+CD3+ T-cells counts of individual group, and (B): FoxP3+CD4+ T-cell population count of individual group.

DETAILED DESCRIPTION OF THE INVENTION

[0044] Several aspects of the invention are described below with reference to example applications for illustration. It should be understood that numerous specific details, relationships, and methods are set forth to provide a full understanding of the invention. One having ordinary skill in the relevant art, however, will readily recognize that the invention can be practiced without one or more of the specific details or with other methods. The present invention is not limited by the illustrated ordering of acts or events, as some acts may occur in different orders and/or concurrently with other acts or events.

[0045] Furthermore, not all illustrated acts or events are required to implement a methodology in accordance with the present invention.

[0046] The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the singular forms "a", "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the extent that the terms "including", "includes", "having", "has", "with", or variants thereof are used in either the detailed description and/or the claims, such terms are intended to be inclusive in a manner similar to the term "comprising".

[0047] The term "about" or "approximately" means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, "about" can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, "about" can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term "about" meaning within an acceptable error range for the particular value should be assumed.

I. Definitions and Abbreviations

[0048] Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry and nucleic acid chemistry and hybridization are those well known and commonly employed in the art. Standard techniques are used for nucleic acid and peptide synthesis. The techniques and procedures are generally performed according to conventional methods in the art and various general references, which are provided throughout this document. The nomenclature used herein and the laboratory procedures in analytical chemistry, and organic synthetic described below are those well- known and commonly employed in the art. Standard techniques, or modifications thereof, are used for chemical syntheses and chemical analyses.

[0049] As used herein, "WNT signaling pathway" or“WNT pathway” refers to the pathway by which binding of the WNT protein to cellular receptors results in changes of cell behavior. The WNT pathway involves a variety of proteins including Frizzled, Disheveled, Axin, APC, GSK3β, β-catenin, LEF/TCF transcription factors, and molecules involved in the synthesis and secretion of WNT proteins. Examples of proteins implicated in the secretion of functional WNTs include, but are not limited to wntless/evenness interrupted (Wls/Evi), porcupine (Porcn), and Vps35p. Wls/Evi is a 7 pass transmembrane protein which resides in the Golgi apparatus and is required for secretion of Wg (drosophila) MOM-2 (c. elegans) and Wnt3A. It contains a conserved structural motif whose structure and function are both unknown. Porcupine (Porcn) is a member of the membrane-bound O- acyltransferase (MBOAT) family of palmitoyl transferases. Fatty acid modification of Wnts is critical for their function. Wnts are palmitoylated on one or two highly conserved sites. Inhibitors of Porcn may therefore block all functional Wnt signaling. Vps35p is a subunit of a multiprotein complex called the retromer complex which is involved in intracellular protein trafficking. Vps35p functions in binding target proteins like WNTs for recruitment into vesicles.

[0050] An "Wnt inhibitor" as used herein reduces the activity of Wnt pathway. Wnt inhibitors are compounds which can inhibit the Wnt signaling pathways, and include the PORCN inhibitors. This inhibition may include, for example, inhibiting PORCN, and its palmitoylation of Wnt, or reducing the association between the Wnt pathway components including Frizzled and Disheveled. Preferably a Wnt inhibitor is a PORCN inhibitor.

[0051] The term "a method of inhibiting WNT pathway" refers to methods of inhibiting known biochemical events associated with production of functional WNT proteins or with cellular responses to WNT proteins. As discussed herein, small organic molecules may inhibit WNT response in accordance with this definition.

[0052] “WNT protein” is a protein binds to Frizzled and LRP5/6 co-receptors so as to activate canonical or non-canonical WNT signaling. Specific examples of WNT proteins include: WNT-1 (NM005430), WNT-2 (NM003391), WNT-2B/WNT-13 (NM004185), WNT-3 (NM030753), WNT3a (NM033131), WNT-4 (NM030761), WNT-5A (NM003392), WNT-5B (NM032642), WNT-6 (NM006522), WNT-7A (NM004625), WNT-7B (NM058238), WNT-8A (NM058244), WNT-8B (NM003393), WNT-9A/WNT-14) (NM003395), WNT-9B/WNT-15 (NM003396), WNT-10A (NM025216), WNT-10B (NM003394), WNT-11 (NM004626), WNT-16 (NM016087).

[0053] “WNT pathway disorder” is a condition or disease state with aberrant WNT signaling. In one aspect, the aberrant WNT signaling is a level of WNT signaling in a cell or tissue suspected of being diseased that exceeds the level of WNT signaling in a normal cell or tissue. In one specific aspect, a WNT-mediated disorder includes cancer or fibrosis.

[0054] The term“cancer” refers to the pathological condition in humans that is characterized by unregulated cell proliferation. Examples include but are not limited to: carcinoma, lymphoma, blastoma, and leukemia. More particular examples of cancers include but are not limited to: lung (small cell and non-small cell), breast, prostate, carcinoid, bladder, gastric, pancreatic, liver (hepatocellular), hepatoblastoma, colorectal, head and neck squamous cell carcinoma, esophageal, ovarian, cervical, endometrial, mesothelioma, melanoma, sarcoma, osteosarcoma, liposarcoma, thyroid, desmoids, chronic myelocytic leukemia (AML), and chronic myelocytic leukemia (CML).

[0055] The term“fibrosis” refers to the pathological condition in humans that is typically characterized by uncontrolled proliferation of fibroblast cells and tissue hardening. Specific examples include but not limited to: lung fibrosis (idiopathic pulmonary fibrosis and radiation- induced fibrosis), renal fibrosis and liver fibrosis including liver cirrhosis.

[0056] “Inhibiting” or“treating” or“treatment” refers to reduction, therapeutic treatment and prophylactic or preventative treatment, wherein the objective is to reduce or prevent the aimed pathologic disorder or condition. In one example, following administering of a WNT signaling inhibitor, a cancer patient may experience a reduction in tumor size. "Treatment" or "treating" includes (1) inhibiting a disease in a subject experiencing or displaying the pathology or symptoms of the disease, (2) ameliorating a disease in a subject that is experiencing or displaying the pathology or symptoms of the disease, and/or (3) affecting any measurable decrease in a disease in a subject or patient that is experiencing or displaying the pathology or symptoms of the disease. To the extent the WNT pathway inhibitor may prevent growth and/or kill cancer cells, it may be cytostatic and/or cytotoxic. [0057] The term“therapeutically effective amount” refers to an amount of a WNT pathway inhibitor (e.g. a Porcupine antagonist) effective to“treat” a WNT pathway disorder in a subject or mammal. In the case of cancer, the therapeutically effective amount of the drug may either reduce the number of cancer cells, reduce the tumor size, inhibit cancer cell infiltration into peripheral organs, inhibit tumor metastasis, inhibit tumor growth to certain extent, and/or relieve one or more of the symptoms associated with the cancer to some extent.

[0058] Administration“in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order. As used herein, the term “pharmaceutical combination” refers to a product obtained from mixing or combining active ingredients, and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound of Formula (1) and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound of Formula (1) and a co- agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the active ingredients in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.

[0059] A“chemotherapeutic agent” is a chemical compound useful in the treatment of cancer. Examples are but not limited to: Gemcitabine, Irinotecan, Doxorubicin, 5-Fluorouracil, Cytosine arabinoside ("Ara-C"), Cyclophosphamide, Thiotepa, Busulfan, Cytoxin, TAXOL, Methotrexate, Cisplatin, Melphalan, Vinblastine and Carboplatin.

[0060] The term“alkyl,” by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated (i.e. C 1 -C 10 means one to ten carbons). Examples of saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. An unsaturated alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers. The term“alkyl,” unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below, such as“heteroalkyl.” Alkyl groups, which are limited to hydrocarbon groups, are termed“homoalkyl”. [0061] The term“alkylene” by itself or as part of another substituent means a divalent radical derived from an alkane, as exemplified, but not limited, by–CH 2 CH 2 CH 2 CH 2 -, and further includes those groups described below as“heteroalkylene.” Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. A“lower alkyl” or“lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.

[0062] The terms“alkoxy,”“alkylamino” and“alkylthio” (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively.

[0063] The term“heteroalkyl,” by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and at least one heteroatom selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule. Examples include, but are not limited to, - CH 2 -CH 2 -O-CH 3 , -CH 2 -CH 2 -NH-CH 3 , -CH 2 -CH 2 -N(CH 3 )-CH 3 , -CH 2 -S-CH 2 -CH 3 , -CH 2 -CH 2 ,-S(O)- CH 3 , -CH 2 -CH 2 -S(O) 2 -CH 3 , -CH=CH-O-CH 3 , -Si(CH 3 ) 3 , -CH 2 -CH=N-OCH 3 , and–CH=CH-N(CH 3 )- CH 3 . Up to two heteroatoms may be consecutive, such as, for example, -CH 2 -NH-OCH 3 and–CH 2 - O-Si(CH 3 ) 3 . Similarly, the term“heteroalkylene” by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH 2 -CH 2 -S-CH 2 -CH 2 - and–CH 2 -S-CH 2 -CH 2 -NH-CH 2 -. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. For example, the formula–C(O) 2 R’- represents both–C(O) 2 R’- and–R’C(O) 2 -.

[0064] In general, an“acyl substituent” is also selected from the group set forth above. As used herein, the term“acyl substituent” refers to groups attached to, and fulfilling the valence of a carbonyl carbon that is either directly or indirectly attached to the polycyclic nucleus of the compounds of the present invention.

[0065] The terms“cycloalkyl” and“heterocycloalkyl”, by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of “alkyl” and“heteroalkyl”, respectively. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include, but are not limited to, 1–(1,2,5,6-tetrahydropyridyl), 1- piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1–piperazinyl, 2-piperazinyl, and the like.

[0066] The terms“halo” or“halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl. For example, the term“halo(C 1 - C 4 )alkyl” is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.

[0067] The term“aryl” means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent, which can be a single ring or multiple rings (preferably from 1 to 3 rings), which are fused together or linked covalently. The term“heteroaryl” refers to aryl groups (or rings) that contain from one to four heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heteroaryl group can be attached to the remainder of the molecule through a heteroatom. Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3- pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4- oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2- furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5- benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5- quinoxalinyl, 3-quinolyl, and 6-quinolyl. Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below.

[0068] For brevity, the term“aryl” when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, the term “arylalkyl” is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2- pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like).

[0069] Each of the above terms (e.g.,“alkyl,”“heteroalkyl,”“aryl” and“heteroaryl”) include both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below.

[0070] Substituents for the alkyl, and heteroalkyl radicals (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) are generally referred to as“alkyl substituents” and“heteroakyl substituents,” respectively, and they can be one or more of a variety of groups selected from, but not limited to: -OR’, =O, =NR’, =N-OR’, -NR’R”, -SR’, -halogen, -SiR’R”R”’, -OC(O)R’, -C(O)R’, -CO 2 R’, -CONR’R”, -OC(O)NR’R”, -NR”C(O)R’, -NR’-C(O)NR”R”’, -NR”C(O) 2 R’, -NR-C(NR’R”R’”)=NR””, -NR-C(NR’R”)=NR’”, -S(O)R’, -S(O) 2 R’, -S(O) 2 NR’R”, -NRSO 2 R’, -CN and–NO 2 in a number ranging from zero to (2m’+1), where m’ is the total number of carbon atoms in such radical. R’, R”, R”’ and R”” each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, e.g., aryl substituted with 1-3 halogens, substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups. When a compound of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R’, R”, R’” and R”” groups when more than one of these groups is present. When R’ and R” are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 5-, 6-, or 7-membered ring. For example, -NR’R” is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term“alkyl” is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., -CF 3 and–CH 2 CF 3 ) and acyl (e.g., -C(O)CH 3 , -C(O)CF 3 , -C(O)CH 2 OCH 3 , and the like).

[0071] Similar to the substituents described for the alkyl radical, the aryl substituents and heteroaryl substituents are generally referred to as“aryl substituents” and“heteroaryl substituents,” respectively and are varied and selected from, for example: halogen, -OR’, =O, =NR’, =N-OR’, - NR’R”, -SR’, -halogen, -SiR’R”R”’, -OC(O)R’, -C(O)R’, -CO 2 R’, -CONR’R”, -OC(O)NR’R”, -NR”C(O)R’, -NR’-C(O)NR”R”’, -NR”C(O) 2 R’, -NR-C(NR’R”)=NR’”, -S(O)R’, -S(O) 2 R’, -S(O) 2 NR’R”, -NRSO 2 R’, -CN and–NO 2 , -R’, -N 3 , -CH(Ph) 2 , fluoro(C 1 -C 4 )alkoxy, and fluoro(C 1 -C 4 )alkyl, in a number ranging from zero to the total number of open valences on the aromatic ring system; and where R’, R”, R”’ and R”” are preferably independently selected from hydrogen, (C 1 -C 8 )alkyl and heteroalkyl, unsubstituted aryl and heteroaryl, (unsubstituted aryl)-(C 1 - C 4 )alkyl, and (unsubstituted aryl)oxy-(C 1 -C 4 )alkyl. When a compound of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R’, R”, R’” and R”” groups when more than one of these groups is present.

[0072] Two of the aryl substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula –T-C(O)-(CRR’) q -U-, wherein T and U are independently–NR-, -O-, -CRR’- or a single bond, and q is an integer of from 0 to 3. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula–A-(CH 2 ) r -B-, wherein A and B are independently–CRR’-, -O-, - NR-, -S-, -S(O)-, -S(O) 2 -, -S(O) 2 NR’- or a single bond, and r is an integer of from 1 to 4. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula–(CRR’) s -X-(CR”R’”) d -, where s and d are independently integers of from 0 to 3, and X is–O-, -NR’-, -S-, -S(O)-, -S(O) 2 -, or–S(O) 2 NR’-. The substituents R, R’, R” and R’” are preferably independently selected from hydrogen or substituted or unsubstituted (C 1 -C 6 ) akyl.

[0073] As used herein, the term“heteroatom” includes oxygen (O), nitrogen (N), sulfur (S), phosphorus (P) and silicon (Si).

II. The Compositions

[0074] In general, the present invention provides therapeutic combinations, pharmaceutical compostions, and methods for treating cancers using combination therapy.

[0075] In one aspect, the present invention provide therapeutic combinations comprising: (i) an effective amount of a therapeutically effective amount of WNT inhibitor (such as an antagonist of Porcupine), and (ii) an effective amount of a PD-L/PD-1 Axis antagonist.

[0076] A therapeutic combination may be provided in a single pharmaceutical composition so that both the WNT inhibitor and the PD-L/PD-1 Axis antagonist compound can be administered together. In alternative embodiments, a therapeutic combination may be provided using more than one pharmaceutical composition. In such embodiments, a WNT inhibitor compound may be provided in one pharmaceutical composition and a PD-L/PD-1 Axis antagonist compound may be provided in a second pharmaceutical composition so that the two compounds can be administered separately such as, for example, at different times, by different routes of administration, and the like. Thus, it also may be possible to provide the WNT inhibitor compound and the PD-L/PD-1 Axis antagonist compound in different dosing regimens.

[0077] Unless otherwise indicated, reference to a compound can include the compound in any pharmaceutically acceptable form, including any isomer (e.g., diastereomer or enantiomer), salt, solvate, polymorph, and the like. In particular, if a compound is optically active, reference to the compound can include each of the compound's enantiomers as well as racemic mixtures of the enantiomers.

A. WNT Inhibitors

[0078] In some embodiments, the Wnt inhibitor is a Porcupine inhibitor suitable for use in human. The Wnt inhibitor may be a Porcupine inhibitor that has a function similar to a known Porcupine inhibitor such as IWP-2, IWP-3 or IWP-4, which are described by Chen B et al. (2009) Nature Chem. Biol. 5: 100-107 and commercially available from Miltenyi Biotech as Stemolecule™ Wnt Inhibitor IWP-2 (#130-095-584), Stemolecule™ Wnt Inhibitor IWP-3 (#130-095-585) and Stemolecule™ Wnt Inhibitor IWP-4. Stemolecule™ IWP-2, Stemolecule™ IWP-3, and Stemolecule™ IWP-4 prevent palmitylation of Wnt proteins by Porcupine (PORCN), a membrane-bound O- acyltransferase. [0079] Alternatively, Wnt inhibitors can be the products of drug design and can be produced using various methods known in the art. See, international patent application WO2010/101849, published 10 September 2010. Various methods of drug design, useful to design mimetics or other compounds useful in the invention are disclosed in Maulik ef al. (1997) Molecular Biotechnology: Therapeutic Applications and Strategies. Wiley-Liss, Inc. (incorporated by reference in its entirety). A Wnt inhibitor can be obtained from molecular diversity strategies (a combination of related strategies allowing the rapid construction of large, chemically diverse molecule libraries), libraries of natural or synthetic compounds, in particular from chemical or combinatorial libraries (i.e., libraries of compounds that differ in sequence or size but that have the similar building blocks) or by rational, directed or random drug design. See, for example, Maulik et al. (1997) Molecular Biotechnology: Therapeutic Applications and Strategies. Wiley-Liss, Inc.. In a molecular diversity strategy, large compound libraries are synthesized, for example, from peptides, oligonucleotides, natural or synthetic steroidal compounds, carbohydrates or natural or synthetic organic and non-steroidal molecules, using biological, enzymatic or chemical approaches. The critical parameters in developing a molecular diversity strategy include subunit diversity, molecular size, and library diversity. The general goal of screening such libraries is to utilize sequential application of combinatorial selection to obtain high- affinity ligands for a desired target, and then to optimize the lead molecules by either random or directed design strategies. Methods of molecular diversity are described in detail in Maulik ef al. (1997) Molecular Biotechnology: TherapeuticApplications and Strategies. Wiley-Liss, Inc.

[0080] In another aspect, the present invention provides a compound as Porcupine antagonist or inhibitor.

[0081] By“PORCN” herein is meant Porcupine, a membrane-bound acyltransferase, required for Wnt post-translational modification. Unless specifically stated otherwise, PORCN as used herein, refers to human PORCN-accession numbers NM_017617.3/NP_060087.

[0082] Wnt Signaling in dentric cells (DCs) leads to immunosuppression. Tumors activate Wnt signaling in DCs through secreting Wnt ligands (or potentially other soluble factors) into their microenvironment. Constitutive Wnt signaling in DCs is also a major mechanism in healthy intestine for maintaining mucosal tolerance to food, commensal microorganisms and self-antigens. Wnt signaling in DCs include: (1) upregulating the expression of immunosuppressive indoleamine 2,3- dioxygenase (IDO) in DCs; (2) promoting indoleamine 2,3-dioxygenase (IDO)-dependent development of immunosuppressive regulatory T cells (Tregs); and (3) suppressing DCs’ ability to prime tumor antigen-specific CD8+ effector T cells.

[0083] In some embodiments, the Porcupine inhibitor has the structure of Formula (I):

or a physiologically acceptable salt thereof, wherein,

X1, X2, X3, X4, X5, X6, X7, X8 are independently CR4 or N

Y 1 is hydrogen or CR 4 ;

Y 2 , Y 3 are independently hydrogen, halo or CR 3 ; R 1 is morpholinyl, piperazinyl, quinolinyl, , aryl, C 1-6 heterocycle, 5 or 6 membered heteroaryl containing 1-2 heteroatoms selected from N, O and S; R 2 is hydrogen, halo, morpholinyl, piperazinyl, quinolinyl, , aryl, C 1-6 heterocycle, 5 or 6 membered heteroaryl containing 1-2 heteroatoms selected from N, O and S;

wherein 5 or 6 membered heteroar l includes the followin selected rou s but is not limited to:

R 1 and R 2 could be independently and optionally substituted with 1-2 R 4 groups;

R 3 is hydrogen, halo, cyano, C 1-6 alkyl, C 1-6 alkoxy optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R 4 is hydrogen, halo, C 1-6 alkoxy,–S(O) 2 R 5 ,–C(O)OR 5 ,–C(O)R 5 ,–C(O)NR 6 R 7 , C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl, each of which can be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R 5 , R 6 and R 7 are independently hydrogen, C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl, each of which may be optionally substituted with halo, amino, hydroxyl, alkoxy or cyano;

R 8 is hydrogen or C 1-6 alkyl. [0084] As used herein, an H atom in any substituent groups (e.g., CH 2 ) encompasses all suitable isotopic variations, e.g., H, 2 H and 3 H.

[0085] As used herein, other atoms in any substituent groups encompasses all suitable isotopic variations, including but not limited to 11C, 13C, 14C, 15N, 17O, 18O, 35S, 18F, 36I and/or 123I.

[0086] In some embodiments, example of the compound of the invention includes but is not limited to:

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpy ridin-4-yl)-2,7-naphthyridin-1-amine; 6-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)methyl) isoquinolin-1-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-1,6-naphthyridin-5-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-2-phenylpyrido[4,3-b]p yrazin-5-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-4-yl)-2,7-n aphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-phenyl-2,7-naphthyri din-1-amine;

6-(3-chlorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

6-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(4-(2-(trifluoromethyl)pyridin-4-yl) benzyl)-2,7-naphthyridin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(3-fluoro phenyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrimidin-5-yl)-2,7 -naphthyridin-1-amine;

6-(5-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

6-(6-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

3-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyrid in-3-yl)benzonitrile;

4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyrid in-3-yl)benzonitrile;

6-(4-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-m-tolyl-2,7-naphthyr idin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-2,7-naphthyridin-1-ami ne;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-2-yl)-2,7-n aphthyridin-1-amine;

6-(2-fluoropyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)-2,7-naphthyridin-1-amine;

6-(2-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-3-yl)-2,7-n aphthyridin-1-amine;

N-(biphenyl-4-ylmethyl)-6-(2-methylpyridin-4-yl)-2,7-naph thyridin-1-amine;

6-(2-methylpyridin-4-yl)-N-((5-phenylpyridin-2-yl)methyl) -2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-((2'-(trifluoromethyl)-2,4'-bipyridi n-5-yl)methyl)-2,7-naphthyridin-1-amine; N-(3-fluoro-4-(2-fluoropyridin-4-yl)benzyl)-6-(2-methylpyrid in-4-yl)-2,7-naphthyridin-1-amine; 6-(2-methylpyridin-4-yl)-N-((2'-(trifluoromethyl)-2,4'-bipyr idin-5-yl)methyl)-2,7-naphthyridin-1- amine; N-((3-fluoro-2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)methyl )-6-(2-methylpyridin-4-yl)-2,7- naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylpy ridin-4-yl)-2,7-naphthyridin-1-amine; N-((2'-fluoro-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyridin -4-yl)-2,7-naphthyridin-1-amine;

4-(5-(((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1-yl)am ino)methyl)pyridine-2-yl)thiomorpholine 1,1-dioxide;

6-(2-methylpyridin-4-yl)-N-(4-(pyridazin-4-yl)benzyl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)-2,7-n aphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridazin-4-yl)-2,7 -naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-morpholino-2,7-napht hyridin-1-amine;

6-(4-methylpiperazin-1-yl)-N-(4-(2-methylpyridin-4-yl)ben zyl)-2,7-naphthyridin-1-amine;

4-(8-((4-(2-methylpyridin-4-yl)benzyl)amino)-2,7-naphthyr idin-3-yl)thiomorpholine 1,1-dioxide; N-(3-fluoro-4-(2-fluoropyridin-4-yl)benzyl)-6-(3-fluoropheny l)-2,7-naphthyridin-1-amine;

N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(3-fluoroph enyl)-2,7-naphthyridin-1-amine;

N-((3-fluoro-2'-(trifluoromethyl)-2,4'-bipyridin-5-yl)met hyl)-6-(3-fluorophenyl)-2,7-naphthyridin-1- amine;

N-((2'-fluoro-2,4'-bipyridin-5-yl)methyl)-6-(3-fluorophen yl)-2,7-naphthyridin-1-amine;

6-(3-fluorophenyl)-N-(3-methyl-4-(2-methylpyridin-4-yl)be nzyl)-2,7-naphthyridin-1-amine;

4-(5-(((6-(3-fluorophenyl)-2,7-naphthyridin-1-yl)amino)me thyl)pyridine-2-yl)thiomorpholine 1,1- dioxide;

N-(4-chlorobenzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyri din-1-amine;

N-(4-methylbenzyl)-6-(2-methylpyridin-4-yl)-2,7-naphthyri din-1-amine;

6-(2-methylpyridin-4-yl)-N-(pyridin-3-ylmethyl)-2,7-napht hyridin-1-amine;

N-benzyl-2-(3-fluorophenyl)-1,6-naphthyridin-5-amine;

2-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)-1,6-naphthyridin-5-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyri din-4-yl)-1,6-naphthyridin-5-amine;

N-((6-(3-fluorophenyl)pyridin-3-yl)methyl)-2-(2-methylpyr idin-4-yl)-1,6-naphthyridin-5-amine; N-(4-(2-fluoropyridin-4-yl)benzyl)-2-(2-methylpyridin-4-yl)- 1,6-naphthyridin-5-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-(trifluoromethyl)pyridin -4-yl)benzyl)-1,6-naphthyridin-5-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyr idin-4-yl)-1,6-naphthyridin-5-amine; N-(biphenyl-4-ylmethyl)-6-(3-fluorophenyl)isoquinolin-1-amin e;

N-((2-fluorobiphenyl-4-yl)methyl)-6-(3-fluorophenyl)isoqu inolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-phenylisoquin olin-1-amine;

6-(3-chlorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-phenylisoquinolin-1- amine;

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-4-yl)isoqui nolin-1-amine; 6-(6-methylpyridin-3-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)i soquinolin-1-amine;

6-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)isoquinolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridin-3-yl)isoqui nolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl)isoqui nolin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(pyridazin-4-yl)isoq uinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-yl )isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin- 2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyridin- 2-yl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(3-fluoro phenyl)isoquinolin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-methyl pyridin-3-yl)isoquinolin-1-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-phenylpyrido[ 4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-(4-(2-methylpyridin-4-yl)benzyl)pyri do[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-((2'-methyl-2,4'-bipyridin-5-yl)meth yl)pyrido[4,3-b]pyrazin-5-amine;

2-(3-fluorophenyl)-N-(3-methyl-4-(2-methylpyridin-4-yl)be nzyl)pyrido[4,3-b]pyrazin-5-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-2-(3-fluoropheny l)pyrido[4,3-b]pyrazin-5-amine;

2-(2-methylpyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzy l)pyrido[4,3-b]pyrazin-5-amine;

N-((2'-methyl-2,4'-bipyridin-5-yl)methyl)-2-(2-methylpyri din-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-2-(2-methylpyrid in-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-2-(2-methylpyrid in-4-yl)pyrido[4,3-b]pyrazin-5-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(pyrazin-2-y l)-2,7-naphthyridin-1-amine;

6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)benzyl) -2,7-naphthyridin-1-amine;

(S)-6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)ben zyl)-2,7-naphthyridin-1-amine;

(R)-6-(2-methylmorpholino)-N-(4-(2-methylpyridin-4-yl)ben zyl)-2,7-naphthyridin-1-amine;

1-(4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthy ridin-3-yl)piperazin-1-yl)ethanone; 6-(1H-imidazol-1-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)-2,7- naphthyridin-1-amine;

6-(4-methyl-1H-imidazol-1-yl)-N-(4-(2-methylpyridin-4-yl) benzyl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(1H-tetrazol-5-yl)-2 ,7-naphthyridin-1-amine;

6-(5-methyl-1,3,4-oxadiazol-2-yl)-N-(4-(2-methylpyridin-4 -yl)benzyl)-2,7-naphthyridin-1-amine; 6-(1-methyl-1H-pyrazol-3-yl)-N-(4-(2-methylpyridin-4-yl)benz yl)-2,7-naphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(thiazol-5-yl)-2,7-n aphthyridin-1-amine;

N-(4-(2-methylpyridin-4-yl)benzyl)-6-(oxazol-5-yl)-2,7-na phthyridin-1-amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-methyl pyridin-3-yl)-2,7-naphthyridin-1-amine; N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(2-methylpyr idin-4-yl)-2,7-naphthyridin-1-amine; N-((3-fluoro-2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(2-meth ylpyridin-4-yl)-2,7-naphthyridin-1- amine;

N-((2',3-dimethyl-2,4'-bipyridin-5-yl)methyl)-6-(5-fluoro pyridin-3-yl)-2,7-naphthyridin-1-amine; N-(3-methyl-4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl) -2,7-naphthyridin-1-amine; N-(3-fluoro-4-(2-methylpyridin-4-yl)benzyl)-6-(pyrazin-2-yl) -2,7-naphthyridin-1-amine;

methyl 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin- 3-yl)piperazine-1-carboxylate; 4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthyridin- 3-yl)piperazin-2-one;

2-(4-(8-(4-(2-methylpyridin-4-yl)benzylamino)-2,7-naphthy ridin-3-yl)piperazin-1-yl)acetonitrile; 2-methyl-4-(4-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1- ylamino)methyl)phenyl)pyridine 1- oxide;

6-(2-chloropyridin-4-yl)-N-((2',3-dimethyl-2,4'-bipyridin -5-yl)methyl)-2,7-naphthyridin-1-amine; 6-(2-chloropyridin-4-yl)-N-(4-(2-methylpyridin-4-yl)benzyl)- 2,7-naphthyridin-1-amine;

2-(2-methylpyridin-4-yl)-5-((6-(2-methylpyridin-4-yl)-2,7 -naphthyridin-1- ylamino)methyl)benzonitrile;

N-(3-methoxy-4-(2-methylpyridin-4-yl)benzyl)-6-(2-methylp yridin-4-yl)-2,7-naphthyridin-1-amine; N-((3-chloro-2'-methyl-2,4'-bipyridin-5-yl)methyl)-6-(2-meth ylpyridin-4-yl)-2,7-naphthyridin-1- amine;

2'-methyl-5-((6-(2-methylpyridin-4-yl)-2,7-naphthyridin-1 -ylamino)methyl)-2,4'-bipyridine-3- carbonitrile; and N-(4-(2-(difluoromethyl)pyridin-4-yl)benzyl)-6-(2-methylpyri din-4-yl)-2,7- naphthyridin-1-amine; or physiologically acceptable salts thereof.

[0087] In some embodiments, examples of the compound of the invention include but are not limited to the compounds provided herein in Examples 1-5 of WO2014165232A1, the disclosure of which is incorporated by reference in its entirety, and Table 1. A person skilled in the art can clearly understand and know that the other compounds could be prepared by the same strategy as Examples 1-5 of WO2014165232A1.

Table 1 Compounds Table

[0088] In some embodiments, the Porcupine antagonist or inhibtor used for the treatment as described herein is any suitable compound as disclosed in the WO2010/101849 A1 (PCT/US10/025813), referabl a com ound of Formula II :

or a physiologically acceptable salt thereof, wherein:

X 1 , X 2 , X 3 and X 4 is selected from N and CR 7 ;

one of X 5 , X 6 , X 7 and X 8 is N and the others are CH;

X 9 is selected from N and CH;

Z is selected from phenyl, pyrazinyl, pyridinyl, pyridazinyl and piperazinyl;

wherein each phenyl, pyrazinyl, pyridinyl, pyridazinyl or piperazinyl of Z is optionally substituted with an R 6 group;

R 1 , R 2 and R 3 are hydrogen;

m is 1 ;

R 4 is selected from hydrogen, halo, difluoromethyl, trifluoromethyl and methyl;

R 6 is selected from hydrogen, halo and -C(O)R i0 ; wherein R 10 is methyl; and

R 7 is selected from hydrogen, halo, cyano, methyl and trifluoromethyl.

[0089] In some embodiments, the compound is selected from the group consisiting of:

N-[5-(3-fluorophenyl)pyridin-2-yl]-2-[5-methyl-6-(pyridazin- 4-yl)pyridin-3- yl]acetamide;

2-[5-methyl-6-(2-methylpyridin-4-yl)pyridin-3-yl]-N-[5-(pyra zin-2-yl)pyridin-2- yl]acetamide (LGK974); N-(2,3'-bipyridin-6'-yl)-2-(2',3-dimethyl-2,4'-bipyridin-5-y l)acetamide;

N-(5-(4-acetylpiperazin-1-yl)pyridin-2-yl)-2-(2'-methyl-3 -(trifluoromethyl)-2,4'- bipyridin-5- yl)acetamide;

N-(5-(4-acetylpiperazin-1 -yl)pyridin-2-yl)-2-(2'-fluoro-3-methyl-2,4'-bipyridin-5- yl)acetamide; and 2-(2'-fluoro-3-methyl-2,4'-bipyridin-5-yl)-N-(5-(pyrazin-2-y l)pyridin-2-yl)acetamide; or a pharmaceutically acceptable salt thereof.

[0090] In some embodiments, thecompound is 2-[5-methyl-6-(2-methylpyridin-4- yl)pyridin-3- yl]-N-[5-(pyrazin-2-yl)pyridin-2-yl]acetamide.

B. PD-L/PD-1 Axis Antagonists

[0091] In general, the combination provided herein comprises an entity, such as a PD-L/PD-1 Axis antagonist that is capable of specifically binding to a particular target, such as PD-L1, PD-L2 or PD-1. The entity is capable of binding to PD-L1, PD-L2, or PD-1 specifically or preferably in comparison to a non-target.

[0092] By“specifically binds” or“preferably binds” herein is meant that the binding between two binding partners (e.g., between a targeting moiety and its binding partner) is selective for the two binding partners and can be discriminated from unwanted or non-specific interactions. For example, the ability of an antigen-binding moiety to bind to a specific antigenic determinant can be measured either through an enzyme- linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance technique (analyzed on a BIAcore instrument) (Liljeblad et al., Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)). The terms“anti- [antigen] antibody” and“an antibody that binds to [antigen]” refer to an antibody that is capable of binding the respective antigen with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting the antigen. In some embodiments, the extent of binding of an anti- [antigen] antibody to an unrelated protein is less than about 10% of the binding of the antibody to the antigen as measured, e.g., by a radioimmunoassay (RIA). In some embodiments, an antibody that binds to [antigen] has a dissociation constant (KD) of < ΙμΜ, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM (e.g. 10 -8 M or less, e.g. from 10 -8 M to 10 -13 M, e.g., from 10 -9 M to 10 -13 M). It is understood that the above definition is also applicable to antigen-binding moieties that bind to an antigen.

[0093] By“PD-L/PD-1 Axis antagonist” herein is meant is a molecule that inhibits the interaction of a PD-L/PD-1 axis binding partner with either one or more of its binding partner, so as to remove T- cell dysfunction resulting from signaling on the PD-L/PD-1 signaling axis - with a result being to restore or enhance T-cell function {e.g., proliferation, cytokine production, target cell killing). As used herein, a PD-L/PD-1 Axis antagonist includes a PD-1binding antagonist, a PD-L1 binding antagonist and a PD-L2 binding antagonist. [0094] By“PD-1 binding antagonists” herein is meant a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-1 with one or more of its binding partners, such as PD-L1, PD-L2. In some embodiments, the PD-1 binding antagonist is a molecule that inhibits the binding of PD-1 to its binding partners. In a specific aspect, the PD-1 binding antagonist inhibits the binding of PD-1 to PD-L1 and/or PD-L2. For example, PD-1 binding antagonists include anti-PD-1 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-1 with PD-L1 and/or PD-L2. In one embodiment, a PD-1 binding antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-1 so as render a dysfunctional T-cell less dysfunctional (e.g., enhancing effector responses to antigen recognition). In some embodiments, the PD-1 binding antagonist is an anti-PD-1 antibody. In a specific aspect, a PD-1 binding antagonist is MDX-1106 described herein. In another specific aspect, a PD-1 binding antagonist is Merck 3745 described herein. In another specific aspect, a PD-1 binding antagonist is CT-011 described herein.

[0095] By“PD-Ll binding antagonists” herein is meants a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-L1 with either one or more of its binding partners, such as PD-1, B7-1. In some embodiments, a PD-L1 binding antagonist is a molecule that inhibits the binding of PD-L1 to its binding partners. In a specific aspect, the PD-L1 binding antagonist inhibits binding of PD-L1 to PD-1 and/or B7-1. In some embodiments, the PD-L1 binding antagonists include anti-PD-L1 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L1 with one or more of its binding partners, such as PD-1, B7-1. In one embodiment, a PD-L1 binding antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L1 so as to render a dysfunctional T-cell less dysfunctional (e.g., enhancing effector responses to antigen recognition). In some embodiments, a PD-L1 binding antagonist is an anti-PD-L1 antibody. In a specific aspect, an anti-PD-L1l antibody is YW243.55.S70 described herein. In another specific aspect, an anti-PD-Ll antibody is MDX-1105 described herein. In still another specific aspect, an anti-PD-L1 antibody is MPDL3280A described herein.

[0096] By“PD-L2 binding antagonists” herein is meant a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-1. In some embodiments, a PD-L2 binding antagonist is a molecule that inhibits the binding of PD-L2 to its binding partners. In a specific aspect, the PD- L2 binding antagonist inhibits binding of PD-L2 to PD-1. In some embodiments, the PD-L2 antagonists include anti-PD-L2 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-1. In one embodiment, a PD-L2 binding antagonist reduces the negative co- stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L2 so as render a dysfunctional T-cell less dysfunctional (e.g., enhancing effector responses to antigen recognition). In some embodiments, a PD-L2 binding antagonist is an immunoadhesin.

Antibodies

[0097] In some embodiments, the targeting moiety comprises an antibody, or a functional fragment thereof.

[0098] By“immunoglobulin” or“antibody” herein is meant a full-length (i.e., naturally occurring or formed by normal immunoglobulin gene fragment recombinatorial processes) immunoglobulin molecule (e.g., an IgG antibody) or an immunologically active (i.e., specifically binding) portion of an immunoglobulin molecule, like an antibody fragment. An antibody or antibody fragment may be conjugated or otherwise derivatized within the scope of the claimed subject matter. Such antibodies include IgGl, lgG2a, IgG3, IgG4 (and IgG4 subforms), as well as IgA isotypes.

[0099] The term“antibody” herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity and comprise an Fc region or a region equivalent to the Fc region of an immunoglobulin The terms“full-length antibody”,“intact antibody”,“and“whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.

[00100] By“native antibodies” herein is meant naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3), also called a heavy chain constant region. Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain, also called a light chain constant region. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain. [00101] By“antibody fragment” herein is meant a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2, diabodies, linear antibodies, single-chain antibody molecules (e.g. scFv), single-domain antibodies, and multispecific antibodies formed from antibody fragments. For a review of certain antibody fragments, see Hudson et al., Nat Med 9, 129-134 (2003). For a review of scFv fragments, see e.g. Pliickthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer- Verlag, New York, pp. 269-315 (1994); see also WO 93/16185; and U.S. Patent Nos. 5,571,894 and 5,587,458. For discussion of Fab and F(ab')2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Patent No. 5,869,046. Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al., Nat Med 9, 129- 134 (2003); and Hollinger et al., Proc Natl Acad Sci USA 90, 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat Med 9, 129-134 (2003). Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see e.g. U.S. Patent No.6,248,516 B l). Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.

[00102] By“antigen binding domain” herein is meant the part of an antibody that comprises the area which specifically binds to and is complementary to part or all of an antigen. An antigen binding domain may be provided by, for example, one or more antibody variable domains (also called antibody variable regions). Particularly, an antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).

[00103] By“variable region” or“variable domain” herein is meant the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). See, e.g., Kindt et al., Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007). A single VH or VL domain may be sufficient to confer antigen-binding specificity.

[00104] By“hypervariable region” or“HVR” herein is meant each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops “"hypervariable loops”). Generally, native four-chain antibodies comprise six HVRs; three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the complementarity determining regions (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. With the exception of CDRl in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. Hypervariable regions (HVRs) are also referred to as“complementarity determining regions” (CDRs), and these terms are used herein interchangeably in reference to portions of the variable region that form the antigen binding regions. This particular region has been described by Kabat et al., U.S. Dept. of Health and Human Services, Sequences of Proteins of Immunological Interest (1983) and by Chothia et al., J Mol Biol 196:901-917 (1987), where the definitions include overlapping or subsets of amino acid residues when compared against each other. Nevertheless, application of either definition to refer to a CDR of an antibody or variants thereof is intended to be within the scope of the term as defined and used herein. The exact residue numbers which encompass a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can routinely determine which residues comprise a particular CDR given the variable region amino acid sequence of the antibody.

[00105] The antibody of the present invention can be chimeric antibodies, humanized antibodies, human antibodies, or antibody fusion proteins.

[00106] By“chimeric antibody” herein is meant a recombinant protein that contains the variable domains of both the heavy and light antibody chains, including the complementarity determining regions (CDRs) of an antibody derived from one species, preferably a rodent antibody, more preferably a murine antibody, while the constant domains of the antibody molecule are derived from those of a human antibody. For veterinary applications, the constant domains of the chimeric antibody may be derived from that of other species, such as a subhuman primate, cat or dog.

[00107] By“humanized antibody” herein is meant a recombinant protein in which the CDRs from an antibody from one species; e.g., a rodent antibody, are transferred from the heavy and light variable chains of the rodent antibody into human heavy and light variable domains. The constant domains of the antibody molecule are derived from those of a human antibody. In some embodiments, specific residues of the framework region of the humanized antibody, particularly those that are touching or close to the CDR sequences, may be modified, for example replaced with the corresponding residues from the original rodent, subhuman primate, or other antibody.

[00108] By“human antibody” herein is meant an antibody obtained, for example, from transgenic mice that have been“engineered” to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. The transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas. Methods for obtaining human antibodies from transgenic mice are described by Green et al, Nature Genet. 7: 13 (1994), Lonberg et al, Nature 368:856 (1994), and Taylor et al, Int. Immun. 6:579 (1994). A fully human antibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art. See for example, McCafferty et al, Nature 348:552-553 (1990) for the production of human antibodies and fragments thereof in vitro, from immunoglobulin variable domain gene repertoires from unimmunized donors. In this technique, antibody variable domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, and displayed as functional antibody fragments on the surface of the phage particle. Because the filamentous particle contains a single- stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. In this way, the phage mimics some of the properties of the B cell. Phage display can be performed in a variety of formats, for their review, see e.g. Johnson and Chiswell, Current Opinion in Structural Biology 3:5564-571 (1993). Human antibodies may also be generated by in vitro activated B cells. See U.S. Patent Nos. 5,567,610 and 5,229,275, which are incorporated herein by reference in their entirety.

[00109] By“antibody fusion protein” herein is meant a recombinantly-produced antigen- binding molecule in which two or more of the same or different natural antibody, single-chain antibody or antibody fragment segments with the same or different specificities are linked. A fusion protein comprises at least one specific binding site. Valency of the fusion protein indicates the total number of binding arms or sites the fusion protein has to antigen(s) or epitope(s); i.e., monovalent, bivalent, trivalent or mutlivalent. The multivalency of the antibody fusion protein means that it can take advantage of multiple interactions in binding to an antigen, thus increasing the avidity of binding to the antigen, or to different antigens. Specificity indicates how many different types of antigen or epitope an antibody fusion protein is able to bind; i.e., monospecific, bispecific, trispecific, multispecific. Using these definitions, a natural antibody, e.g., an IgG, is bivalent because it has two binding arms but is monospecific because it binds to one type of antigen or epitope. A monospecific, multivalent fusion protein has more than one binding site for the same antigen or epitope. For example, a monospecific diabody is a fusion protein with two binding sites reactive with the same antigen. The fusion protein may comprise a multivalent or multispecific combination of different antibody components or multiple copies of the same antibody component. The fusion protein may additionally comprise a therapeutic agent.

[00110] In some embodiments, the targeting moiety comprises a probody, such as those disclosed in US Patent Nos: 8,518,404; 8,513,390; and US Pat. Appl. Pub. Nos: 20120237977A1, 20120149061A1, and 20130150558A1, the disclosures of which are incorporated by reference in their entireties. [00111] Probodies are monoclonal antibodies that are selectively activated within the cancer microenvironment, focusing the activity of therapeutic antibodies to tumors and sparing healthy tissue.

[00112] In general, the porbody comprises at least an antibody or antibody fragment thereof (collectively referred to as“AB”), capable of specifically binding a target, wherein the AB is modified by a masking moiety (MM). When the AB is modified with a MM and is in the presence of the target, specific binding of the AB to its target is reduced or inhibited, as compared to the specific binding of the AB not modified with an MM or the specific binding of the parental AB to the target. The dissociation constant (Kd) of the MM towards the AB is generally greater than the Kd of the AB towards the target. When the AB is modified with a MM and is in the presence of the target, specific binding of the AB to its target can be reduced or inhibited, as compared to the specific binding of the AB not modified with an MM or the specific binding of the parental AB to the target. When an AB is coupled to or modified by a MM, the MM can‘mask’ or reduce, or inhibit the specific binding of the AB to its target. When an AB is coupled to or modified by a MM, such coupling or modification can effect a structural change which reduces or inhibits the ability of the AB to specifically bind its target.

[00113] The present invention relates to therapeutic combinations comprising WNT inhibitors and methods for treating cancers using combination therapy.

[00114] In some embodiments, the probody is an activatable antibodies (AAs) where the AB modified by an MM can further include one or more cleavable moieties (CM). Such AAs exhibit activatable/switchable binding, to the AB's target. AAs generally include an antibody or antibody fragment (AB), modified by or coupled to a masking moiety (MM) and a modifiable or cleavable moiety (CM). In some embodiments, the CM contains an amino acid sequence that serves as a substrate for a protease of interest. In other embodiments, the CM provides a cysteine-cysteine disulfide bond that is cleavable by reduction. In yet other embodiments the CM provides a photolytic substrate that is activatable by photolysis.

[00115] The CM and AB of the AA may be selected so that the AB represents a binding moiety for a target of interest, and the CM represents a substrate for a protease that is co-localized with the target at a treatment site in a subject. Alternatively, or in addition, the CM is a cysteine-cysteine disulfide bond that is cleavable as a result of reduction of this disulfide bond. AAs contain at least one of a protease-cleavable CM or a cysteine-cysteine disulfide bond, and in some embodiments include both kinds of CMs. The AAs can alternatively or further include a photolabile substrate, activatable by a light source. The AAs disclosed herein find particular use where, for example, a protease capable of cleaving a site in the CM is present at relatively higher levels in target-containing tissue of a treatment site (for example diseased tissue; for example, for therapeutic treatment or diagnostic treatment) than in tissue of non-treatment sites (for example in healthy tissue). The AAs disclosed herein also find particular use where, for example, a reducing agent capable of reducing a site in the CM is present at relatively higher levels in target-containing tissue of a treatment or diagnostic site than in tissue of non-treatment non-diagnostic sites. The AAs disclosed herein also find particular use where, for example, a light source, for example, by way of laser, capable of photolysing a site in the CM is introduced to a target-containing tissue of a treatment or diagnostic site.

[00116] In some embodiments, AAs can provide for reduced toxicity and/or adverse side effects that could otherwise result from binding of the AB at non-treatment sites if the AB were not masked or otherwise inhibited from binding its target. Where the AA contains a CM that is cleavable by a reducing agent that facilitates reduction of a disulfide bond, the ABs of such AAs may selected to exploit activation of an AB where a target of interest is present at a desired treatment site characterized by elevated levels of a reducing agent, such that the environment is of a higher reduction potential than, for example, an environment of a non-treatment site.

[00117] In general, an AA can be designed by selecting an AB of interest and constructing the remainder of the AA so that, when conformationally constrained, the MM provides for masking of the AB or reduction of binding of the AB to its target. Structural design criteria to be taken into account to provide for this functional feature.

Anti-PD-1 Antibodies

[00118] In some embodiments, the TM is a monoclonal anti-PD-1 antibody.

[00119] Programmed Death-1 (“PD-1”) is a receptor of PD-L1 (also known as CD274, B7-H1, or B7-DC). PD-1 is an approximately 31 kD type I membrane protein member of the extended CD28/CTLA4 family of T cell regulators (Ishida, Y. et al. (1992) EMBO J. 11:3887-3895; US Pat. Appl. Pub. No. 2007/0202100; 2008/0311117; 2009/00110667; U.S. Pat. Nos. 6,808,710; 7,101,550; 7,488,802; 7,635,757; 7,722,868; PCT Publication No. WO 01/14557). In comparisoin to CTLA4, PD-1 more broadly negatively regulates immune responses.

[00120] PD-1 is expressed on activated T cells, B cells, and monocytes (Agata, Y. et al. (1996) Int. Immunol. 8(5):765-772; Yamazaki, T. et al. (2002 J. Immunol. 169:5538-5545) and at low levels in natural killer (NK) T cells (Nishimura, H. et al. (2000) J. Exp. Med.191:891-898; Martin-Orozco, N. et al. (2007), Semin. Cancer Biol.17(4):288-298).

[00121] The extracellular region of PD-1 consists of a single immunoglobulin (Ig) V domain with 23% identity to the equivalent domain in CTLA4 (Martin-Orozco, N. et al. (2007) Semin. Cancer Biol. 17(4):288-298). The extracellular IgV domain is followed by a transmembrane region and an intracellular tail. The intracellular tail contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif, which suggests that PD-1 negatively regulates TCR signals (Ishida, Y. et al. (1992 EMBO J.11:3887- 3895; Blank, C. et al. (Epub 2006 Dec.29) Immunol. Immunother.56(5):739-745). [00122] Antibodies capable of immunospecifically binding to murine PD-1 have been reported (see, e.g., Agata, T. et al. (1996) Int. Immunol.8(5):765-772).

[00123] Anti-PD-1 antibodies bind to PD-1 and enhance T-cell function to upregulate cell- mediated immune responses and for the treatment of T cell dysfunctional disorders, such as tumor immunity.

[00124] In some embodiments, the anti-PD-1 antibody is MK-3475 (formerly lambrolizumab, Merck), AMP-514, AMP-224 (MedImmune/AstraZeneca), BMS-936558 (MDX-1106, Bristol-Myers Squibb), or CT-011 (Curetech).

[00125] Pembrolizumab (MK-3475) is a humanized, monoclonal anti-PD-1 antibody designed to reactivate anti-tumor immunity. Pembrolizumab exerts dual ligand blockade of the PD-1 pathway by inhibiting the interaction of PD-1 on T cells with its ligands PD-L1 and PD-L2.

[00126] In some embodiments, the anti-PD-1 antibody is one of the antibodies disclosed in US 8,354,509, and US 8,168,757, the disclosure of which is incorporated by reference in their entirety.

[00127] Nivolumab (also known as BMS-936558 or MDX1106, is a fully human IgG4 monoclonal antibody developed by Bristol-Myers Squibb for the treatment of cancer.

[00128] In some embodiments, the anti-PD-1 antibody is one of the antibodies disclosed in WO2004/056875, US 7,488,802 and US 8,008,449, the disclosure of which is incorporated by reference in their entirety.

[00129] AMP-514 and AMP-224 are an anti-programmed cell death 1 (PD-1) monoclonal antibody (mAb) developed by Amplimmune, which was acquired by MedImmune.

[00130] In some embodiments, the anti-PD-1 antibody is one of the antibodies disclosed in US Appl. Pub. No.20140044738, the disclosure of which is incorporated by reference in their entirety.

[00131] In some embodiments, the six CDRs are: (A) the three light chain and the three heavy chain CDRs of anti-PD-1 antibody 1E3; (B) the three light chain and the three heavy chain CDRs of anti-PD-1 antibody 1E8; or (C) the three light chain and the three heavy chain CDRs of anti-PD-1 antibody 1H3.

[00132] Pidilizumab (CT-011) is an anti-PD-1 monoclonal antibody developed by Israel-based Curetech Ltd.

[00133] In some embodiments, the anti-PD-1 antibody is one of the antibodies disclosed in US Pat. Appl. Pub. Nos. 20080025980 and 20130022595, the disclosure of which is incorporated by reference in their entirety.

Anti-PD-L1 Antibodies [00134] In some embodiments, the TM is a monoclonal anti-PD-L1 antibody.

[00135] Programmed cell death 1 ligand 1 (PD-L1, also known as CD274 and B7-H1) is a ligand for PD-1, found on activated T cells, B cells, myeloid cells and macrophages. Although there are two endogenous ligands for PD-1, PD-L1 and PD-L2, anti-tumor therapies have focused on anti-PD- L1 antibodies. The complex of PD-1 and PD-L1 inhibits proliferation of CD8+ T cells and reduces the immune response (Topalian et al., 2012, N Engl J Med 366:2443-54; Brahmer et al., 2012, N Eng J Med 366:2455-65). Anti-PD-L1 antibodies have been used for treatment of non-small cell lung cancer, melanoma, colorectal cancer, renal-cell cancer, pancreatic cancer, gastric cancer, ovarian cancer, breast cancer, and hematologic malignancies (Brahmer et al., N Eng J Med 366:2455-65; Ott et al., 2013, Clin Cancer Res 19:5300-9; Radvanyi et al., 2013, Clin Cancer Res 19:5541; Menzies & Long, 2013, Ther Adv Med Oncol 5:278-85; Berger et al., 2008, Clin Cancer Res 14:13044-51). PD- L1 is a B7 family member that is expressed on many cell types, including APCs and activated T cells (Yamazaki et al. (2002) J. Immunol.169:5538). PD-L1 binds to both PD-1 and B7-1. Both binding of T-cell-expressed B7-1 by PD-L1 and binding of T-cell-expressed PD-L1 by B7-1 result in T cell inhibition (Butte et al. (2007) Immunity 27:111). There is also evidence that, like other B7 family members, PD-L1 can also provide costimulatory signals to T cells (Subudhi et al. (2004) J. Clin. Invest.113:694; Tamura et al. (2001) Blood 97:1809).

[00136] By“PD-L1” herein is meant to include any variants or isoforms which are naturally expressed by cells, and/or fragments thereof having at least one biological activity of the full-length polypeptide, unless otherwise expressly defined. In addition, the term“PD-L1” includes PD- L1 (Freeman et al. (2000) J. Exp. Med.192:1027) and any variants or isoforms which are naturally expressed by cells, and/or fragments thereof having at least one biological activity of the full-length polypeptides. For example, PD-L1 sequences from different species, including humans, are well known in the art (see, for example, herein incorporated in their entirety by reference, Chen et al., U.S. Pat. No. 6,803,192, which discloses human and mouse PD-L1 sequences; Wood et al., U.S. Pat. No. 7,105,328, which discloses human PD-L1 sequences.

[00137] Anti-PD-L1 antibodies bind to PD-L1 and enhance T-cell function to upregulate cell- mediated immune responses and for the treatment of T cell dysfunctional disorders, such as tumor immunity.

[00138] In some embodiments, the anti-PD-L1 antibody is MPDL3280A and YW243.55.S70, (Genentech/Roche), MEDI-4736 (MedImmune/AstraZeneca), BMS-936559 (MDX-1105, Bristol- Myers Squibb), and MSB0010718C (EMD Serono/Merck KGaA).

[00139] MPDL3280A (TECENTRIQ™, or atezolizumab, Genentech) is an engineered anti-PD-L1 antibody designed to target PD-L1 expressed on tumor cells and tumor-infiltrating immune cells. MPDL3280A is designed to prevent PD-L1 from binding to PD-1 and B7.1. This blockade of PD-L1 may enable the activation of T cells, restoring their ability to detect and attack tumor cells. MPDL3280A contains an engineered fragment crystallizable (Fc) domain designed to optimize efficacy and safety by minimizing antibody-dependent cellular cytotoxicity (ADCC).

[00140] In some embodiments, the anti-PD-L1 antibody is one of the antibodies disclosed in US 7,943,743, the disclosure of which is incorporated by reference in their entirety.

[00141] BMS-936559 (MDX-1105, Bristol-Myers Squibb) is a fully human IgG4 anti-PD-L1 mAb that inhibits the binding of the PD-L1 ligand to both PD-1 and CD80.

[00142] In some embodiments, the anti-PD-L1 antibody is one of the antibodies disclosed in US 7,943,743, the disclosure of which is incorporated by reference in their entirety.

[00143] MSB0010718C (EMD Serono of Merck KGaA) is fully human IgG1 monoclonal antibody that binds to PD-L1.

[00144] In some embodiments, the anti-PD-L1 antibody is one of the antibodies disclosed in WO 2013079174 A1, the disclosure of which is incorporated by reference in their entirety.

[00145] MEDI4736 (MedImmune/AstraZeneca) is a human IgG1 antibody which binds specifically to PD-L1, preventing binding to PD-1 and CD80.

[00146] In some embodiments, the anti-PD-L1 antibody is one of the antibodies disclosed in WO 2011066389 A1 and US 8,779,108, the disclosure of which is incorporated by reference in their entirety.

[00147] In some embodiments, the anti-PD-L1 antibody is one of the antibodies disclosed in US 8,552,154, the disclosure of which is incorporated by reference in their entirety

[00148] In some embodiments, the targeting moiety comprises a Fab, Fab’, F(ab’)2, single domain antibody, T and Abs dimer, Fv, scFv, dsFv, ds-scFv, Fd, linear antibody, minibody, diabody, bispecific antibody fragment, bibody, tribody, sc-diabody, kappa (lamda) body, BiTE, DVD-Ig, SIP, SMIP, DART, or an antibody analogue comprising one or more CDRs.

PD-L/PD-1 Axis antagonist Comprising a Targeting Moiety

[00149] In some aspects, the PD-L/PD-1 Axis antagonist is a targeted therapeutic comprise a targeting moiety, such as an ADC.

[00150] By“targeting moiety (TM)” or“targeting agent” here in is meant a molecule, complex, or aggregate, that binds specifically or selectively to a target molecule, cell, particle, tissue or aggregate, which generally is referred to as a“target” or a“marker,” and these are discussed in further detail herein.

[00151] In some embodiments, the targeting moiety comprises an immunoglobulin, a protein, a peptide, a small molecule, a nanoparticle, or a nucleic acid. [00152] Exemplary targeting agents such as antibodies (e.g., chimeric, humanized and human), ligands for receptors, lecitins, and saccharides, and substrate for certain enzymes are recognized in the art and are useful without limitation in practicing the present invention. Other targeting agents include a class of compounds that do not include specific molecular recognition motifs include nanoparticles, macromolecules such as poly(ethylene glycol), polysaccharide, and polyamino acids which add molecular mass to the activating moiety. The additional molecular mass affects the pharmacokinetics of the activating moiety, e.g., serum half-life.

[00153] In some embodiments, a targeting moiety is an antibody, antibody fragment, bispecific antibody or other antibody-based molecule or compound. However, other examples of targeting moieties are known in the art and may be used, such as aptamers, avimers, receptor-binding ligands, nucleic acids, biotin-avidin binding pairs, binding peptides or proteins, etc. The terms“targeting moiety” and“binding moiety” are used synonymously herein.

[00154] By“target” or“marker” herein is meant any entity that is capable of specifically binding to a particular targeting moiety. In some embodiments, targets are specifically associated with one or more particular cell or tissue types. In some embodiments, targets are specifically associated with one or more particular disease states. In some embodiments, targets are specifically associated with one or more particular developmental stages. For example, a cell type specific marker is typically expressed at levels at least 2 fold greater in that cell type than in a reference population of cells. In some embodiments, the cell type specific marker is present at levels at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 50 fold, at least 100 fold, or at least 1,000 fold greater than its average expression in a reference population. Detection or measurement of a cell type specific marker may make it possible to distinguish the cell type or types of interest from cells of many, most, or all other types. In some embodiments, a target can comprise a protein, a carbohydrate, a lipid, and/or a nucleic acid, as described herein.

[00155] A substance is considered to be“targeted” for the purposes described herein if it specifically binds to a nucleic acid targeting moiety. In some embodiments, a nucleic acid targeting moiety specifically binds to a target under stringent conditions. An inventive complex or compound comprising targeting moiety is considered to be“targeted” if the targeting moiety specifically binds to a target, thereby delivering the entire complex or compound composition to a specific organ, tissue, cell, extracellular matrix component, and/or intracellular compartment.

[00156] In certain embodiments, compound in accordance with the present invention comprise a targeting moiety which specifically binds to one or more targets (e.g. antigens) associated with an organ, tissue, cell, extracellular matrix component, and/or intracellular compartment. In some embodiments, compounds comprise a targeting moiety which specifically binds to targets associated with a particular organ or organ system. In some embodiments, compounds in accordance with the present invention comprise a nuclei targeting moiety which specifically binds to one or more intracellular targets (e.g. organelle, intracellular protein). In some embodiments, compounds comprise a targeting moiety which specifically binds to targets associated with diseased organs, tissues, cells, extracellular matrix components, and/or intracellular compartments. In some embodiments, compounds comprise a targeting moiety which specifically binds to targets associated with particular cell types (e.g. endothelial cells, cancer cells, malignant cells, prostate cancer cells, etc.).

[00157] In some embodiments, compounds in accordance with the present invention comprise a targeting moiety which binds to a target that is specific for one or more particular tissue types (e.g. liver tissue vs. prostate tissue). In some embodiments, compounds in accordance with the present invention comprise a targeting moiety which binds to a target that is specific for one or more particular cell types (e.g. T cells vs. B cells). In some embodiments, compounds in accordance with the present invention comprise a targeting moiety which binds to a target that is specific for one or more particular disease states (e.g. tumor cells vs. healthy cells). In some embodiments, compounds in accordance with the present invention comprise a targeting moiety which binds to a target that is specific for one or more particular developmental stages (e.g. stem cells vs. differentiated cells).

[00158] In some embodiments, a target may be a marker that is exclusively or primarily associated with one or a few cell types, with one or a few diseases, and/or with one or a few developmental stages. A cell type specific marker is typically expressed at levels at least 2 fold greater in that cell type than in a reference population of cells which may consist, for example, of a mixture containing cells from a plurality (e.g., 5-10 or more) of different tissues or organs in approximately equal amounts. In some embodiments, the cell type specific marker is present at levels at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 50 fold, at least 100 fold, or at least 1000 fold greater than its average expression in a reference population. Detection or measurement of a cell type specific marker may make it possible to distinguish the cell type or types of interest from cells of many, most, or all other types.

[00159] In some embodiments, a target comprises a protein, a carbohydrate, a lipid, and/or a nucleic acid. In some embodiments, a target comprises a protein and/or characteristic portion thereof, such as a tumor-marker, integrin, cell surface receptor, transmembrane protein, intercellular protein, ion channel, membrane transporter protein, enzyme, antibody, chimeric protein, glycoprotein, etc. In some embodiments, a target comprises a carbohydrate and/or characteristic portion thereof, such as a glycoprotein, sugar (e.g., monosaccharide, disaccharide, polysaccharide), glycocalyx (i.e., the carbohydrate-rich peripheral zone on the outside surface of most eukaryotic cells) etc. In some embodiments, a target comprises a lipid and/or characteristic portion thereof, such as an oil, fatty acid, glyceride, hormone, steroid (e.g., cholesterol, bile acid), vitamin (e.g. vitamin E), phospholipid, sphingolipid, lipoprotein, etc. In some embodiments, a target comprises a nucleic acid and/or characteristic portion thereof, such as a DNA nucleic acid; RNA nucleic acid; modified DNA nucleic acid; modified RNA nucleic acid; nucleic acid that includes any combination of DNA, RNA, modified DNA, and modified RNA.

[00160] Numerous markers are known in the art. Typical markers include cell surface proteins, e.g., receptors. Exemplary receptors include, but are not limited to, the transferrin receptor; LDL receptor; growth factor receptors such as epidermal growth factor receptor family members (e.g., EGFR, Her2, Her3, Her4) or vascular endothelial growth factor receptors, cytokine receptors, cell adhesion molecules, integrins, selectins, and CD molecules. The marker can be a molecule that is present exclusively or in higher amounts on a malignant cell, e.g., a tumor antigen.

[00161] In some embodiments, the targeting moiety binds to a tumor cell specifically or preferably in comparison to a non-tumor cell.

[00162] The binding of target moiety to tumor cell can be measured using assays known in the art.

[00163] In some embodiments, the tumor cell is of a carcinoma, a sarcoma, a lymphoma, a myeloma, or a central nervous system cancer.

[00164] In some embodiments, the targeting moiety is capable of binding to a tumor antigen specifically or preferably in comparison to a non-tumor antigen.

[00165] In certain specific embodiments, a target is a tumor marker. In some embodiments, a tumor marker is an antigen that is present in a tumor that is not present in normal organs, tissues, and/or cells. In some embodiments, a tumor marker is an antigen that is more prevalent in a tumor than in normal organs, tissues, and/or cells. In some embodiments, a tumor marker is an antigen that is more prevalent in malignant cancer cells than in normal cells.

[00166] In some embodiments, the targeting moiety comprises folic acid or a derivative thereof.

[00167] In recent years, research on folic acid had made great progress. Folic acid is a small molecule vitamin that is necessary for cell division. Tumor cells divide abnormally and there is a high expression of folate receptor (FR) on tumor cell surface to capture enough folic acid to support cell division.

[00168] Data indicate FR expression in tumor cells is 20-200 times higher than normal cells. The expression rate of FR in various malignant tumors are: 82% in ovarian cancer, 66% in non-small cell lung cancer, 64% in kidney cancer, 34% in colon cancer, and 29% in breast cancer (Xia W, Low PS. Late-targeted therapies for cancer. J Med Chem. 2010; 14; 53 (19):6811-24). The expression rate of FA and the degree of malignancy of epithelial tumor invasion and metastasis is positively correlated. FA enters cell through FR mediated endocytosis, and FA through its carboxyl group forms FA complexes with drugs which enter the cells. Under acidic conditions (pH value of 5), FR separates from the FA, and FA releases drugs into the cytoplasm. [00169] Clinically, the system can be used to deliver drugs selectively attack the tumor cells. Folic acid has small molecular weight, has non-immunogenicity and high stability, and is inexpensive to synthesis. More importantly, chemical coupling between the drug and the carrier is simple, and as such using FA as targeting molecule to construct drug delivery system has become a research hotspot for cancer treatment. Currently EC145 (FA chemotherapy drug conjugate compound) that is in clinical trials can effectively attack cancer cells (Pribble P and Edelman MJ. EC145: a novel targeted agent for adenocarcinoma of the lung. Expert Opin. Investig. Drugs (2012) 21:755-761).

[00170] In some embodiments, the targeting moiety comprises extracellular domains (ECD) or soluble form of PD-1, PDL-1, CTLA4, CD47, BTLA, KIR, TIM3, 4-1BB, and LAG3, full length of partial of a surface ligand Amphiregulin, Betacellulin, EGF, Ephrin, Epigen, Epiregulin, IGF, Neuregulin, TGF, TRAIL, or VEGF.

[00171] In some embodiments, the targeting moiety comprises a Fab, Fab’, F(ab’)2, single domain antibody, T and Abs dimer, Fv, scFv, dsFv, ds-scFv, Fd, linear antibody, minibody, diabody, bispecific antibody fragment, bibody, tribody, sc-diabody, kappa (lamda) body, BiTE, DVD-Ig, SIP, SMIP, DART, or an antibody analogue comprising one or more CDRs.

[00172] n some embodiments, the targeting moiety is an antibody, or antibody fragment, that is selected based on its specificity for an antigen expressed on a target cell, or at a target site, of interest. A wide variety of tumor-specific or other disease-specific antigens have been identified and antibodies to those antigens have been used or proposed for use in the treatment of such tumors or other diseases. The antibodies that are known in the art can be used in the compounds of the invention, in particular for the treatment of the disease with which the target antigen is associated. Examples of target antigens (and their associated diseases) to which an antibody-linker-drug conjugate of the invention can be targeted include: CD2, CD19, CD20, CD22, CD27, CD33, CD37, CD38, CD40, CD44, CD47, CD52, CD56, CD70, CD79, CD137, 4-1BB, 5T4,AGS-5, AGS-16, Angiopoietin 2, B7.1, B7.2, B7DC, B7H1, B7H2, B7H3, BT-062, BTLA, CAIX, carcinoembryonic antigen, CTLA4, Cripto, ED-B, ErbB1, ErbB2, ErbB3, ErbB4, EGFL7, EpCAM, EphA2, EphA3, EphB2, FAP, fibronectin, folate receptor, ganglioside GM3, GD2, glucocorticoid-induced tumor necrosis factor receptor (GITR), gp100, gpA33, GPNMB, ICOS, IGF1R, integrin αν, integrin ανβ, KIR, LAG-3, lewis Y, mesothelin, c-MET, MN carbonic anhydrase IX, MUC1, MUC16, Nectin-4, NKGD2, NOTCH, OX40, OX40L, PD-1, PDL1, PSCA, PSMA, RANKL, ROR1, ROR2, SLC44A4, syndecan-1, TACI, TAG-72, tenascin, TIM3, TRAILR1, TRAILR2,VEGFR-1, VEGFR-2, VEGFR-3.

[00173] In some embodiments, the targeting moiety comprises a particle (target particle), preferably a nanoparticle, optionally a targeted nanoparticle that attached to a targeting molecule that can binds specifically or preferably to a target. In some embodiments, the targeting particle by itself guides the compound of the present invention (such as by enrichment in tumor cells or tissue) and there is no additional targeting molecules attached therein.

[00174] By“nanoparticle” herein is meant any particle having a diameter of less than 1000 nm. In some embodiments, a therapeutic agent and/or targeting molecule can be associated with the polymeric matrix. In some embodiments, the targeting molecule can be covalently associated with the surface of a polymeric matrix. In some embodiments, covalent association is mediated by a linker. In some embodiments, the therapeutic agent can be associated with the surface of, encapsulated within, surrounded by, and/or dispersed throughout the polymeric matrix. US Pat. No. 8,246,968, which is incorporated in its entirety.

[00175] In general, nanoparticles of the present invention comprise any type of particle. Any particle can be used in accordance with the present invention. In some embodiments, particles are biodegradable and biocompatible. In general, a biocompatible substance is not toxic to cells. In some embodiments, a substance is considered to be biocompatible if its addition to cells results in less than a certain threshold of cell death. In some embodiments, a substance is considered to be biocompatible if its addition to cells does not induce adverse effects. In general, a biodegradable substance is one that undergoes breakdown under physiological conditions over the course of a therapeutically relevant time period (e.g., weeks, months, or years). In some embodiments, a biodegradable substance is a substance that can be broken down by cellular machinery. In some embodiments, a biodegradable substance is a substance that can be broken down by chemical processes. In some embodiments, a particle is a substance that is both biocompatible and biodegradable. In some embodiments, a particle is a substance that is biocompatible, but not biodegradable. In some embodiments, a particle is a substance that is biodegradable, but not biocompatible.

[00176] In some embodiments, particles are greater in size than the renal excretion limit (e.g. particles having diameters of greater than 6 nm). In some embodiments, particles are small enough to avoid clearance of particles from the bloodstream by the liver (e.g. particles having diameters of less than 1000 nm). In general, physiochemical features of particles should allow a targeted particle to circulate longer in plasma by decreasing renal excretion and liver clearance.

[00177] It is often desirable to use a population of particles that is relatively uniform in terms of size, shape, and/or composition so that each particle has similar properties. For example, at least 80%, at least 90%, or at least 95% of the particles may have a diameter or greatest dimension that falls within 5%, 10%, or 20% of the average diameter or greatest dimension. In some embodiments, a population of particles may be heterogeneous with respect to size, shape, and/or composition.

[00178] Zeta potential is a measurement of surface potential of a particle. In some embodiments, particles have a zeta potential ranging between−50 mV and +50 mV. In some embodiments, particles have a zeta potential ranging between−25 mV and +25 mV. In some embodiments, particles have a zeta potential ranging between−10 mV and +10 mV. In some embodiments, particles have a zeta potential ranging between−5 mV and +5 mV. In some embodiments, particles have a zeta potential ranging between 0 mV and +50 mV. In some embodiments, particles have a zeta potential ranging between 0 mV and +25 mV. In some embodiments, particles have a zeta potential ranging between 0 mV and +10 mV. In some embodiments, particles have a zeta potential ranging between 0 mV and +5 mV. In some embodiments, particles have a zeta potential ranging between−50 mV and 0 mV. In some embodiments, particles have a zeta potential ranging between−25 mV and 0 mV. In some embodiments, particles have a zeta potential ranging between−10 mV and 0 mV. In some embodiments, particles have a zeta potential ranging between−5 mV and 0 mV. In some embodiments, particles have a substantially neutral zeta potential (i.e. approximately 0 mV).

[00179] A variety of different particles can be used in accordance with the present invention. In some embodiments, particles are spheres or spheroids. In some embodiments, particles are spheres or spheroids. In some embodiments, particles are flat or plate-shaped. In some embodiments, particles are cubes or cuboids. In some embodiments, particles are ovals or ellipses. In some embodiments, particles are cylinders, cones, or pyramids.

[00180] In some embodiments, particles are microparticles (e.g. microspheres). In general, a “microparticle” refers to any particle having a diameter of less than 1000 μm. In some embodiments, particles are picoparticles (e.g. picospheres). In general, a“picoparticle” refers to any particle having a diameter of less than 1 nm. In some embodiments, particles are liposomes. In some embodiments, particles are micelles.

[00181] Particles can be solid or hollow and can comprise one or more layers (e.g., nanoshells, nanorings). In some embodiments, each layer has a unique composition and unique properties relative to the other layer(s). For example, particles may have a core/shell structure, wherein the core is one layer and the shell is a second layer. Particles may comprise a plurality of different layers. In some embodiments, one layer may be substantially cross-linked, a second layer is not substantially cross-linked, and so forth. In some embodiments, one, a few, or all of the different layers may comprise one or more therapeutic or diagnostic agents to be delivered. In some embodiments, one layer comprises an agent to be delivered, a second layer does not comprise an agent to be delivered, and so forth. In some embodiments, each individual layer comprises a different agent or set of agents to be delivered.

[00182] In some embodiments, a particle is porous, by which is meant that the particle contains holes or channels, which are typically small compared with the size of a particle. For example, a particle may be a porous silica particle, e.g., a mesoporous silica nanoparticle or may have a coating of mesoporous silica (Lin et al., 2005, J. Am. Chem. Soc., 17:4570). Particles may have pores ranging from about 1 nm to about 50 nm in diameter, e.g., between about 1 and 20 nm in diameter. Between about 10% and 95% of the volume of a particle may consist of voids within the pores or channels.

[00183] Particles may have a coating layer. Use of a biocompatible coating layer can be advantageous, e.g., if the particles contain materials that are toxic to cells. Suitable coating materials include, but are not limited to, natural proteins such as bovine serum albumin (BSA), biocompatible hydrophilic polymers such as polyethylene glycol (PEG) or a PEG derivative, phospholipid-(PEG), silica, lipids, polymers, carbohydrates such as dextran, other nanoparticles that can be associated with inventive nanoparticles etc. Coatings may be applied or assembled in a variety of ways such as by dipping, using a layer-by-layer technique, by self-assembly, conjugation, etc. Self-assembly refers to a process of spontaneous assembly of a higher order structure that relies on the natural attraction of the components of the higher order structure (e.g., molecules) for each other. It typically occurs through random movements of the molecules and formation of bonds based on size, shape, composition, or chemical properties.

[00184] Examples of polymers include polyalkylenes (e.g. polyethylenes), polycarbonates (e.g. poly(1,3-dioxan-2one)), polyanhydrides (e.g. poly(sebacic anhydride)), polyhydroxyacids (e.g. poly(β-hydroxyalkanoate)), polyfumarates, polycaprolactones, polyamides (e.g. polycaprolactam), polyacetals, polyethers, polyesters (e.g. polylactide, polyglycolide), poly(orthoesters), polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, and polyamines. In some embodiments, polymers in accordance with the present invention include polymers which have been approved for use in humans by the U.S. Food and Drug Administration (FDA) under 21 C.F.R. §177.2600, including but not limited to polyesters (e.g. polylactic acid, polyglycolic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(1,3-dioxan-2one)); polyanhydrides (e.g. poly(sebacic anhydride)); polyethers (e.g., polyethylene glycol); polyurethanes; polymethacrylates; polyacrylates; and polycyanoacrylates.

[00185] In some embodiments, particles can be non-polymeric particles (e.g. metal particles, quantum dots, ceramic particles, polymers comprising inorganic materials, bone-derived materials, bone substitutes, viral particles, etc.). In some embodiments, a therapeutic or diagnostic agent to be delivered can be associated with the surface of such a non-polymeric particle. In some embodiments, a non-polymeric particle is an aggregate of non-polymeric components, such as an aggregate of metal atoms (e.g. gold atoms). In some embodiments, a therapeutic or diagnostic agent to be delivered can be associated with the surface of and/or encapsulated within, surrounded by, and/or dispersed throughout an aggregate of non-polymeric components.

[00186] Particles (e.g. nanoparticles, microparticles) may be prepared using any method known in the art. For example, particulate formulations can be formed by methods as nanoprecipitation, flow focusing fluidic channels, spray drying, single and double emulsion solvent evaporation, solvent extraction, phase separation, milling, microemulsion procedures, microfabrication, nanofabrication, sacrificial layers, simple and complex coacervation, and other methods well known to those of ordinary skill in the art. Alternatively or additionally, aqueous and organic solvent syntheses for monodisperse semiconductor, conductive, magnetic, organic, and other nanoparticles have been described (Pellegrino et al., 2005, Small, 1:48; Murray et al., 2000, Ann. Rev. Mat. Sci., 30:545; and Trindade et al., 2001, Chem. Mat., 13:3843).

[00187] Methods for making microparticles for delivery of encapsulated agents are described in the literature (see, e.g., Doubrow, Ed.,“Microcapsules and Nanoparticles in Medicine and Pharmacy,” CRC Press, Boca Raton, 1992; Mathiowitz et al., 1987, J. Control. Release, 5:13; Mathiowitz et al., 1987, Reactive Polymers, δ: 275; and Mathiowitz et al., 1988, J. Appl. Polymer Sci., 35:755).

[00188] In some embodiments, the targeting moiety comprises an nucleic acid targeting moiety.

[00189] In general, a nucleic acid targeting moiety is any polynucleotide that binds to a component associated with an organ, tissue, cell, extracellular matrix component, and/or intracellular compartment (the target).

[00190] In some embodiments, the nucleic acid targeting moieties are aptamers.

[00191] An aptamer is typically a polynucleotide that binds to a specific target structure that is associated with a particular organ, tissue, cell, extracellular matrix component, and/or intracellular compartment. In general, the targeting function of the aptamer is based on the three-dimensional structure of the aptamer. In some embodiments, binding of an aptamer to a target is typically mediated by the interaction between the two- and/or three-dimensional structures of both the aptamer and the target. In some embodiments, binding of an aptamer to a target is not solely based on the primary sequence of the aptamer, but depends on the three-dimensional structure(s) of the aptamer and/or target. In some embodiments, aptamers bind to their targets via complementary Watson-Crick base pairing which is interrupted by structures (e.g. hairpin loops) that disrupt base pairing.

[00192] In some embodiments, the nucleic acid targeting moieties are spiegelmers (PCT Publications WO 98/08856, WO 02/100442, and WO 06/117217). In general, spiegelmers are synthetic, mirror-image nucleic acids that can specifically bind to a target (i.e. mirror image aptamers). Spiegelmers are characterized by structural features which make them not susceptible to exo- and endo-nucleases.

[00193] One of ordinary skill in the art will recognize that any nucleic acid targeting moiety (e.g. aptamer or spiegelmer) that is capable of specifically binding to a target can be used in accordance with the present invention. In some embodiments, nucleic acid targeting moieties to be used in accordance with the present invention may target a marker associated with a disease, disorder, and/or condition. In some embodiments, nucleic acid targeting moieties to be used in accordance with the present invention may target cancer-associated targets. In some embodiments, nucleic acid targeting moieties to be used in accordance with the present invention may target tumor markers. Any type of cancer and/or any tumor marker may be targeted using nucleic acid targeting moieties in accordance with the present invention. To give but a few examples, nucleic acid targeting moieties may target markers associated with prostate cancer, lung cancer, breast cancer, colorectal cancer, bladder cancer, pancreatic cancer, endometrial cancer, ovarian cancer, bone cancer, esophageal cancer, liver cancer, stomach cancer, brain tumors, cutaneous melanoma, and/or leukemia.

[00194] Nucleic acids of the present invention (including nucleic acid nucleic acid targeting moieties and/or functional RNAs to be delivered, e.g., RNAi-inducing entities, ribozymes, tRNAs, etc., described in further detail below) may be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, enzymatic or chemical cleavage of a longer precursor, etc. Methods of synthesizing RNAs are known in the art (see, e.g., Gait, M. J. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, D.C.: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in molecular biology, v.288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005).

[00195] The nucleic acid that forms the nucleic acid nucleic acid targeting moiety may comprise naturally occurring nucleosides, modified nucleosides, naturally occurring nucleosides with hydrocarbon linkers (e.g., an alkylene) or a polyether linker (e.g., a PEG linker) inserted between one or more nucleosides, modified nucleosides with hydrocarbon or PEG linkers inserted between one or more nucleosides, or a combination of thereof. In some embodiments, nucleotides or modified nucleotides of the nucleic acid nucleic acid targeting moiety can be replaced with a hydrocarbon linker or a polyether linker provided that the binding affinity and selectivity of the nucleic acid nucleic acid targeting moiety is not substantially reduced by the substitution (e.g., the dissociation constant of the nucleic acid nucleic acid targeting moiety for the target should not be greater than about 1×10 -3 M).

[00196] It will be appreciated by those of ordinary skill in the art that nucleic acids in accordance with the present invention may comprise nucleotides entirely of the types found in naturally occurring nucleic acids, or may instead include one or more nucleotide analogs or have a structure that otherwise differs from that of a naturally occurring nucleic acid. U.S. Pat. Nos. 6,403,779; 6,399,754; 6,225,460; 6,127,533; 6,031,086; 6,005,087; 5,977,089; and references therein disclose a wide variety of specific nucleotide analogs and modifications that may be used. See Crooke, S. (ed.) Antisense Drug Technology: Principles, Strategies, and Applications (1st ed), Marcel Dekker; ISBN: 0824705661; 1st edition (2001) and references therein. For example, 2′-modifications include halo, alkoxy and allyloxy groups. In some embodiments, the 2′-OH group is replaced by a group selected from H, OR, R, halo, SH, SR, NH2, NHR, NR2 or CN, wherein R is C1-C6 alkyl, alkenyl, or alkynyl, and halo is F, Cl, Br, or I. Examples of modified linkages include phosphorothioate and 5′-N- phosphoramidite linkages.

[00197] Nucleic acids comprising a variety of different nucleotide analogs, modified backbones, or non-naturally occurring internucleoside linkages can be utilized in accordance with the present invention. Nucleic acids of the present invention may include natural nucleosides (i.e., adenosine, thymidine, guanosine, cytidine, uridine, deoxyadenosine, deoxythymidine, deoxyguanosine, and deoxycytidine) or modified nucleosides. Examples of modified nucleotides include base modified nucleoside (e.g., aracytidine, inosine, isoguanosine, nebularine, pseudouridine, 2,6-diaminopurine, 2- aminopurine, 2-thiothymidine, 3-deaza-5-azacytidine, 2′-deoxyuridine, 3-nitorpyrrole, 4-methylindole, 4-thiouridine, 4-thiothymidine, 2-aminoadenosine, 2-thiothymidine, 2-thiouridine, 5-bromocytidine, 5-iodouridine, inosine, 6-azauridine, 6-chloropurine, 7-deazaadenosine, 7-deazaguanosine, 8- azaadenosine, 8-azidoadenosine, benzimidazole, M1-methyladenosine, pyrrolo-pyrimidine, 2-amino- 6-chloropurine, 3-methyl adenosine, 5-propynylcytidine, 5-propynyluridine, 5-bromouridine, 5- fluorouridine, 5-methylcytidine, 7-deazaadenosine, 7-deazaguanosine, 8-oxoadenosine, 8- oxoguanosine, O(6)-methylguanine, and 2-thiocytidine), chemically or biologically modified bases (e.g., methylated bases), modified sugars (e.g., 2′-fluororibose, 2′-aminoribose, 2′-azidoribose, 2′-O- methylribose, L-enantiomeric nucleosides arabinose, and hexose), modified phosphate groups (e.g., phosphorothioates and 5′-N-phosphoramidite linkages), and combinations thereof. Natural and modified nucleotide monomers for the chemical synthesis of nucleic acids are readily available. In some cases, nucleic acids comprising such modifications display improved properties relative to nucleic acids consisting only of naturally occurring nucleotides. In some embodiments, nucleic acid modifications described herein are utilized to reduce and/or prevent digestion by nucleases (e.g. exonucleases, endonucleases, etc.). For example, the structure of a nucleic acid may be stabilized by including nucleotide analogs at the 3′ end of one or both strands order to reduce digestion.

[00198] Modified nucleic acids need not be uniformly modified along the entire length of the molecule. Different nucleotide modifications and/or backbone structures may exist at various positions in the nucleic acid. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid is not substantially affected. To give but one example, modifications may be located at any position of a nucleic acid targeting moiety such that the ability of the nucleic acid targeting moiety to specifically bind to the target is not substantially affected. The modified region may be at the 5′-end and/or the 3′-end of one or both strands. For example, modified nucleic acid targeting moieties in which approximately 1-5 residues at the 5′ and/or 3′ end of either of both strands are nucleotide analogs and/or have a backbone modification have been employed. The modification may be a 5′ or 3′ terminal modification. One or both nucleic acid strands may comprise at least 50% unmodified nucleotides, at least 80% unmodified nucleotides, at least 90% unmodified nucleotides, or 100% unmodified nucleotides.

[00199] Nucleic acids in accordance with the present invention may, for example, comprise a modification to a sugar, nucleoside, or internucleoside linkage such as those described in U.S. Patent Application Publications 2003/0175950, 2004/0192626, 2004/0092470, 2005/0020525, and 2005/0032733. The present invention encompasses the use of any nucleic acid having any one or more of the modification described therein. For example, a number of terminal conjugates, e.g., lipids such as cholesterol, lithocholic acid, aluric acid, or long alkyl branched chains have been reported to improve cellular uptake. Analogs and modifications may be tested using, e.g., using any appropriate assay known in the art, for example, to select those that result in improved delivery of a therapeutic or diagnostic agent, improved specific binding of an nucleic acid targeting moiety to a target, etc. In some embodiments, nucleic acids in accordance with the present invention may comprise one or more non-natural nucleoside linkages. In some embodiments, one or more internal nucleotides at the 3′-end, 5′-end, or both 3′- and 5′-ends of the nucleic acid targeting moiety are inverted to yield a linkage such as a 3′-3′ linkage or a 5′-5′ linkage.

[00200] In some embodiments, nucleic acids in accordance with the present invention are not synthetic, but are naturally-occurring entities that have been isolated from their natural environments.

[00201] Any method can be used to design novel nucleic acid targeting moieties (see, e.g., U.S. Pat. Nos. 6,716,583; 6,465,189; 6,482,594; 6,458,543; 6,458,539; 6,376,190; 6,344,318; 6,242,246; 6,184,364; 6,001,577; 5,958,691; 5,874,218; 5,853,984; 5,843,732; 5,843,653; 5,817,785; 5,789,163; 5,763,177; 5,696,249; 5,660,985; 5,595,877; 5,567,588; and 5,270,163; and U.S. Patent Application Publications 2005/0069910, 2004/0072234, 2004/0043923, 2003/0087301, 2003/0054360, and 2002/0064780). The present invention provides methods for designing novel nucleic acid targeting moieties. The present invention further provides methods for isolating or identifying novel nucleic acid targeting moieties from a mixture of candidate nucleic acid targeting moieties.

[00202] Nucleic acid targeting moieties that bind to a protein, a carbohydrate, a lipid, and/or a nucleic acid can be designed and/or identified. In some embodiments, nucleic acid targeting moieties can be designed and/or identified for use in the complexes of the invention that bind to proteins and/or characteristic portions thereof, such as tumor-markers, integrins, cell surface receptors, transmembrane proteins, intercellular proteins, ion channels, membrane transporter proteins, enzymes, antibodies, chimeric proteins etc. In some embodiments, nucleic acid targeting moieties can be designed and/or identified for use in the complexes of the invention that bind to carbohydrates and/or characteristic portions thereof, such as glycoproteins, sugars (e.g., monosaccharides, disaccharides and polysaccharides), glycocalyx (i.e., the carbohydrate-rich peripheral zone on the outside surface of most eukaryotic cells) etc. In some embodiments, nucleic acid targeting moieties can be designed and/or identified for use in the complexes of the invention that bind to lipids and/or characteristic portions thereof, such as oils, saturated fatty acids, unsaturated fatty acids, glycerides, hormones, steroids (e.g., cholesterol, bile acids), vitamins (e.g. vitamin E), phospholipids, sphingolipids, lipoproteins etc. In some embodiments, nucleic acid targeting moieties can be designed and/or identified for use in the complexes of the invention that bind to nucleic acids and/or characteristic portions thereof, such as DNA nucleic acids; RNA nucleic acids; modified DNA nucleic acids; modified RNA nucleic acids; and nucleic acids that include any combination of DNA, RNA, modified DNA, and modified RNA; etc.

[00203] Nucleic acid targeting moieties (e.g. aptamers or spiegelmers) may be designed and/or identified using any available method. In some embodiments, nucleic acid targeting moieties are designed and/or identified by identifying nucleic acid targeting moieties from a candidate mixture of nucleic acids. Systemic Evolution of Ligands by Exponential Enrichment (SELEX), or a variation thereof, is a commonly used method of identifying nucleic acid targeting moieties that bind to a target from a candidate mixture of nucleic acids.

[00204] Nucleic acid targeting moieties that bind selectively to any target can be isolated by the SELEX process, or a variation thereof, provided that the target can be used as a target in the SELEX process.

III. Pharmaceutical Formulations and Administration

[00205] The present invention further relates to a pharmaceutical formulation comprising a compound of the invention or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, and a pharmaceutical formulation comprising a combination of the invention.

[00206] The compounds described herein including pharmaceutically acceptable carriers such as addition salts or hydrates thereof, can be delivered to a patient using a wide variety of routes or modes of administration. Suitable routes of administration include, but inhalation, transdermal, oral, rectal, transmucosal, intestinal and parenteral administration, including intramuscular, subcutaneous and intravenous injections. Preferably, the compouds of the invention comprising an antibody or antibody fragment as the targeting moiety are administered parenterally, more preferably intravenously.

[00207] As used herein, the terms“administering” or“administration” are intended to encompass all means for directly and indirectly delivering a compound to its intended site of action.

[00208] The compounds described herein, or pharmaceutically acceptable salts and/or hydrates thereof, may be administered singly, in combination with other compounds of the invention, and/or in cocktails combined with other therapeutic agents. Of course, the choice of therapeutic agents that can be co-administered with the compounds of the invention will depend, in part, on the condition being treated.

[00209] For example, when administered to patients suffering from a disease state caused by an organism that relies on an autoinducer, the compounds of the invention can be administered in cocktails containing agents used to treat the pain, infection and other symptoms and side effects commonly associated with the disease. Such agents include, e.g., analgesics, antibiotics, etc.

[00210] When administered to a patient undergoing cancer treatment, the compounds may be administered in cocktails containing anti-cancer agents and/or supplementary potentiating agents. The compounds may also be administered in cocktails containing agents that treat the side-effects of radiation therapy, such as anti-emetics, radiation protectants, etc.

[00211] Supplementary potentiating agents that can be co-administered with the compounds of the invention include,e.g., tricyclic anti-depressant drugs (e.g., imipramine, desipramine, amitriptyline, clomipramine, trimipramine, doxepin, nortriptyline, protriptyline, amoxapine and maprotiline); non- tricyclic and anti-depressant drugs (e.g., sertraline, trazodone and citalopram); Ca+2 antagonists (e.g., verapamil, nifedipine, nitrendipine and caroverine); amphotericin; triparanol analogues (e.g., tamoxifen); antiarrhythmic drugs (e.g., quinidine); antihypertensive drugs (e.g., reserpine); thiol depleters (e.g., buthionine and sulfoximine); and calcium leucovorin.

[00212] The active compound(s) of the invention are administered per se or in the form of a pharmaceutical composition wherein the active compound(s) is in admixture with one or more pharmaceutically acceptable carriers, excipients or diluents. Pharmaceutical compositions for use in accordance with the present invention are typically formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.

[00213] For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.

[00214] For oral administration, the compounds can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, and suspensions for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxyniethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

[00215] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.

[00216] Pharmaceutical preparations, which can be used orally, include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.

[00217] For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.

[00218] For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.

[00219] The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Injection is a preferred method of administration for the compositions of the current invention. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

[00220] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions. For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.

[00221] Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.

[00222] The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.

[00223] In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation or transcutaneous delivery (e.g., subcutaneously or intramuscularly), intramuscular injection or a transdermal patch. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

[00224] The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include calcium carbonate, calcium phosate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.

[00225] A preferred pharmaceutical composition is a composition formulated for injection such as intravenous injection and includes about 0.01 % to about 100% by weight of the compound of the present invention, based upon 100% weight of total pharmaceutical composition. The drug- ligand conjugate may be an antibody-cytotoxin conjugatewhere the antibody has been selected to target a particular cancer.

[00226] In some embodiments, the pharmaceutical composition of the present invention further comprises an additional therapeutic agent.

[00227] In some embodiments, the additional therapeutic agent is an anticancer agent.

[00228] In some embodiments, the additional anticancer agent is selected from an antimetabolite, an inhibitor of topoisomerase I and II, an alkylating agent, a microtubule inhibitor, an antiandrogen agent, a GNRh modulator or mixtures thereof.

[00229] In some embodiments, the additional therapeutic agent is a chemotherapeutic agent. [00230] By“chemotherapeutic agent” herein is meant a chemical compound useful in the treatment of cancer. Examples are but not limited to: Gemcitabine, Irinotecan, Doxorubicin, 5-Fluorouracil, Cytosine arabinoside ("Ara-C"), Cyclophosphamide, Thiotepa, Busulfan, Cytoxin, TAXOL, Methotrexate, Cisplatin, Melphalan, Vinblastine and Carboplatin.

[00231] In some embodiments, the second chemotherapeutic agent is selected from the group consisting of tamoxifen, raloxifene, anastrozole, exemestane, letrozole, imatanib, paclitaxel, cyclophosphamide, lovastatin, minosine, gemcitabine, cytarabine, 5- fluorouracil, methotrexate, docetaxel, goserelin, vincristine, vinblastine,nocodazole, teniposide etoposide, gemcitabine, epothilone, vinorelbine, camptothecin, daunorubicin, actinomycin D, mitoxantrone, acridine, doxorubicin, epirubicin, or idarubicin.

IV. Kits

[00232] In another aspect, the present invention provides kits containing the therapeutic combinations provided herein and directions for using the therapeutic combinations. The kit may also include a container and optionally one or more vial, test tube, flask, bottle, or syringe. Other formats for kits will be apparent to those of skill in the art and are within the scope of the present invention. IV. Medical and Pharmaceutical Uses

[00233] In another aspect, the present invention provides a method for treating a disease condition in a subject that is in need of such treatment, comprising: administering to the subject a therapeteutic combination or pharmaceutical composition comprising a therapeutically effective amount of the compound of the present invention or a pharmaceutically acceptable salt thereof, and a pharmaceutical acceptable carrier.

[00234] In addition to the compositions and constructs described above, the present invention also provides a number of uses of the combinations of the invention. Uses of the combinations of the current invention include: killing or inhibiting the growth, proliferation or replication of a tumor cell or cancer cell, treating cancer, treating a pre-cancerous condition, preventing the multiplication of a tumor cell or cancer cell, preventing cancer, preventing the multiplication of a cell that expresses an auto-immune antibody. These uses comprise administering to an animal such as a mammal or a human in need thereof an effective amount of a compound of the present invention.

[00235] The combination of the current invention is useful for treating diseases such as cancer in a subject, such as a human being. Combinations and uses for treating tumors by providing a subject the composition in a pharmaceutically acceptable manner, with a pharmaceutically effective amount of a composition of the present invention are provided.

[00236] By“cancer” herein is meant the pathological condition in humans that is characterized by unregulated cell proliferation. Examples include but are not limited to: carcinoma, lymphoma, blastoma, and leukemia. More particular examples of cancers include but are not limited to: lung (small cell and non-small cell), breast, prostate, carcinoid, bladder, gastric, pancreatic, liver (hepatocellular), hepatoblastoma, colorectal, head and neck squamous cell carcinoma, esophageal, ovarian, cervical, endometrial, mesothelioma, melanoma, sarcoma, osteosarcoma, liposarcoma, thyroid, desmoids, chronic myelocytic leukemia (AML), and chronic myelocytic leukemia (CML).

[00237] By“uncontrolled growth”, we include an increase in the number and/or size of cancer cells (also referred to herein as“proliferation”). By“metastasis” we mean the movement or migration (e.g. invasiveness) of cancer cells from a primary tumor site in the body of a subject to one or more other areas within the subject’s body (where the cells can then form secondary tumors). Thus, in one embodiment the invention provides compounds and methods for inhibiting, in whole or in part, the formation of secondary tumors in a subject with cancer.

[00238] Advantageously, the compounds of the invention may be capable of inhibiting the proliferation and/or metastasis of cancer cells selectively.

[00239] By“selectively” we mean that the compounds of the invention may inhibit the proliferation and/or metastasis of cancer cells to a greater extent than it modulates the function (e.g. proliferation) of non-cancer cells. Preferably, the compounds of the invention inhibit the proliferation and/or metastasis of cancer cells only.

[00240] In another aspect, the present invention provides a pharmaceutical composition comprising the compound of the present invention and at least one pharmaceutically acceptable carrier or diluent, wherein said compound is in free form or in a pharmaceutically acceptable salt form. Such composition may be an oral composition, injectable composition or suppository. And the composition may be manufactured in a conventional manner by mixing, granulating or coating methods.

[00241] In an embodiment of the invention, the composition is an oral composition and it may be a tablet or gelatin capsule. Preferably, the oral composition comprises the present compound together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets, together with c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragamayth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; and if desired, d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) additives, e.g., absorbents, colorants, flavors and sweeteners.

[00242] In another embodiment of the invention, the composition is an injectable composition, and may be an aqueous isotonic solution or suspension.

[00243] In yet another embodiment of the invention, the composition is a suppository and may be prepared from fatty emulsion or suspension. [00244] Preferably, the composition is sterilized and/or contains adjuvant. Such adjuvant can be preserving, stabilizing, wetting or emulsifying agent, solution promoter, salt for regulating the osmotic pressure, buffer and/or any combination thereof.

[00245] Alternatively or in addition, the composition may further contain other therapeutically valuable substances for different applications, like solubilizers, stabilizers, tonicity enhancing agents, buffers and/ or preservatives.

[00246] In an embodiment of the invention, the composition may be a formulation suitable for transdermal application. Such formulation includes an effective amount of the compound of the present invention and a carrier. Preferably, the carrier may include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. A transdermal device contain the formulation may also be used. The transdermal device may be in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Otherwise, a matrix transdermal formulation may also be used.

[00247] In another embodiment of the invention, the composition may be a formulation suitable for topical application, such as to the skin and eyes, and may be aqueous solution, ointment, cream or gel well known in the art.

[00248] In another aspect, the present invention provides a method of inhibiting WNT secretion from a cell.

[00249] In one embodiment, the cell is contained within a mammal, and the administered amount is a therapeutically effective amount. In another embodiment, the inhibition of WNT signaling further results in the inhibition of the growth of the cell. In a further embodiment, the cell is a cancer cell. In yet another embodiment, the cell is a fibrogenic cell.

[00250] Cell proliferation is measured by using methods known to those skilled in the art. For example, a convenient assay for measuring cell proliferation is the CellTiter-Glo™ Assay commercially available from Promega (Madison, WI). The assay procedure involves adding the CellTiter-Glo® reagent to cells cultured on multi-well dishes. The luminescent signal, measured by a luminometer or an imaging device, is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture. In addition, cell proliferation may also be measured using colony formation assays known in the art.

[00251] The present invention also provides a method for treating cancers or fibroses related to the WNT signaling pathway with the present compound. Those skilled in the art would readily be able to determine whether a cancer is related to the Wnt pathway by analyzing cancer cells using one of several techniques known in the art. For example, one could examine cancer cells for aberrations in the levels of proteins or mRNAs involved in Wnt signaling using immune and nucleic acid detection methods.

[00252] Cancers or fibroses related to the Wnt pathway include those in which activity of one or more components of the Wnt signaling pathways are upregulated from basal levels. In one embodiment, inhibiting the Wnt pathway may involve inhibiting Wnt secretion. As another example, inhibiting the Wnt pathway may involve inhibiting components downstream of the cell surface receptors. In another embodiment, inhibition of Wnt secretion may involve inhibiting the activity of any of the proteins implicated in the secretion of functional WNTs.

[00253] Furthermore, the invention provides a method for treating a WNT pathway disorder in a subject suffering from the disorder by administering to the subject a therapeutically effective amount of a WNT inhibitor. In one embodiment, the disorder is a cell proliferative disorder associated with aberrant, e.g., increased, activity of WNT signaling. In another embodiment, the disorder results from increased amount of a WNT protein. In yet another embodiment, the cell proliferative disorder is cancer, include but are not limited to: lung (small cell and non-small cell), breast, prostate, carcinoid, bladder, gastric, pancreatic, liver (hepatocellular), hepatoblastoma, colorectal, head cancer and neck squamous cell carcinoma, esophageal, ovarian, cervical, endometrial, mesothelioma, melanoma, sarcoma, osteosarcoma, liposarcoma, thyroid, desmoids, chronic myelocytic leukemia (AML), and chronic myelocytic leukemia (CML). In yet another embodiment, the cell proliferative disorder is fibrosis, include but are not limited to: lung fibrosis, such as idiopathic pulmonary fibrosis and radiation-induced fibrosis, renal fibrosis and liver fibrosis including liver cirrhosis. In yet another embodiment, the disorder is osteoarthritis, Parkinson’s disease, retinopathy, macular degeneration.

[00254] For therapeutically use, the compound of the present invention could be administered in a therapeutically effective amount via any acceptable way known in the art singly. As used herein, the therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. Generally, the satisfactory result is indicated to be obtained systemically at a daily dosage of about 0.03 to 2.5 mg/kg per body weight of the subject. In one embodiment, the indicated daily dosage for larger mammal as human is in the range from about 0.5mg to about 100mg. Preferably, the compound is administered in divided doses up to four times a day or in retard form. In another embodiment, suitable unit dosage forms for oral administration comprise from ca.1 to 100 mg active ingredient.

[00255] Alternatively, the compound of the present invention may be administered in a therapeutically effective amount as the active ingredient in combination with one or more therapeutic agents, such as pharmaceutical combinations. There may be synergistic effects when the compound of the present invention is used with a chemotherapeutic agent known in the art. The dosage of the co- administered compounds could vary depending on the type of co-drug employed, the specific drug employed, the condition being treated and so forth.

[00256] The compound of the present invention or the composition thereof may be administered by any conventional route. In one embodiment, it is administered enterally, such as orally, and in the form of tablets or capsules. In another embodiment, it is administered parenterally and in the form of injectable solutions or suspensions. In yet another embodiment, it is administered topically and in the form of lotions, gels, ointments or creams, or in a nasal or suppository form.

[00257] In another aspect, the invention also provides a pharmaceutical combination, preferably, a kit, comprising a) a first agent which is the compound of the present invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent. In addition, the kit may comprise instructions for its administration.

[00258] The combination of the present invention may be used in vitro or in vivo. Preferably, the desired therapeutic benefit of the administration may be achieved by contacting cell, tissue or organism with a single composition or pharmacological formulation that includes the compound of the present invention and one or more agents, or by contacting the cell with two or more distinct compositions or formulations, wherein one composition includes one agent and the other includes another. The agents of the combination may be administered at the same time or separately within a period of time. Preferably, the separate administration can result in a desired therapeutic benefit. The present compound may precede, be co-current with and /or follow the other agents by intervals ranging from minutes to weeks. A person skilled in the art could generally ensure the interval of the time of each delivery, wherein the agents administered separately could still be able to exert an advantageously combined effect on the cell, tissue or organism. In one embodiment, it is contemplated that one may contact the cell, tissue or organism with two, three, four or more modalities substantially simultaneously as the candidate substance, i.e., with less than about one minute. In another embodiment, one or more agents may be administered about between 1 minute to 14 days.

[00259] By“inhibiting” or“treating” or“treatment” herein is meant to reduction, therapeutic treatment and prophylactic or preventative treatment, wherein the objective is to reduce or prevent the aimed pathologic disorder or condition. In one example, following administering of a compound of the present invention, a cancer patient may experience a reduction in tumor size. “Treatment” or “treating” includes (1) inhibiting a disease in a subject experiencing or displaying the pathology or symptoms of the disease, (2) ameliorating a disease in a subject that is experiencing or displaying the pathology or symptoms of the disease, and/or (3) affecting any measurable decrease in a disease in a subject or patient that is experiencing or displaying the pathology or symptoms of the disease. To the extent a compound of the present invention may prevent growth and/or kill cancer cells, it may be cytostatic and/or cytotoxic.

[00260] By“therapeutically effective amount” herein is meant an amount of a compound provided herein effective to“treat” a disorder in a subject or mammal. In the case of cancer, the therapeutically effective amount of the drug may either reduce the number of cancer cells, reduce the tumor size, inhibit cancer cell infiltration into peripheral organs, inhibit tumor metastasis, inhibit tumor growth to certain extent, and/or relieve one or more of the symptoms associated with the cancer to some extent.

[00261] Administration“in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order. As used herein, the term “pharmaceutical combination” refers to a product obtained from mixing or combining active ingredients, and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound of Formula (1) and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound of Formula (1) and a co- agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the active ingredients in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.

[00262] In another aspect, the present invention provides a method of treating a tumor/cancer in a subject comprising administering to the subject a therapeutically effective amount of the compounds of the present invention. In some embodiments, the tumor or cancer can be at any stage, e.g., early or advanced, such as a stage I, II, III, IV or V tumor or cancer. In some embodiments, the tumor or cancer can be metastatic or non-metastatic. In the context of metastasis, the methods of the present invention can reduce or inhibit metastasis of a primary tumor or cancer to other sites, or the formation or establishment of metastatic tumors or cancers at other sites distal from the primary tumor or cancer therapy. Thus, the methods of the present invention include, among other things, 1) reducing or inhibiting growth, proliferation, mobility or invasiveness of tumor or cancer cells that potentially or do develop metastases (e.g., disseminated tumor cells, DTC); 2) reducing or inhibiting formation or establishment of metastases arising from a primary tumor or cancer to one or more other sites, locations or regions distinct from the primary tumor or cancer; 3) reducing or inhibiting growth or proliferation of a metastasis at one or more other sites, locations or regions distinct from the primary tumor or cancer after a metastasis has formed or has been established; and 4) reducing or inhibiting formation or establishment of additional metastasis after the metastasis has been formed or established.

[00263] In some embodiments, the tumor or cancer is solid or liquid cel mass. A "solid" tumor refers to cancer, neoplasia or metastasis that typically aggregates together and forms a mass. Specific non-limiting examples include breast, ovarian, uterine, cervical, stomach, lung, gastric, colon, bladder, glial, and endometrial tumors/cancers, etc. A "liquid tumor," which refers to neoplasia that is dispersed or is diffuse in nature, as they do not typically form a solid mass. Particular examples include neoplasia of the reticuloendothelial or hematopoietic system, such as lymphomas, myelomas and leukemias. Non-limiting examples of leukemias include acute and chronic lymphoblastic, myeolblastic and multiple myeloma. Typically, such diseases arise from poorly differentiated acute leukemias, e.g., erythroblastic leukemia and acute megakaryoblastic leukemia. Specific myeloid disorders include, but are not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML). Lymphoid malignancies include, but are not limited to, acute lymphoblastic leukemia (ALL), which includes B-lineage ALL (B-ALL) and T-lineage ALL (T-ALL), chronic lymphocytic leukemia (CLL), prolymphocyte leukemia (PLL), hairy cell leukemia (HLL) and Waldenstroem's macroglobulinemia (WM). Specific malignant lymphomas include, non-Hodgkin lymphoma and variants, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Sternberg disease.

[00264] In some embodiments, the abnormal proliferation is of cancer cells.

[00265] In some embodiments, the cancer is selected from the group consisting of: breast cancer, colorectal cancer, diffuse large B-cell lymphoma, endometrial cancer, follicular lymphoma, gastric cancer, glioblastoma, head and neck cancer, hepatocellular cancer, lung cancer, melanoma, multiple myeloma, ovarian cancer, pancreatic cancer, prostate cancer, and renal cell carcinoma.

[00266] In some embodiments, the methods of the present invention can be practiced with other treatments or therapies (e.g., surgical resection, radiotherapy, ionizing or chemical radiation therapy, chemotherapy, immunotherapy, local or regional thermal (hyperthermia) therapy, or vaccination). Such other treatments or therapies can be administered prior to, substantially contemporaneously with (separately or in a mixture), or following administration of the compounds of the present invention.

[00267] In some embodiments, the methods of the present invention comprise administering a therapeutically effective amount of a compound of the present invention in combination with an additional therapeutic agent. In some embodiments, the additional therapeutic agent is an anticancer/antitumor agent. In some embodiments, the additional therapeutic agent is an antimetabolite, an inhibitor of topoisomerase I and II, an alkylating agent, a microtubule inhibitor, an antiandrogen agent, a GNRh modulator or mixtures thereof. In some embodiemnts, the additional therapeutic agent is selected from the group consisting of tamoxifen, raloxifene, anastrozole, exemestane, letrozole, imatanib, paclitaxel, cyclophosphamide, lovastatin, minosine, gemcitabine, cytarabine, 5- fluorouracil, methotrexate, docetaxel, goserelin, vincristine, vinblastine,nocodazole, teniposide etoposide, gemcitabine, epothilone, vinorelbine, camptothecin, daunorubicin, actinomycin D, mitoxantrone, acridine, doxorubicin, epirubicin, or idarubicin.

[00268] Administration“in combination with” one or more additional therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order. As used herein, the term “pharmaceutical combination” refers to a product obtained from mixing or combining active ingredients, and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound of Formula (1) and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound of Formula (1) and a co- agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the active ingredients in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.

[00269] In some embodiments, the present invention provides a compound for use in killing a cell. The compound is administered to the cell in an amount sufficient to kill said cell. In an exemplary embodiment, the compound is administered to a subject bearing the cell. In a further exemplary embodiment, the administration serves to retard or stop the growth of a tumor that includes the cell (e.g., the cell can be a tumor cell). For the administration to retard the growth, the rate of growth of the cell should be at least 10% less than the rate of growth before administration. Preferably, the rate of growth will be retarded at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or completely stopped.

[00270] Additionally, the present invention provides a compound or a pharmaceutical composition of the present invention for use as a medicament. The present invention also provides a compound or a pharmaceutical composition for killing, inhibiting or delaying proliferation of a tumor or cancer cell, or for treating a disease.

Effective Dosages

[00271] Pharmaceutical compositions suitable for use with the present invention include compositions wherein the active ingredient is contained in a therapeutically effective amount, i.e., in an amount effective to achieve its intended purpose. The actual amount effective for a particular application will depend, inter alia, on the condition being treated. Determination of an effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure herein.

[00272] For any compound described herein, the therapeutically effective amount can be initially determined from cell culture assays. Target plasma concentrations will be those concentrations of active compound(s) that are capable of inhibition cell growth or division. In preferred embodiments, the cellular activity is at least 25% inhibited. Target plasma concentrations of active compound(s) that are capable of inducing at least about 30%, 50%, 75%, or even 90% or higher inhibition of cellular activity are presently preferred. The percentage of inhibition of cellular activity in the patient can be monitored to assess the appropriateness of the plasma drug concentration achieved, and the dosage can be adjusted upwards or downwards to achieve the desired percentage of inhibition.

[00273] As is well known in the art, therapeutically effective amounts for use in humans can also be determined from animal models. For example, a dose for humans can be formulated to achieve a circulating concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring cellular inhibition and adjusting the dosage upwards or downwards, as described above.

[00274] A therapeutically effective dose can also be determined from human data for compounds which are known to exhibit similar pharmacological activities. The applied dose can be adjusted based on the relative bioavailability and potency of the administered compound as compared with the known compound.

[00275] Adjusting the dose to achieve maximal efficacy in humans based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.

[00276] In the case of local administration, the systemic circulating concentration of administered compound will not be of particular importance. In such instances, the compound is administered so as to achieve a concentration at the local area effective to achieve the intended result.

[00277] For use in the prophylaxis and/or treatment of diseases related to abnormal cellular proliferation, a circulating concentration of administered compound of about 0.001 µM to 20 µM is preferred, with about 0.01 µM to 5 µM being preferred.

[00278] Patient doses for oral administration of the compounds described herein, typically range from about 1 mg/day to about 10,000 mg/day, more typically from about 10 mg/day to about 1,000 mg/day, and most typically from about 50 mg/day to about 500 mg/day. Stated in terms of patient body weight, typical dosages range from about 0.01 to about 150 mg/kg/day, more typically from about 0.1 to about 15 mg/kg/day, and most typically from about 1 to about 10 mg/kg/day, for example 5 mg/kg/day or 3 mg/kg/day.

[00279] In at least some embodiments, patient doses that retard or inhibit tumor growth can be 1 µmol/kg/day or less. For example, the patient doses can be 0.9, 0.6, 0.5, 0.45, 0.3, 0.2, 0.15, or 0.1 µmol/kg/day or less (referring to moles of the drug). Preferably, the antibody with drug conjugates retards growth of the tumor when administered in the daily dosage amount over a period of at least five days. [00280] For other modes of administration, dosage amount and interval can be adjusted individually to provide plasma levels of the administered compound effective for the particular clinical indication being treated. For example, in one embodiment, a compound according to the invention can be administered in relatively high concentrations multiple times per day. Alternatively, it may be more desirable to administer a compound of the invention at minimal effective concentrations and to use a less frequent administration regimen. This will provide a therapeutic regimen that is commensurate with the severity of the individual's disease.

[00281] Utilizing the teachings provided herein, an effective therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the clinical symptoms demonstrated by the particular patient. This planning should involve the careful choice of active compound by considering factors such as compound potency, relative bioavailability, patient body weight, presence and severity of adverse side effects, preferred mode of administration and the toxicity profile of the selected agent.

[00282] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

[00283] REFERENCE:

Akiri G, Cherian MM, Vijayakumar S, Liu G, Bafico A, Aaronson SA. Wnt pathway aberrations including autocrine Wnt activation occur at high frequency in human non-small-cell lung carcinoma. Oncogene.2009 May 28;28(21):2163-72.

Bafico A, Liu G, Goldin L, Harris V, Aaronson SA. An autocrine mechanism for constitutive Wnt pathway activation in human cancer cells. Cancer Cell.2004 Nov;6(5):497-506.

Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov. 2006 Dec;5(12):997-1014.

Blom AB, van Lent PL, van der Kraan PM, van den Berg WB. To seek shelter from the WNT in osteoarthritis? WNT-signaling as a target for osteoarthritis therapy. Curr Drug Targets. 2010 May;11(5):620-9.

Boonen RA, van Tijn P, Zivkovic D. Wnt signaling in Alzheimer's disease: up or down, that is the question. Ageing Res Rev.2009 Apr;8(2):71-82. Camilli TC, Weeraratna AT. Striking the target in Wnt-y conditions: intervening in Wnt signaling during cancer progression. Biochem Pharmacol.2010 Sep 1;80(5):702-11.

Chan SL, Cui Y, van Hasselt A, Li H, Srivastava G, Jin H, Ng KM, Wang Y, Lee KY, Tsao GS, Zhong S, Robertson KD, Rha SY, Chan AT, Tao Q. The tumor suppressor Wnt inhibitory factor 1 is frequently methylated in nasopharyngeal and esophageal carcinomas. Lab Invest.2007 Jul;87(7):644- 50.

Chen B, Dodge ME, Tang W, Lu J, Ma Z, Fan CW, Wei S, Hao W, Kilgore J, Williams NS, Roth MG, Amatruda JF, Chen C, Lum L. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat Chem Biol.2009 Feb;5(2):100-7.

Cheng JH, She H, Han YP, Wang J, Xiong S, Asahina K, Tsukamoto H. Wnt antagonism inhibits hepatic stellate cell activation and liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2008;294(1):G39-49.

Chun JS, Oh H, Yang S, Park M. Wnt signaling in cartilage development and degeneration. BMB Rep. 2008 Jul 31;41(7):485-94.

Chien AJ, Moon RT. WNTS and WNT receptors as therapeutic tools and targets in human disease processes. Front Biosci.2007 Jan 1;12:448-57.

DeAlmeida VI, Miao L, Ernst JA, Koeppen H, Polakis P, Rubinfeld B. The soluble wnt receptor Frizzled-8CRD-hFc inhibits the growth of teratocarcinomas in vivo. Cancer Res. 2007 Jun 1;67(11):5371-9

D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK,Baetge EE. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol.2006 Nov;24(11):1392-401.

Herbst A, Kolligs FT. Wnt signaling as a therapeutic target for cancer. Method Mol Biol. 2007;361:63-91.

Hoeppner LH, Secreto FJ, Westendorf JJ. Wnt signaling as a therapeutic target for bone diseases. Expert Opin Ther Targets.2009 Apr;13(4):485-96.

Hwang I, Seo EY, Ha H. Wnt/beta-catenin signaling: a novel target for therapeutic intervention of fibrotic kidney disease. Arch Pharm Res.2009 Dec;32(12):1653-62.

Inestrosa NC, Arenas E. Emerging roles of Wnts in the adult nervous system. Nat Rev Neurosci.2010 Feb;11(2):77-86.

Kansara M, et al. Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice. J Clin Invest.2009 Apr;119(4):837-51 Lie DC, Colamarino SA, Song HJ, Désiré L, Mira H, Consiglio A, Lein ES, Jessberger S, Lansford H, Dearie AR, Gage FH. WNT signalling regulates adult hippocampal neurogenesis. Nature 437 (7063): 1370–5, 2005.

MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell.2009 Jul;17(1):9-26.

Mikels AJ, Nusse R. Wnts as ligands: processing, secretion and reception. Oncogene. 2006 Dec 4;25(57):7461-8.

Moon RT. Wnt/beta-catenin pathway. Sci STKE.;2005(271):cm1.

Morrisey EE. Wnt signaling and pulmonary fibrosis. Am J Pathol.2003 May;162(5):1393-7.

Nusse R. WNT signaling and stem cell control". Cell Res.18 (5): 523–7, 2008

Ouchi N, Higuchi A, Ohashi K, Oshima Y, Gokce N, Shibata R, Akasaki Y, Shimono A, Walsh K. Sfrp5 is an anti-inflammatory adipokine that modulates metabolic dysfunction in obesity. Science. 2010 Jul 23;329(5990):454-7.

Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature.2005 Apr 14;434(7035):843-50. Rhee CS, Sen M, Lu D, Wu C, Leoni L, Rubin J, Corr M, Carson DA. Wnt and frizzled receptors as potential targets for immunotherapy in head and neck squamous cell carcinomas. Oncogene.2002 Sep 26;21(43):6598-605.

Sullivan GJ, et al. Generation of functional human hepatic endoderm from human induced pluripotent stem cells. Hepatology.2010 Jan;51(1):329-35.

Takahashi-Yanaga F, Kahn M. Targeting Wnt signaling: can we safely eradicate cancer stem cells? Clin Cancer Res.2010 Jun 15;16(12):3153-62.

Ten Berge, D. et al. WNT signaling mediates self-organization and axis formation in embryoid bodies. Cell Stem Cell 3, 508–518, 2008.

Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, Henckaerts E, Bonham K, Abbott GW,Linden RM, Field LJ, Keller GM. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature.2008 May 22;453(7194):524-8.

EXAMPLES

[00284] The present invention is further exemplified, but not limited, by the following and Examples that illustrate the preparation of the compounds of the invention.

Example 1

[00285] In vivo evaluation in CT26 murine carcinoma syngeneic mouse model [00286] Materials and Methods

[00287] Animals: Species: Mus Musculus; Strain: BALB/c; Age: 6-8 weeks (estimated age at inoculation); Sex: female; Body weight: 18-20 g; and Animal supplier: Beijing HFK Bio-Technology Co. Ltd. (HFK, Beijing, China).

[00288] Animal housing: The animals were housed in individual ventilated cages (up to 5 mice per cage) under the following conditions:

− Temperature: 20~26ºC

− Humidity: 30-70%

− Light cycle: 12 hours light and 12 hours darkness

− Polysulfone IVC cage: size of 325 mm x 210 mm x 180 mm

− Bedding material: corn cob

− Diet: Mouse diet, C060 irradiation sterilized dry granule food. Animals will have free access during the entire study period.

− Water: RO water, autoclaved before using. Animals will have free access to sterile drinking water

− Cage identification label: number of animals, sex, strain, receiving date, treatment, study number, group number, and the starting date of the treatment, etc

− Animal identification: Animals will be marked by 1 ear coding (notch) 0 ear tag − Adapt housing: the animals will be adapted in the facility for at least 7 days.

[00289] Tumor inoculation: all mice were lightly anesthetized with isoflurane before implantation, and then each mouse will be inoculated subcutaneously at the right lower flank with CT-26 tumor cells (3 x 10 4 ) in 0.1 ml of PBS for tumor development. The treatments were started with CGX1321 at inoculation. Treatment of anti-PD-1 were started either at 5 days after inoculation or when the mean tumor size reaches approximately 50 mm 3 , whichever is earlier. The date of grouping is denoted as day 0 of post grouping (PG-D0).

[00290] Evaluation of CGX1321 treatment efficacy in combination with PD-1 antibody

[00291] Product identification: PD-1 antibody (clone RMP1-14, BioXCell, Lot No.: 5311-2/1214).

[00292] The major endpoint was to see if the tumor growth could be delayed or regressed. Tumor measurement is conducted twice weekly with a caliper and the tumor volume (mm 3 ) is estimated using the formula: TV=a × b 2 /2, where a and b are long and short diameters of a tumor, respectively.

[00293] Tumor growth inhibition (TGI): TGI% is an indication of antitumor effectiveness, and expressed as: TGI (%)=100 x (1-T/C). T and C are the mean tumor volume (or weight) of the treated and control groups, respectively, on a given day. [00294] After tumor cells inoculation, the animals will be checked daily for morbidity and mortality. At the time of routine monitoring, the animals were checked for any effects of tumor growth and treatments on normal behavior such as mobility, visual estimation of food and water consumption, body weight gain/loss (body weights will be measured twice weekly), eye/hair matting and any other abnormal effect. Death and observed clinical signs were recorded in the comment of datasheet for each animal in detail.

[00295] Flow cytometric analysis: at the end of treatment, lymphocytes from spleens and tumors were isolated and processed for antibody labeling. Cells were stained with mouse antibodies of CD3, CD4 CD8 and FOXP3.