Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION OF A PI3K-INHIBITOR WITH AN ANDROGEN RECEPTOR ANTAGONIST
Document Type and Number:
WIPO Patent Application WO/2019/002068
Kind Code:
A1
Abstract:
The present invention relates to : • combinations of : * a component A : which is a PI3K kinase inhibitor, such as copanlisib, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a physiologically acceptable salt, or a mixture of same, for example; * a component B : which is an androgen receptor antagonist, such as darolutamide or enzalutamide for example; and, optionally, * one or more pharmaceutical agents C; in which optionally either or both of said components A and B are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially; • use of such combinations : * in the treatment or prophylaxis of a cancer, particularly prostate cancer, or * for the preparation of a medicament for the treatment or prophylaxis of a cancer, particularly prostate cancer; • methods of treatment or prophylaxis of a cancer, particularly prostate cancer, in a subject, comprising administering to said subject a therapeutically effective amount of such a combination; • compositions containing such a combination, together with pharmaceutically acceptable ingredients; and • kits comprising such a combination.

Inventors:
HAENDLER BERNARD (DE)
Application Number:
PCT/EP2018/066524
Publication Date:
January 03, 2019
Filing Date:
June 21, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAYER CONSUMER CARE AG (CH)
BAYER PHARMA AG (DE)
International Classes:
A61K31/517; A61K31/4155; A61K31/433; A61K31/519; A61K45/06; A61P35/00
Domestic Patent References:
WO2016142313A12016-09-15
WO2016087488A12016-06-09
WO2015082322A12015-06-11
WO2008070150A12008-06-12
WO2012062743A12012-05-18
WO2012062745A12012-05-18
WO2012062748A12012-05-18
WO2004029055A12004-04-08
WO2012136553A12012-10-11
WO2011051540A12011-05-05
Foreign References:
US5023252A1991-06-11
US5011472A1991-04-30
Other References:
PAUL TOREN ET AL: "Combination AZD5363 with Enzalutamide Significantly Delays Enzalutamide-resistant Prostate Cancer in Preclinical Models", EUROPEAN UROLOGY, vol. 67, no. 6, 1 June 2015 (2015-06-01), AMSTERDAM, NL, pages 986 - 990, XP055505771, ISSN: 0302-2838, DOI: 10.1016/j.eururo.2014.08.006
BRETT?S. CARVER ET AL: "Reciprocal Feedback Regulation of PI3K and Androgen Receptor Signaling in PTEN-Deficient Prostate Cancer", CANCER CELL, vol. 19, no. 5, 1 May 2011 (2011-05-01), pages 575 - 586, XP055039150, ISSN: 1535-6108, DOI: 10.1016/j.ccr.2011.04.008
SCHWARTZ SARIT ET AL: "Feedback Suppression of PI3K[alpha] Signaling inPTEN-Mutated Tumors Is Relieved by Selective Inhibition of PI", CANCER CELL, CELL PRESS, US, vol. 27, no. 1, 24 December 2014 (2014-12-24), pages 109 - 122, XP029131333, ISSN: 1535-6108, DOI: 10.1016/J.CCELL.2014.11.008
NEAL D. SHORE: "Darolutamide (ODM-201) for the treatment of prostate cancer", EXPERT OPINION ON PHARMACOTHERAPY, vol. 18, no. 9, 13 June 2017 (2017-06-13), LONDON, UK, pages 945 - 952, XP055470984, ISSN: 1465-6566, DOI: 10.1080/14656566.2017.1329820
L.A. TORRE ET AL., CANCER EPIDEMIOL. BIOMARKERS PREV., vol. 25, 2016, pages 16 - 27
T. MITCHELL; D.E. NEAL, BR., J. CANCER., vol. 113, 2015, pages 193 - 198
GUNDEM ET AL., NATURE, vol. 520, 2015, pages 353 - 357
G. ATTARD ET AL., LANCET, vol. 387, 2016, pages 70 - 82
S.J. BAUMGART; B. HAENDLER, INT. J. MOL. SCI., vol. 18, 2017, pages E1017
T.A. YAP ET AL., NAT. REV. DRUG DISCOV., vol. 15, 2016, pages 699 - 718
C.S. GRASSO ET AL., NATURE, vol. 487, 2012, pages 239 - 243
J. HOFFMAN-CENSITS; W.K. KELLY, CLIN. CANCER RES., vol. 203, no. 19, pages 1335 - 1339
C.A. THOMPSON, AM. J. HEALTH SYST. PHARM., vol. 68, 2011, pages 960
G. GALETTI ET AL., CANCER TREAT. REV., vol. 57, 2017, pages 16 - 27
D. ROBINSON, CELL, vol. 161, 2015, pages 1215 - 1228
D.E. SPRATT ET AL., NAT. REV., vol. 13, 2016, pages 597 - 610
R. FERRALDESCHI ET AL., EUR. UROL., vol. 67, 2015, pages 795 - 802
D.H. HOVELSON ET AL., NEOPLASIA, vol. 17, 2015, pages 385 - 399
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
PURE APPL CHEM, vol. 45, 1976, pages 11 - 30
A.M. MOILANEN ET AL., SCI. REP., vol. 5, 2015, pages 12007
C. TRAN ET AL., SCIENCE, vol. 324, 2009, pages 787 - 790
POWELL, M.F. ET AL.: "Compendium of Excipients for Parenteral Formulations", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 52, no. 5, 1998, pages 238 - 311, XP009119027
STRICKLEY, R.G: "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 53, no. 6, 1999, pages 324 - 349
NEMA, S. ET AL.: "Excipients and Their Use in Injectable Products", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 51, no. 4, 1997, pages 166 - 171
GARCIA-ECHEVERRIA ET AL., ONCOGENE, vol. 27, 2008, pages 551 - 5526
AIELLO ET AL., NEW ENGL. J. MED., vol. 331, 1994, pages 1480
PEER ET AL., LAB. INVEST., vol. 72, 1995, pages 638
LOPEZ ET AL., INVEST. OPTHTHALMOL. VIS. SCI., vol. 37, 1996, pages 855
"Merck Index, 11th ed.", 1996, .
GOODMAN; GILMAN'S ET AL.: "The Pharmacological Basis of Therapeutics, 9th ed.", 1996, MCGRAW-HILL, pages: 1225 - 1287
N. LIU ET AL., MOL. CANCERTHER., vol. 12, 2013, pages 2319 - 2330
D.P. SUTHERLIN ET AL., J. MED. CHEM., vol. 54, 2011, pages 7579 - 7587
S.M. MAIRA ET AL., MOL. CANCER THER., vol. 7, 2008, pages 1851 - 1863
CHOU; TALALAY, ADV. ENZYME REGUL., vol. 22, 1984, pages 27 - 55
Attorney, Agent or Firm:
BIP PATENTS (DE)
Download PDF:
Claims:
CLAIMS

1. A combination of : o a component A : which is a PI3K kinase inhibitor, such as copanlisib, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a physiologically acceptable salt, or a mixture of same, for example ; o a component B : which is an androgen receptor antagonist, such as darolutamide or enzalutamide for example ; and, optionally, o one or more pharmaceutical agents C ; in which optionally either or both of said components A and B are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially.

2. The combination of at least two components, component A and component B, according to claim 1, in which said component A is a compound of general formula :

(I) or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, in which :

R1 represents -(CH2)n-(CH R4)-(CH2)m-N(R5)(R5') ;

R2 represents a heteroaryl optionally substituted with 1, 2 or 3 R6 groups ;

R3 represents alkyl or cycloalkyl ;

R4 represents hydrogen or alkoxy ;

R5 and R5' may be the same or different and are independently, hydrogen, alkyl, cycloalkylalkyl, or alkoxyalkyl or R5 and R5' may be taken together with the nitrogen atom to which they are bound to form a 3-7 membered nitrogen containing heterocyclic ring optionally containing at least one additional heteroatom selected from oxygen, nitrogen or sulfur and which may be optionally substituted with 1 or more R6' groups, or R4 and R5 may be taken together with the atoms to which they are bound to form a 5-6 membered nitrogen containing heterocyclic ring optionally containing 1 or more nitrogen, oxygen or sulfur atoms and which may be optionally substituted with 1 or more

R6' groups; each occurrence of R6 may be the same or different and is independently halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalklyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclic ring, heterocyclolalkyl, alkyl-OR7, alkyl-SR7, alkyl-N(R7)(R7'), alkyl-COR7,-CN, -COOR7, -CON (R7)(R7'), -OR7, -SR7, -N(R7)(R7'), or -N R7COR7 each of which may be optionally substituted with 1 or more R8 groups; each occurrence of R6' may be the same or different and is independently alkyl, cycloalkylalklyl, or alkyl-OR7; each occurrence of R7 and R7' may be the same or different and is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalklyl, cycloalkenyl, aryl, arylalkyl, heteroaryl, heterocyclic ring, heterocyclolalkyl, or heteroarylalkyl; each occurrence of R8 is independently nitro, hydroxy, cyano, formyl, acetyl, halogen, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkylalklyl, cycloalkenyl, aryl, arylalkyl, heteroaryl, heterocyclic ring, heterocyclolalkyl, or heteroarylalkyl; n is an integer from 1-4 and m is an integer from 0-4 with the proviso that when when R4 and R5 are taken together with the atoms to which they are bound to form a 3- 7 membered nitrogen containing ring, n + m < 4 ;

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. 3. The combination according to claim 1 or 2, in which said component A is a compound of formula (I) of claim 2, in which R4 and R5 are taken together with the atoms to which they are bound to form a 5-6 membered nitrogen containing heterocyclic ring optionally containing 1 or more nitrogen, oxygen or sulfur atoms and which may be optionally substituted with 1 or more R6' groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

4. The combination according to any one of claims 1 to 3, in which said component A is a compound of formula (I) of claim 2, in which R2 is pyridine, pyridazine, pyrimidine, pyrazine, pyrole, oxazole, thiazole, furan or thiophene, optionally substituted with 1, 2 or 3 R6 groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

5. The combination according to any one of claims 1 to 4, in which said component A is a compound of formula (I), which has the formula :

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

6. The combination of claim 5, in which, in said compound of formula (I), R2 is pyridine, pyridazine, pyrimidine, pyrazine, pyrole, oxazole, thiazole, furan or thiophene, optionally substituted with 1, 2 or 3 R6 groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

7. The combination of any one of claims 1 to 6, in which said component A is a compound selected from the list consisting of :

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-methoxy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide ;

N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-methoxy-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-2,4-dimethyl-l,3-thiazole-5- carboxamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]-l,3-thiazole-5-carboxamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]isonicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]-4-methyl-l,3-thiazole-5-carboxamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-4-propylpyrimidine-5-carboxamide;

N-{8-[2-(4-ethylmorpholin-2-yl)ethoxy]-7-methoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl}nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}pyrimidine-5-carboxamide;

N-(8-{3-[2-(hydroxymethyl)morpholin-4-yl]propoxy}-7-methoxy-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide; N-(8-{3-[2-(hydroxymethyl)morpholin-4-yl]propoxy}-7-nnethoxy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-{8-[3-(dimethylamino)propoxy]-7-nnethoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide 1-oxide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl]-6-(2-pyrrolidin-l-ylethyl)nicotinamide;

6-(cyclopentylamino)-N-[7-nnethoxy-8-(3-nnorpholin-4-ylpropoxy)- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[8-(2-hydroxy-3-morpholin-4-ylpropoxy)-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-{7-methoxy-8-[3-(3-methylmorpholin-4-yl)propoxy]-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-(8-{3-[2-(hydroxymethyl)morpholin-4-yl]propoxy}-7-methoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-(8-{2-[4-(cyclobutylmethyl)morpholin-2-yl]ethoxy}-7-methoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-(7-methoxy-8-{2-[4-(2-methoxyethyl)morpholin-2-yl]ethoxy}-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-{8-[(4-ethylmorpholin-2-yl)methoxy]-7-methoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-(7-methoxy-8-{[4-(2-methoxyethyl)morpholin-2-yl]methoxy}-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-{7-methoxy-8-[(4-methylmorpholin-2-yl)methoxy]-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-4-carboxamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-4-carboxamide; N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-l-methyl-lH-innidazole-4-carboxannide;

rel-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-nnethoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)pyrimidine-5-carboxannide;

rel-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-nnethoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)-6-methylnicotinannide;

rel-6-acetamido-N-(8-{3-[(2R,6S)-2,6-dimethylnnorpholin-4-yl]propoxy}-7- methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinannide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl]-l-methyl-lH-innidazole-5-carboxannide;

6-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-methylnicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-4-methylpyrimidine-5-carboxannide;

6-amino-5-bromo-N-[7-nnethoxy-8-(3-nnorpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-l,3-oxazole-5-carboxamide;

N-[7-methoxy-8-(morpholin-2-ylmethoxy)-2,3-dihydroimidazo[l,2-c]quinazolin- 5-yl]nicotinamide;

2-{[2-(dimethylamino)ethyl]amino}-N-{8-[3-(dinnethylannino)propoxy]-7- methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl}pyrimidine-5-carboxamide; 2-amino-N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}-l,3-thiazole-5-carboxamide;

rel-2-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-nnethoxy-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)pyrimidine-5-carboxamide;

rel-6-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-nnethoxy-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

2-[(2-hydroxyethyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-carboxamide; N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-[(3-methoxypropyl)amino]pyrimidine-5-carboxamide; 2-amino-N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-[(3-morpholin-4-ylpropyl)amino]pyrimidine-5-carboxamide; 2-[(2-methoxyethyl)amino]-N-[7-nnethoxy-8-(3-nnorpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-carboxannide;

2-{[2-(dimethylamino)ethyl]amino}-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-carboxannide;

6-amino-N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-pyrrolidin-l-ylpyrimidine-5-carboxannide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-(4-methylpiperazin-l-yl)pyrimidine-5-carboxannide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-morpholin-4-ylpyrimidine-5-carboxannide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-piperazin-l-ylnicotinamide hydrochloride;

6-[(3S)-3-aminopyrrolidin-l-yl]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide hydrochloride hydrate;

6-[(3R)-3-aminopyrrolidin-l-yl]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide hydrochloride;

6-[(4-fluorobenzyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-[(2-furylmethyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-[(2-methoxyethyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide; N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-(lH-pyrrol-l-yl)nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-morpholin-4-ylnicotinamide;

N-{7-methoxy-8-[3-(methylannino)propoxy]-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

6-[(2,2-dimethylpropanoyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-[(cyclopropylcarbonyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-(2,2,2-trifluoroethoxy)nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-(trifluoromethyl)nicotinamide;

6-(isobutyrylamino)-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-{7-methoxy-8-[3-(4-methylpiperazin-l-yl)propoxy]-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-{[(methylamino)carbonyl]amino}-l,3-thiazole-4- carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-{[(methylamino)carbonyl]amino}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-(methylamino)-l,3-thiazole-4-carboxamide;

N-[7-methoxy-8-(2-morpholin-4-ylethoxy)-2,3-dihydroimidazo[l,2-c]quinazolin- 5-yl]nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}-2,4-dimethyl-l,3-thiazole-5-carboxamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}-6-methylnicotinamide; 6-{[(isopropylamino)carbonyl]amino}-N-[7-methoxy-8-(3-morpholin-4- ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-pyrrolidin-l-ylnicotinamide;

6-(dimethylamino)-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-piperidin-l-ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin- 5-yl]nicotinamide;

N-[7-methoxy-8-(2-pyrrolidin-l-ylethoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-

5- yl]nicotinamide;

N-[7-methoxy-8-(2-piperidin-l-ylethoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5- yl]nicotinamide;

6- {[(ethylamino)carbonyl]amino}-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-fluoro-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-l,3-oxazole-4-carboxamide;

2-(ethylamino)-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]-l,3-thiazole-4-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrazine-2-carboxamide;

N-[8-(2-aminoethoxy)-7-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5- yl]nicotinamide;

6-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]isonicotinamide;

N-{8-[3-(diethylamino)propoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}nicotinamide; N 8-[2-(diisopropylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N 8-[2-(diethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin

5- yl}nicotinamide;

N-{8-[3-(dimethylamino)propoxy]-7-nnethoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-nnethoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl]-2-(methylamino)pyrinnidine-5-carboxannide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl]-2-(methylthio)pyrimidine-5-carboxannide;

N-[8-(3-aminopropoxy)-7-methoxy-2,3-dihydroinnidazo[l,2-c]quinazolin-5- yl] nicotinamide trifluoroacetate;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]thiophene-2-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2,4-dimethyl-l,3-thiazole-5-carboxamide;

2-methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2 c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-3-furamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]thiophene-3-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2-methyl-l,3-thiazole-4-carboxamide;

6- methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2 c]quinazolin-5-yl]nicotinamide;

5-methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2 c]quinazolin-5-yl]nicotinamide; N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-methylnicotinamide;

6-(acetylamino)-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]nicotinamide ; preferably, N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-6-methylnicotinamide;

5- methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]-2,4-dimethyl-l,3-thiazole-5-carboxamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydroinnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

6- {[(isopropylamino)carbonyl]amino}-N-[7-methoxy-8-(3-nnorpholin-4- ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}-2,4-dimethyl-l,3-thiazole-5-carboxamide;

N-[7-methoxy-8-(2-morpholin-4-ylethoxy)-2,3-dihydroimidazo[l,2-c]quinazolin- 5-yl]nicotinamide;

rel-6-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-methoxy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

rel-2-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-methoxy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)pyrimidine-5-carboxamide; 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N 8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl}pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

8. The combination of any one of claims 1 to 7, in which said component A is 2-amino- N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5- yl]pyrimidine-5-carboxamide,

or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a physiologically acceptable salt, or a mixture of same. 9. The combination of any one of claims 1 to 8, in which said component A is 2-amino- N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5- yl]pyrimidine-5-carboxamide.

10. The combination of any one of claims 1 to 8, in which said component A is 2-amino- N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5- yl]pyrimidine-5-carboxamide dihydrochloride.

11. The combination of any one of claims 1 to 10, in which component B is darolutamide or enzalutamide.

12. The combination of any one of claims 1 to 11, in which component B is darolutamide.

13. The combination of any one of claims 1 to 11, in which component B is enzalutamide.

14. The combination according to any one of claims 1 to 7, wherein said component A is 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide and said component B is darolutamide.

15. The combination according to any one of claims 1 to 7, wherein said component A is 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide dihydrochloride and said component B is darolutamide.

16. The combination according to any one of claims 1 to 7, wherein said component A is 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide and said component B is enzalutamide.

17. The combination according to any one of claims 1 to 7, wherein said component A is 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide dihydrochloride and said component B is enzalutamide.

18. A combination according to any one of claims 1 to 17 for use in the treatment or prophylaxis of a cancer, particularly prostate cancer.

19. Use of a combination according to any one of claims 1 to 17 in the treatment or prophylaxis of a cancer, particularly prostate cancer.

20. Use of a combination according to any one of claims 1 to 17 for the preparation of a medicament for the treatment or prophylaxis of a cancer, particularly prostate cancer.

21. A method of treatment or prophylaxis of a cancer, particularly prostate cancer, in a subject, comprising administering to said subject a therapeutically effective amount of a combination according to any one of claims 1 to 17.

22. A composition containing a combination according to claims 1 to 17 together with pharmaceutically acceptable ingredients.

23. A kit comprising a combination according to any one of claims 1 to 17.

Description:
COMBINATION OF A PI3K-INHIBITOR WITH AN ANDROGEN RECEPTOR

ANTAGONIST

The present invention relates to :

• combinations of :

o a component A : which is a PI3K kinase inhibitor, such as copanlisib, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a physiologically acceptable salt, or a mixture of same, for example ;

o a component B : which is an androgen receptor antagonist, such as darolutamide or enzalutamide for example ; and, optionally, o one or more pharmaceutical agents C ;

in which optionally either or both of said components A and B are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially ;

• use of such combinations :

o in the treatment or prophylaxis of a cancer, particularly prostate cancer, or

o for the preparation of a medicament for the treatment or prophylaxis of a cancer, particularly prostate cancer ;

• methods of treatment or prophylaxis of a cancer, particularly prostate cancer, in a subject, comprising administering to said subject a therapeutically effective amount of such a combination ;

• compositions containing such a combination, together with pharmaceutically acceptable ingredients ; and

• kits comprising such a combination.

Component A may be administered by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.

Component B may be administered by the oral, intravenous, topical, local installations, intraperitoneal or nasal route. BACKGROUND to the INVENTION

Cancer is the second most prevalent cause of death in the United States, causing 450,000 deaths per year. While substantial progress has been made in identifying some of the likely environmental and hereditary causes of cancer, there is a need for additional therapeutic modalities that target cancer and related diseases. In particular there is a need for therapeutic methods for treating diseases associated with dysregulated growth / proliferation.

Cancer is a complex disease arising after a selection process for cells with acquired functional capabilities like enhanced survival / resistance towards apoptosis and a limitless proliferative potential. Thus, it is preferred to develop drugs for cancer therapy addressing distinct features of established tumors.

Prostate cancer is the most frequent cancer affecting men in developed countries, with an increasing incidence linked to the general ageing of the population (L.A. Torre et al., Cancer Epidemiol. Biomarkers Prev., 2016, 25:16-27). It is characterized by a high heterogeneity, due to the multifocal origin of the tumor and the genetic and epigenetic changes that accumulate during the long and usually slow progression of the disease (T. Mitchell and D.E. Neal, Br. J. Cancer. 2015, 113:193-198; G. Gundem et al., Nature, 2015, 520:353-357; G. Attard et al., Lancet, 2016, 387:70-82 ; S.J. Baumgart and B. Haendler, Int. J. Mol. Sci., 2017, 18:E1017).

Local therapies such as surgery and irradiation are the first options for confined prostate cancer but later stages are treated with androgen deprivation based on surgical castration and medical castration with gonadotropin-releasing hormone receptor agonists or antagonists (T.A. Yap et al., Nat. Rev. Drug Discov., 2016, 15:699- 718). This usually leads to a rapid response but, unfortunately, therapy resistance almost invariably appears and patients then progress to castration-resistant prostate cancer (CRPC). Nonetheless, androgen signalling is usually still maintained and essential during late stage prostate cancer, as documented by the overexpression and mutations reported for the androgen receptor (AR) in many tumor samples (C.S. Grasso et al., Nature, 2012, 487, 239-243). In line with this, the AR antagonist enzalutamide and the androgen synthesis inhibitor abiraterone have shown significant efficacy in clinical studies and are now approved for metastatic CRPC (J. Hoffman-Censits and W.K. Kelly, Clin. Cancer Res., 203, 19:1335-1339; C.A. Thompson, Am. J. Health Syst. Pharm., 2011, 68:960). Unfortunately these treatments only help for about 18 months, after which few therapy options are currently available (G. Galetti et al., Cancer Treat. Rev., 2017, 57:16-27). Numerous reports show that beside AR signaling, the PI3K/AKT pathway also plays a causal role in prostate cancer progression (C.S. Grasso et al., Nature, 2012, 487, 239- 243; D. Robinson, Cell, 2015, 161:1215-1228; D.E. Spratt et al., Nat. Rev., 2016, 13:597- 610). It is involved in tumor proliferation, survival and metabolism. Both in primary and in advanced prostate tumors, mutations are often observed in PTEN, a tumor suppressor which normally restrains activity of the PI3K/AKT pathway. Abnormalities in additional players involved in this pathway such as PIK3CA, PIK3CB, PIK3R1 and MTOR have also been reported, but less frequently. A poor clinical outcome and a worse response to abiraterone treatment have been reported for patients harboring such mutations (R. Ferraldeschi et al., Eur. Urol., 2015, 67:795-802). Importantly, a reciprocal regulation of the AR and PI3K/AKT signaling pathways has been claimed (D.H. Hovelson et al., Neoplasia, 2015, 17:385-399).

All these studies show that both the AR and the PI3K signaling pathways play essential roles in prostate cancer progression. It is therefore desirable to demonstrate that the combination of compounds that block either of these pathways possesses superior efficacy in controlling prostate cancer cell proliferation.

Different PI3K inhibitors are disclosed in e.g. WO2008/070150, WO2012/062743, WO2012/062745, WO2012/062748. However, the state of the art does not disclose the combinations of the present invention comprising the specific PI3K inhibitor copanlisib, or a physiologically acceptable salt thereof, and darolutamide or enzalutamide.

SUMMARY of the INVENTION

Surprisingly, it was observed that by administering of copanlisib or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, in combination with darolutamide or enzalutamide, a synergistic anti-proliferative effect could be achieved in cell lines representing human prostate cancer : the present invention describes the antiproliferative activity of the combination of PI3K inhibitors and androgen receptor antagonists in prostate cancer cell lines and reveals the synergistic effects of these two compound classes.

Hence, in accordance with a first aspect, the present invention relates to :

• combinations of :

o a component A : which is a PI3K kinase inhibitor, such as copanlisib, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a physiologically acceptable salt, or a mixture of same, for example ;

o a component B : which is an androgen receptor antagonist, such as darolutamide or enzalutamide for example ; and, optionally, o one or more pharmaceutical agents C ;

in which optionally either or both of said components A and B are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially.

In accordance with a second aspect, the present invention relates to :

• use of such combinations according to the first aspect of the present invention :

o in the treatment or prophylaxis of a cancer, particularly prostate cancer, or

o for the preparation of a medicament for the treatment or prophylaxis of a cancer, particularly prostate cancer.

In accordance with a third aspect, the present invention relates to :

• methods of treatment or prophylaxis of a cancer, particularly prostate cancer, in a subject, comprising administering to said subject a therapeutically effective amount of such combinations according to the first aspect of the present invention.

In accordance with a fourth aspect, the present invention relates to :

• compositions containing such a combination, together with pharmaceutically acceptable ingredients.

In accordance with a fifth aspect, the present invention relates to :

• kits comprising such combinations according to the first aspect of the present invention.

In the second and third aspects, component A may be administered by the oral, intravenous, topical, local installations, intraperitoneal or nasal route, and component B may be administered by the oral, intravenous, topical, local installations, intraperitoneal or nasal route. DETAILED DESCRIPTION OF THE INVENTION

Definitions The terms as mentioned in the present text have preferably the following meanings :

The term " alkyl " refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing solely of carbon and hydrogen atoms, containing no unsaturation, having from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, such as illustratively, methyl, ethyl, n-propyl 1-methylethyl (isopropyl), n-butyl, n-pentyl, and 1,1-dimethylethyl (t-butyl).

The term "alkenyl " refers to an aliphatic hydrocarbon group containing a carbon- carbon double bond and which may be a straight or branched or branched chain having about 2 to about 10 carbon atoms, e.g., ethenyl, 1-propenyl, 2-propenyl (allyl), iso-propenyl, 2-methyl-l-propenyl, 1-butenyl, 2-and butenyl.

The term "alkynyl" refers to a straight or branched chain hydrocarbonyl radicals having at least one carbon-carbon triple bond, and having in the range of about 2 up to 12 carbon atoms (with radicals having in the range of about 2 up to 10 carbon atoms presently being preferred) e.g., ethynyl.

The term "alkoxy" denotes an alkyl group as defined herein attached via oxygen linkage to the rest of the molecule. Representative examples of those groups are methoxy and ethoxy.

The term "alkoxyakyl" denotes an alkoxy group as defined herein attached via oxygen linkage to an alkyl group which is then attached to the main structure at any carbon from alkyl group that results in the creation of a stable structure the rest of the molecule. Representative examples of those groups are -CH2OCH3, - CH2OC2H5 .

The term "cycloalkyi" denotes a non-aromatic mono or multicyclic ring system of about 3 to 12 carbon atoms such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and examples of multicyclic cycloalkyi groups include perhydronapththyl, adamantyl and norbornyl groups bridged cyclic group or sprirobicyclic groups e.g spiro (4,4) non-2-yl.

The term "cycloalkylalkyl" refers to cyclic ring-containing radicals containing in the range of about 3 up to 8 carbon atoms directly attached to alkyl group which is then also attached to the main structure at any carbon from the alkyl group that results in the creation of a stable structure such as cyclopropylmethyl, cyclobutylethyl, cyclopentylethyl.

The term "aryl" refers to aromatic radicals having in the range of 6 up to 14 carbon atoms such as phenyl, naphthyl, tetrahydronapthyl, indanyl, biphenyl .

The term "arylalkyl" refers to an aryl group as defined herein directly bonded to an alkyl group as defined herein which is then attached to the main structure at any carbon from alkyl group that results in the creation of a stable structure the rest of the molecule, e.g., -CH2C6H5, -C2H5C6H5 .

The term "heterocyclic ring" refers to a stable 3- to 15 membered ring radical which consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, phosphorus, oxygen and sulfur. For purposes of this invention, the heterocyclic ring radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused, bridged or spiro ring systems, and the nitrogen, phosphorus, carbon, oxygen or sulfur atoms in the heterocyclic ring radical may be optionally oxidized to various oxidation states. In addition, the nitrogen atom may be optionally quaternized; and the ring radical may be partially or fully saturated (i.e., heteroaromatic or heteroaryl aromatic). Examples of such heterocyclic ring radicals include, but are not limited to, azetidinyl, acridinyl, benzodioxolyl, benzodioxanyl, benzofurnyl, carbazolyl cinnolinyl dioxolanyl, indolizinyl, naphthyridinyl, perhydroazepinyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazil, pyridyl, pteridinyl, purinyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrazoyl, imidazolyl tetrahydroisouinolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxoazepinyl, azepinyl, pyrrolyl, 4-piperidonyl, pyrrolidinyl, pyrazinyl, pyrimidinyl pyridazinyl, oxazolyl oxazolinyl oxasolidinyl, triazolyl, indanyl, isoxazolyl, isoxasolidinyl, morpholinyl, thiazolyl, thiazolinyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, indolyl, isoindolyl, indolinyl, isoindolinyl, octahydroindolyl, octahydroisoindolyl quinolyl, isoquinolyl, decahydroisoquinolyl, benzimidazolyl, thiadiazolyl, benzopyranyl, benzothiazolyl, benzooxazolyl, furyl, tetrahydrofurtyl, tetrahydropyranyl, thienyl, benzothienyl, thiamorpholinyl, thiamorpholinyl sulfoxide thiamorpholinyl sulfone, dioxaphospholanyl, oxadiazolyl, chromanyl, isochromanyl.

The term "heteroaryl" refers to heterocyclic ring radical as defined herein which are aromatic. The heteroaryl ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure.

The heterocyclic ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. The term "heteroarylalkyi" refers to heteroaryl ring radical as defined herein directly bonded to alkyl group. The heteroarylalkyi radical may be attached to the main structure at any carbon atom from alkyl group that results in the creation of a stable structure. The term "heterocyclyl" refers to a heterocylic ring radical as defined herein. The heterocyclyl ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure.

The term "heterocyclylalkyl" refers to a heterocylic ring radical as defined herein directly bonded to alkyl group. The heterocyclylalkyl radical may be attached to the main structure at carbon atom in the alkyl group that results in the creation of a stable structure.

The term "carbonyl" refers to an oxygen atom bound to a carbon atom of the molecule by a double bond.

The term "halogen" refers to radicals of fluorine, chlorine, bromine and iodine.

The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties.

Ring system substituent means a substituent attached to an aromatic or nonaromatic ring system which, for example, replaces an available hydrogen on the ring system.

As used herein, the term "one or more times", e.g. in the definition of the substituents of the compounds of the present invention (e.g. component A, B or C), is understood as meaning "one, two, three, four or five times, particularly one, two, three or four times, more particularly one, two or three times, even more particularly one or two times".

Where the plural form of the word compounds, salts, polymorphs, hydrates, solvates and the like, is used herein, this is taken to mean also a single compound, salt, polymorph, isomer, hydrate, solvate or the like. By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.

The term "carbonyl" refers to an oxygen atom bound to a carbon atom of the molecule by a double bond.

The compounds of this invention may contain one or more asymmetric centers, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms may be present in the (/?)- and/or (Sj-configuration, resulting in racemic mixtures in the case of a single asymmetric center, and diastereomeric mixtures in the case of multiple asymmetric centers. In certain instances, asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds. Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention. Preferred compounds are those, which produce the more desirable biological activity. Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of this invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.

Tautomers, sometimes referred to as proton-shift tautomers, are two or more compounds that are related by the migration of a hydrogen atom accompanied by the switch of one or more single bonds and one or more adjacent double bonds. The compounds of this invention may exist in one or more tautomeric forms. For example, a compound of Formula I may exist in tautomeric form la, tautomeric form lb, or tautomeric form lc, or may exist as a mixture of any of these forms. It is intended that all such tautomeric forms are included within the scope of the present invention.

The present invention also relates to useful forms of the compounds as disclosed herein, such as pharmaceutically acceptable salts, co-precipitates, metabolites, hydrates, solvates and prodrugs of all the compounds of examples. The term "pharmaceutically acceptable salt" refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19. Pharmaceutically acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succinic acid and citric acid. Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an appropriate base to form, e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts. Those skilled in the art will further recognize that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.

Representative salts of the compounds of this invention include the conventional nontoxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, sulfate, tartrate, thiocyanate, tosylate, and undecanoate.

Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, or butyl chlorides, bromides and iodides; dialkyi sulfates like dimethyl, diethyl, dibutyl sulfate, or diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.

A solvate for the purpose of this invention is a complex of a solvent and a compound of the invention in the solid state. Exemplary solvates would include, but are not limited to, complexes of a compound of the invention with ethanol or methanol. Hydrates are a specific form of solvate wherein the solvent is water.

Constituents which are optionally substituted as stated herein, may be substituted, unless otherwise noted, one or more times, independently from one another at any possible position. When any variable occurs more than one time in any constituent, each definition is independent.

The heteroarylic, or heterocyclic groups mentioned herein can be substituted by their given substituents or parent molecular groups, unless otherwise noted, at any possible position, such as e.g. at any substitutable ring carbon or ring nitrogen atom. Analogously it is being understood that it is possible for any heteroaryl or heterocyclyl group to be attached to the rest of the molecule via any suitable atom if chemically suitable. Unless otherwise noted, any heteroatom of a heteroarylic ring with unsatisfied valences mentioned herein is assumed to have the hydrogen atom(s) to satisfy the valences. Unless otherwise noted, rings containing quaterniza ble amino- or imino-type ring nitrogen atoms (-N=) may be prefera bly not quaternized on these amino- or imino-type ring nitrogen atoms by the mentioned substituents or parent molecular groups.

Preferred compounds are those which produce the more desira ble biological activity. Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of this invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques already known in the art.

The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers. Examples of appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid. Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation. The optically active bases or acids are then liberated from the separated diastereomeric salts. A different process for separation of optical isomers involves the use of chiral chromatography (e.g., chiral H PLC columns), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers. Suitable chiral H PLC columns are manufactured by Diacel, e.g., Chiracel OD and Chiracel OJ among many others, all routinely selectable. Enzymatic separations, with or without derivatisation, are also useful. The optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.

If in the context of the invention "em bodiment" is mentioned it should be understood to include a plurality of possible combinations.

In order to limit different types of isomers from each other reference is made to l U PAC Rules Section E (Pure Appl Chem 45, 11-30, 1976). The invention also includes all suitable isotopic variations of a compound of the invention. An isotopic variation of a compound of the invention is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature. Examples of isotopes that can be incorporated into a compound of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 17 0, 18 0, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 CI, 82 Br, 123 l, 124 l, 129 l and 131 l, respectively. Certain isotopic variations of a compound of the invention, for example, those in which one or more radioactive isotopes such as 3 H or 14 C are incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopic variations of a compound of the invention can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.

The present invention includes all possible stereoisomers of the compounds of the present invention as single stereoisomers, or as any mixture of said stereoisomers, in any ratio. Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of the present invention may be achieved by any suitable state of the art method, such as chromatography, especially chiral chromatography, for example.

The present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio. Furthermore, the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorphs, or as a mixture of more than one polymorph, in any ratio. Component A of the combinations of the present invention

Component A can be selected from inhibitors of PI3K-kinase specifically or generically disclosed e.g. in the publications as mentioned above which are incorporated herein by reference.

In an embodiment, said component A is a compound of general formula (A) :

(A)

in which

X represents CR 5 R 6 or N H;

Y 1 represents CR 3 or N;

the chemical bond between γ 2 ==γ 3 represents a single bond or double bond, with the proviso that when theY 2 =Y 3 represents a double bond, Y 2 and Y 3 independently represent CR 4 or N, and

when Y 2 ==Y 3 represents a single bond, Y 2 and Y 3 independently represent CR 3 R 4 or N R 4 ; Z 1 , Z 2 , Z 3 and Z 4 independently represent CH , CR 2 or N; represents aryl optionally having 1 to 3 substituents selected from R 11 , C3-8 cycloalkyl optionally having 1 to 3 substituents selected from R 11 , Ci-6 alkyl optionally substituted by aryl, heteroaryl, Ci-6 alkoxyaryl, aryloxy, heteroaryloxy or one or more halogen,

Ci-6 alkoxy optionally substituted by carboxy, aryl, heteroaryl, Ci-6 alkoxyaryl, aryloxy, heteroaryloxy or one or more halogen,

or

a 3 to 15 membered mono- or bi-cyclic heterocyclic ring that is saturated or unsaturated, optionally having 1 to 3 substituents selected from R 11 , and contains 1 to 3 heteroatoms selected from the group consisting of N, O and S, wherein

R 11 represents halogen, nitro, hydroxy, cyano, carboxy, amino, N- (Ci-6alkyl)amino, N-(hydroxyCi-6alkyl)amino, N,N-di(Ci-6alkyl)amino, N- (Ci-6acyl)amino, N-(formyl)-N-(Ci-6alkyl)amino, N-(Ci-6alkanesulfonyl) amino, N-(carboxyCi-6alkyl)-N-(Ci-6alkyl)amino, N-(Ci-

6alkoxycabonyl)amino, N-[N,N-di(Ci-6alkyl)amino methylene]amino, N- [N,N-di(Ci-6alkyl)amino (Ci-6alkyl)methylene]amino, N-[N,N-di(Ci- 6alkyl)amino C2-6alkenyl]amino, aminocarbonyl, N-

(Ci-6alkyl)aminocarbonyl, N,N-di(Ci-6alkyl)aminocarbonyl, C3-scycloalkyl, Ci-6 alkylthio, Ci-6alkanesulfonyl, sulfamoyl, Ci-6al koxycarbonyl,

N-arylamino wherein said aryl moiety is optionally having 1 to 3 substituents selected from R 101 , N-(aryl Ci-6alkyl)amino wherein said aryl moiety is optionally having 1 to 3 substituents selected from R 101 , aryl Ci-6alkoxycarbonyl wherein said aryl moiety is optionally having 1 to 3 substituents selected from R 101 ,

Ci-6alkyl optionally substituted by mono-, di- or tri- halogen, amino, N- (Ci-6alkyl)amino or N,N-di(Ci-6alkyl)amino, Ci-6alkoxy optionally su bstituted by mono-, di- or tri- halogen, N-

(Ci-6alkyl)sulfonamide, or N-(aryl)sulfonamide,

or

a 5 to 7 membered saturated or unsaturated ring having 1 to 3 heteroatoms selected from the group consisting of O, S and N, and optionally having 1 to 3 substituents selected from R 101 wherein

R 101 represents halogen, carboxy, amino, N-(Ci-6 alkyl)amino, N,N-di(Ci- 6alkyl)amino, aminocarbonyl, N-(Ci-6alkyl)aminocarbonyl, N,N- di(Ci-6alkyl)aminocarbonyl, pyridyl,

Ci-6 alkyl optionally substituted by cyano or mono- di- or tri- halogen,

and

Ci-6alkoxy optionally su bstituted by cyano, carboxy, amino, N-(Ci-6 alkyl)amino, N,N-di(Ci-6alkyl)amino, aminocarbonyl, N-(Ci- 6alkyl)aminocarbonyl, N, N-di(Ci-6alkyl)aminocarbonyl or mono-, di- or tri- halogen; represents hydroxy, halogen, nitro, cyano, amino, N-(Ci-6alkyl)amino, N, N-di(Ci-6alkyl)amino, N-(hydroxyCi-6alkyl)amino, N-(hydroxyCi-6alkyl)- N-(Ci-6alkyl)amino, Ci-6 acyloxy, aminoCi-6 acyloxy, C2-6alkenyl, aryl, a 5-7 membered saturated or unsaturated heterocyclic ring having 1 to 3 heteroatoms selected from the group consisting O, S and N, and optionally su bstituted by hydroxy, Ci-6 alkyl, Ci-6 alkoxy, oxo, amino, amino Ci-ealkyl, N- (Ci-6alkyl)amino, N, N-di(Ci-6alkyl)amino, N-(Ci-6 acyl)amino, N- (Ci-6alkyl)carbonylamino, phenyl, phenyl Ci-6 alkyl, carboxy, Ci-6alkoxycarbonyl, aminocarbonyl, N-(Ci-6alkyl)aminocarbonyl, or N,N- di(Ci- 6 alkyl)amino, -C(O)- R 20

wherein

R 20 represents Ci-6 alkyl, Ci-6 alkoxy, amino, N-(Ci-6alkyl)amino, N,N- di(Ci-6alkyl)amino, N-(Ci-6 acyl)amino, or a 5-7 membered saturated or unsaturated heterocyclic ring having 1 to 3 heteroatoms selected from the group consisting O, S and N, and optionally substituted by Ci-6 alkyl, Ci-6 alkoxy, oxo, amino, N-(Ci- 6alkyl)amino, N,N-di(Ci-6alkyl)amino, N-(Ci-6 acyl)amino, phenyl, or benzyl,

Ci-6 alkyl optionally substituted by R 21 ,

or

Ci-6 alkoxy optionally substituted by R 21 ,

wherein

R 21 represents cyano, mono-, di or tri- halogen, amino, N-(Ci- 6alkyl)amino, N,N-di(Ci-6alkyl)amino, N- (hydroxyCi-6 alkyl) amino, N- (halophenylCi-6 alkyl) amino, amino C2-6 alkylenyl, Ci-6 alkoxy, hydroxyCi-6 alkoxy, -C(O)- R 201 , -

NHC(O)- R 201 , C 3 - 8 cycloalkyl, isoindolino, phthalimidyl, 2- oxo-l,3-oxazolidinyl, aryl or a 5 or 6 membered saturated or unsaturated heterocyclic ring having 1 to 4 heteroatoms selected from the group consisting O, S and N , and optionally substituted by hydroxy, Ci-6 alkyl, Ci-

6 alkoxy, Ci-6 alkoxycarbonyl, hydroxyCi-6 alkoxy, oxo, amino, aminoCi-6alkyl, N-(Ci-6alkyl)amino, N,N-di(Ci-6alk- yl)amino, N-(Ci-6 acyl)amino, or benzyl, wherein R 201 represents hydroxy, amino, N-(Ci-6alkyl)amino, N,N- di(Ci-6alkyl)amino, N- (halophenylCi-6 alkyl) amino, Ci-6alkyl, aminoCi-6 alkyl, aminoC2-6 alkylenyl, Ci-6 alkoxy, a 5 or 6 membered saturated or unsaturated heterocyclic ring having 1 to 4 heteroatoms selected from the group consisting O, S and N, and optionally substituted by hydroxy, Ci-6 alkyl, Ci-6 alkoxy, Ci-6 alkoxycarbonyl, hydroxyCi-6 alkoxy, oxo, amino, N-(Ci-6alkyl)amino, N,N-di(Ci-6alkyl)amino, N-(Ci-6 acyl)amino or benzyl;

R 3 represents hydrogen, halogen, aminocarbonyl, or Ci-6 alkyl optionally substituted by aryl Ci-6 alkoxy or mono-, di- or tri- halogen;

R 4 represents hydrogen or Ci-6 alkyl;

R 5 represents hydrogen or Ci-6 alkyl; and R 6 represents halogen, hydrogen or Ci-6 alkyl,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

In an embodiment, said component A is a compound of general formula (A), supra, which is selected from the list consisting of :

N-(7,8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl )nicotinamide;

2-(7, 8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-l-pyri din-3-ylethylenol; N-(7, 8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH-ben zimidazole-5- carboxamide; 6-(acetamido)-N-(7,8-dimethoxy-2,3-dihydroimidazo[l,2-c]quin azolin-5- yl)nicotinamide;

N-{5-[2-(7,8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5 -yl)-l- hydroxyvinyl]pyridin-2-yl}acetamide;

2-({5-[2-hydroxy-2-pyridin-3-ylvinyl]-7-methoxy-2,3-dihyd roimidazo[l,2-c]quinazolin-8- yl}oxy)-N,N-dimethylacetamide;

2-[7-methoxy-8-(tetrahydro-2H-pyran-2-ylmethoxy)-2,3-dihydro innidazo[l,2- c]quinazolin-5-yl]-l-pyridin-3-ylethylenol;

2-[8-(2-hydroxyethoxy)-7-methoxy-2,3-dihydroimidazo[l,2-c]qu inazolin-5-yl]-l-pyridin- 3-ylethylenol;

({5-[2-hydroxy-2-pyridin-3-ylvinyl]-7-methoxy-2,3-dihydroimi dazo[l,2-c]quinazolin-8- yl}oxy)acetic acid;

4- ({5-[2-hydroxy-2-pyridin-3-ylvinyl]-7-methoxy-2,3-dihydroimi dazo[l,2-c]quinazolin-8- yl}oxy)butanoic acid;

({5-[2-hydroxy-2-pyridin-3-ylvinyl]-7-methoxy-2,3-dihydro imidazo[l,2-c]quinazolin-8- yl}oxy)acetonitrile;

2-[7-methoxy-8-(2H-tetrazol-5-ylmethoxy)-2,3-dihydroimidazo[ l,2-c]quinazolin-5-yl]-l- pyridin-3-ylethylenol;

2-[7-methoxy-8-(4-morpholin-4-yl-4-oxobutoxy)-2,3-dihydroimi dazo[l,2-c]quinazolin-5- yl]-l-pyridin-3-ylethylenol;

5- [l-hydroxy-2-(8-morpholin-4-yl-2,3-dihydroimidazo[l,2-c]quin azolin-5- yl)vinyl]pyridin-3-ol ;

N-(2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-5-hydroxynicoti namide;

6- (acetamido)-N-(7,9-dimethoxy-8-methyl-2,3-dihydroimidazo[l,2 -c]quinazolin-5- yl)nicotinamide;

N-(8,9-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-5 -hydroxynicotinamide; 5-hydroxy-N-(7-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5 -yl)nicotinamide;

N-(7,8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl )-5-[(4- methoxybenzyl)oxy]nicotinamide;

N-(7,8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl )-5-hydroxynicotinamide; 5- hydroxy-N-[8-(trifluoromethyl)-2,3-dihydroimidazo[l,2-c]quin azolin-5- yl]nicotinamide;

N-{8-[3-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)propoxy]-2,3 -dihydroimidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-(7-bromo-8-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin- 5-yl)nicotinamide;

6- amino-N-(8-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl) nicotinamide;

1- (lH-benzimidazol-5-yl)-2-(8,9-dimethoxy-2,3-dihydroimidazo[l ,2-c]quinazolin-5- yl)ethylenol;

2- (8,9-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-l-( 2,4-dimethyl-l,3-thiazol- 5-yl)ethylenol;

N-(9-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH-be nzimidazole-5- carboxamide;

N-(8-bromo-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotina nnide;

N-(8-bromo-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH-b enzimidazole-5- carboxamide;

N-(8-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH-be nzimidazole-5- carboxamide;

N-(8-methyl-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH-ben zimidazole-5- carboxamide;

N-[8-(trifluoromethyl)-2 -dihydroimidazo[l,2-c]quinazolin-5-yl]-lH-benzimidazole-5- carboxamide;

N-(7-fluoro-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH-ben zimidazole-5- carboxamide;

N-(7-methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicoti nannide;

N-(8-chloro-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-lH- benzimidazole-5- carboxamide;

6-(acetamido)-N-(8-morpholin-4-yl-2,3-dihydroinnidazo[l,2-c] quinazolin-5- yl)nicotinamide;

l-(lH-benzimidazol-5-yl)-2-(8-morpholin-4-yl-2,3-dihydroi midazo[l,2-c]quinazolin-5- yl)ethylenol; N 5-[l-hydroxy-2-(8-morpholin-4-yl-2,3-dihydroimidazo[l,2-c]qu inazolin-5- yl)vinyl]pyridin-2-yl}acetamide;

6-methyl-N-(8-morpholin-4-yl-2 -dihydroimidazo[l,2 ]quinazolin-5-yl)nicotinamide;

1- (lH-benzimidazol-5-yl)-2-[8-(4-methylpiperazin-l-yl)-2,3-dih ydroimidazo[l,2- c]quinazolin-5-yl]ethylenol;

N-(2 -dihydroimidazo[l,2-c]quinazolin-5-yl)-3H-imidazo[4,5-b]pyri dine-6-carboxamide; N-(7,8-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-3 H-imidazo[4,5-b]pyridine- 6-carboxamide;

N-[7-(trifluoromethyl)-2 -dihydroimidazo[l,2-c]quinazolin-5-yl]-lH-benzimidazole-5- carboxamide;

N-(7,9-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-l H-benzimidazole-5- carboxamide;

N-{5-[2-(7,9-dimethoxy-8-methyl-2,3-dihydroimidazo[l,2-c]qui nazolin-5-yl)-l- hydroxyvinyl]pyridin-2-yl}acetamide;

N-{5-[2-(7-bromo-9-nnethyl-2,3-dihydroinnidazo[l,2-c]quin azolin-5-yl)-l- hydroxyvinyl]pyridin-2-yl}acetamide; and

2- (8,9-dimethoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-l-p yridin-3-ylethylenol; In an embodiment, said component A is a compound having the formula (I) :

(I) or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, in which :

R 1 represents -(CH 2 )n-(CH R 4 )-(CH 2 )m-N(R 5 )(R 5' ) ;

R 2 represents a heteroaryl optionally substituted with 1, 2 or 3 R 6 groups ;

R 3 represents alkyl or cycloalkyi ;

R 4 represents hydrogen or alkoxy ; and

R 5 and R 5' may be the same or different and represent independently, hydrogen, alkyl, cycloalkylalklyl, or alkoxyalkyl or R 5 and R 5' may be taken together with the nitrogen atom to which they are bound to form a 3-7 membered nitrogen containing heterocyclic ring optionally containing at least one additional heteroatom selected from oxygen, nitrogen or sulfur and which may be optionally substituted with 1 or more R 6' groups, or R 4 and R 5 may be taken together with the atoms to which they are bound to form a 5-6 membered nitrogen containing heterocyclic ring optionally containing 1 or more nitrogen, oxygen or sulfur atoms and which may be optionally substituted with 1 or more R 6' groups ; each occurrence of R 6 may be the same or different and is independently halogen, alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkylalklyl, aryl, arylalkyi, heteroaryl, heteroarylalkyl, heterocyclic ring, heterocyclylalkyl, alkyl-OR 7 , alkyl-SR 7 , alkyl-N(R 7 )(R 7' ), alkyl-COR 7 ,-CN, -COOR 7 , -CON(R 7 )(R 7' ), -OR 7 , -SR 7 , -N(R 7 )(R 7' ), or -N R 7 COR 7 each of which may be optionally substituted with 1 or more R 8 groups ; each occurrence of R 6' may be the same or different and is independently alkyl, cycloalkylalklyl, or alkyl-OR 7 ; each occurrence of R 7 and R 7' may be the same or different and is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkylalklyl, cycloalkenyl, aryl, arylalkyi, heteroaryl, heterocyclic ring, heterocyclylalkyl, or heteroarylalkyl ; each occurrence of R 8 is independently nitro, hydroxy, cyano, formyl, acetyl, halogen, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkylalklyl, cycloalkenyl, aryl, arylalkyl, heteroaryl, heterocyclic ring, heterocyclylalkyl, or heteroarylalkyl ; n is an integer from 1-4 and m is an integer from 0-4 with the proviso that when R 4 and R 5 are taken together with the atoms to which they are bound to form a 5-6 membered nitrogen containing ring, n + m < 4 ;

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In an embodiment, said component A is a compound having the formula (I), supra, in which R 2 is a nitrogen containing heteroaryl optionally substituted with 1, 2 or 3 R 6 groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In an embodiment, said component A is a compound of general formula (I), supra, in which R 5 and R 5' are independently alkyl,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

In an embodiment, said component A is a compound of general formula (I), supra, in which R 5 and R 5' are taken together with the nitrogen atom to which they are bound to form a 5-6 membered nitrogen containing heterocyclic ring containing at least one additional heteroatom selected from oxygen, nitrogen or sulfur and which may be optionally substituted with 1 or more R 6' groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

In an embodiment, said component A is a compound of formula (I) in which R 4 and R 5 are taken together with the atoms to which they are bound to form a 5-6 membered nitrogen containing heterocyclic ring optionally containing 1 or more nitrogen, oxygen or sulfur atoms and which may be optionally substituted with 1 or more R 6 groups, or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In an embodiment, said component A is a compound of formula (I) in which R 3 is methyl,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In an embodiment, said component A is a compound of formula (I), wherein R 2 is pyridine, pyridazine, pyrimidine, pyrazine, pyrole, oxazole, thiazole, furan or thiophene, optionally substituted with 1, 2 or 3 R 6 groups; more preferably pyridine, pyridazine, pyrimidine, pyrazine, pyrole, oxazole or thiazole, optionally substituted with 1, 2 or 3 R 6 groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In an embodiment, said component A is a compound of formula (la) :

(la) or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein R 2 is as defined above for formula (I).

In an embodiment, said component A is a compound of formula (lb) :

(lb)

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein R 2 is as defined above for formula (I).

In an embodim

(Ic) or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein R 2 is as defined above for formula (I).

(Id) or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein R 2 and R 4 are as defined above for formula (I).

In an embodiment, said component A is a compound of the formula (le) :

(le) or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein R 2 and R 4 are as defined above for formula (I).

In an embodiment, said component A is a compound of formula (I) - (le), wherein R 2 is pyridine, pyridazine, pyrimidine, pyrazine, pyrole, oxazole, thiazole, furan or thiophene, optionally substituted with 1, 2 or 3 R 6 groups; more preferably wherein R 2 is pyridine, pyridazine, pyrimidine, pyrazine, pyrole, oxazole or thiazole, optionally substituted with 1, 2 or 3 R 6 groups,

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In an embodiment, said component A is a compound selected from the list consisting of :

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-metho xy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide ;

N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}-7-metho xy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)-2,4-dimethyl-l,3-t hiazole-5- carboxamide;

2-amino-N-[7-methoxy-8-(3-nnorpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]-l,3-thiazole-5-carboxa mide;

2-amino-N-[7-methoxy-8-(3-nnorpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]isonicotinamide;

2-amino-N-[7-methoxy-8-(3-nnorpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]-4-methyl-l,3-thiazole- 5-carboxamide; 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]-4-propylpyrimidine-5-carboxannide;

N-{8-[2-(4-ethylmorpholin-2-yl)ethoxy]-7-methoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl}nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-nnethoxy-2,3-dihydroinnidaz o[l,2- c]quinazolin-5-yl}pyrimidine-5-carboxamide;

N-(8-{3-[2-(hydroxymethyl)morpholin-4-yl]propoxy}-7-nnethoxy -

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-(8-{3-[2-(hydroxymethyl)morpholin-4-yl]propoxy}-7-nnethoxy -

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-{8-[3-(dimethylamino)propoxy]-7-nnethoxy-2,3-dihydroinnida zo[l,2- c]quinazolin-5-yl}nicotinamide 1-oxide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]-6-(2-pyrrolidin-l-ylethyl)nicotinamide;

6-(cyclopentylamino)-N-[7-nnethoxy-8-(3-nnorpholin-4-ylpr opoxy)- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[8-(2-hydroxy-3-morpholin-4-ylpropoxy)-7-methoxy-2,3-dihyd roimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-{7-methoxy-8-[3-(3-methylmorpholin-4-yl)propoxy]-2,3-dihyd roimidazo[l,2- c]quinazolin-5-yl}nicotinamide; N-(8-{3-[2-(hydroxymethyl)morpholin-4-yl]propoxy}-7-nnethoxy -2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-(8-{2-[4-(cyclobutylmethyl)nnorpholin-2-yl]ethoxy}-7-nneth oxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N-(7-methoxy-8-{2-[4-(2-methoxyethyl)nnorpholin-2-yl]ethoxy} -2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N 8-[(4-ethylmorpholin-2-yl)methoxy]-7-methoxy-2,3-dihydroimid azo[l,2- c]quinazolin-5-yl}nicotinamide;

N-(7-methoxy-8-{[4-(2-methoxyethyl)nnorpholin-2-yl]nnethoxy} -2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

N 7-methoxy-8-[(4-methylmorpholin-2-yl)methoxy]-2,3-dihydroimi dazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]pyrimidine-4-carboxamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]pyrimidine-4-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]-l-methyl-lH-innidazole-4-carboxannide;

rel-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}- 7-nnethoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)pyrimidine-5-carboxanni de;

rel-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]propoxy}- 7-nnethoxy-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl)-6-methylnicotinannide;

rel-6-acetamido-N-(8-{3-[(2R,6S)-2,6-dimethylnnorpholin-4 -yl]propoxy}-7- methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinanni de;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]-l-methyl-lH-innidazole-5-carboxannide;

6-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]-2-methylnicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]-4-methylpyrimidine-5-carboxannide; 6-amino-5-bromo-N-[7-nnethoxy-8-(3-nnorpholin-4-ylpropoxy)-2 ,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]-l,3-oxazole-5-carboxamide;

N-[7-methoxy-8-(morpholin-2-ylmethoxy)-2,3-dihydroimidazo[l, 2-c]quinazolin-

5- yl]nicotinamide;

2-{[2-(dimethylamino)ethyl]amino}-N-{8-[3-(dinnethylannino)p ropoxy]-7- methoxy-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl}pyrimidine- 5-carboxamide; 2-amino-N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydr oinnidazo[l,2- c]quinazolin-5-yl}-l,3-thiazole-5-carboxamide;

rel-2-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]p ropoxy}-7-nnethoxy-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)pyrimidine-5-car boxamide;

rel-6-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]p ropoxy}-7-nnethoxy-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

2-[(2-hydroxyethyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpr opoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-carboxamid e;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-[(3-methoxypropyl)amino]pyrimidine-5-ca rboxannide;

2-amino-N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dih ydroinnidazo[l,2- c]quinazolin-5-yl}pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-[(3-morpholin-4-ylpropyl)amino]pyrimidi ne-5-carboxannide;

2-[(2-methoxyethyl)amino]-N-[7-methoxy-8-(3-nnorpholin-4- ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-carboxamid e;

2-{[2-(dimethylamino)ethyl]amino}-N-[7-nnethoxy-8-(3-nnorpho lin-4-ylpropoxy)-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-car boxamide;

6- amino-N-{8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydroi nnidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-pyrrolidin-l-ylpyrimidine-5-carboxamide ; N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-(4-methylpiperazin-l-yl)pyrimidine-5-ca rboxannide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-morpholin-4-ylpyrimidine-5-carboxannide ;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimida zo[l,2- c]quinazolin-5-yl]-6-piperazin-l-ylnicotinamide hydrochloride;

6-[(3S)-3-aminopyrrolidin-l-yl]-N-[7-methoxy-8-(3-morpholin- 4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide hydrochloride hydrate;

6-[(3R)-3-aminopyrrolidin-l-yl]-N-[7-methoxy-8-(3-morphol in-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide hydrochloride;

6-[(4-fluorobenzyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpr opoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-[(2-furylmethyl)amino]-N-[7-methoxy-8-(3-morpholin-4-ylpro poxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-[(2-methoxyethyl)amino]-N-[7-methoxy-8-(3-morpholin-4-y lpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-(lH-pyrrol-l-yl)nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-morpholin-4-ylnicotinamide;

N-{7-methoxy-8-[3-(methylamino)propoxy]-2,3-dihydroimidazo[l ,2- c]quinazolin-5-yl}nicotinamide;

6-[(2,2-dimethylpropanoyl)amino]-N-[7-methoxy-8-(3-morpholin -4-ylpropoxy)-

2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

6-[(cyclopropylcarbonyl)amino]-N-[7-methoxy-8-(3-morpholi n-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-(2,2,2-trifluoroethoxy)nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-(trifluoromethyl)nicotinamide; 6-(isobutyrylamino)-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy) -2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-{7-methoxy-8-[3-(4-methylpiperazin-l-yl)propoxy]-2,3-dihyd roimidazo[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-{[(methylamino)carbonyl]amino}-l,3-thia zole-4- carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-{[(methylamino)carbonyl]amino}nicotinan nide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-(methylamino)-l,3-thiazole-4-carboxamid e;

N-[7-methoxy-8-(2-morpholin-4-ylethoxy)-2,3-dihydroimidazo[l ,2-c]quinazolin-

5- yl]nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl}-2,4-dimethyl-l,3-thiazole-5-carboxamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroinnid azo[l,2- c]quinazolin-5-yl}-6-methylnicotinamide;

6- {[(isopropylamino)carbonyl]amino}-N-[7-methoxy-8-(3-nnorphol in-4- ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimida zo[l,2- c]quinazolin-5-yl]-6-pyrrolidin-l-ylnicotinamide;

6-(dimethylamino)-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy )-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-piperidin-l-ylpropoxy)-2,3-dihydroimidazo[ l,2-c]quinazolin- 5-yl]nicotinamide;

N-[7-methoxy-8-(2-pyrrolidin-l-ylethoxy)-2,3-dihydroimidazo[ l,2-c]quinazolin-

5- yl]nicotinamide;

N-[7-methoxy-8-(2-piperidin-l-ylethoxy)-2,3-dihydroimidazo[l ,2-c]quinazolin-5- yl]nicotinamide;

6- {[(ethylamino)carbonyl]amino}-N-[7-methoxy-8-(3-morpholin-4- ylpropoxy)- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide; 6-fluoro-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydr oimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]-l,3-oxazole-4-carboxamide;

2-(ethylamino)-N-[7-methoxy-8-(3-nnorpholin-4-ylpropoxy)- 2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]-l,3-thiazole-4-carboxa mide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]pyrazine-2-carboxamide;

N-[8-(2-aminoethoxy)-7-methoxy-2,3-dihydroinnidazo[l,2-c]qui nazolin-5- yl]nicotinamide;

6-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]isonicotinamide;

N-{8-[3-(diethylamino)propoxy]-7-methoxy-2,3-dihydroinnid azo[l,2- c]quinazolin-5-yl}nicotinamide;

N-{8-[2-(diisopropylamino)ethoxy]-7-methoxy-2,3-dihydroinnid azo[l,2- c]quinazolin-5-yl}nicotinamide;

N 8-[2-(diethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2- c]quinazolin- 5-yl}nicotinamide;

N-{8-[3-(dimethylamino)propoxy]-7-nnethoxy-2,3-dihydroinnida zo[l,2- c]quinazolin-5-yl}nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-nnethoxy-2,3-dihydroinnidaz o[l,2- c]quinazolin-5-yl}nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnid azo[l,2- c]quinazolin-5-yl]-2-(methylamino)pyrinnidine-5-carboxannide ;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroinnidazo [l,2- c]quinazolin-5-yl]-2-(methylthio)pyrimidine-5-carboxannide;

N-[8-(3-aminopropoxy)-7-methoxy-2,3-dihydroinnidazo[l,2-c]qu inazolin-5- yl]nicotinamide trifluoroacetate; N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]thiophene-2-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2,4-dimethyl-l,3-thiazole-5-carboxamide;

2-methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihyd roimidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-3-furamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]thiophene-3-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2-methyl-l,3-thiazole-4-carboxamide;

6-methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihyd roimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

5-methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihyd roimidazo[l,2- c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-methylnicotinamide;

6-(acetylamino)-N-[7-methoxy-8-(3-nnorpholin-4-ylpropoxy)-2, 3- dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]nicotinamide;

or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In another embodiment, said component A is a compound selected from the list consisting of :

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimida zo[l,2- c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-6-methylnicotinamide; 5-methoxy-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihyd roimidazo[l,2 c]quinazolin-5-yl]nicotinamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]-2,4-dimethyl-l,3-thiazole-5-carboxamide;

N 8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[l,2 - c]quinazolin-5-yl}nicotinamide;

N 8-[3-(dimethylamino)propoxy]-7-methoxy-2,3-dihydroimidazo[l, 2- c]quinazolin-5-yl}nicotinamide;

6 [(isopropylamino)carbonyl]amino}-N-[7-methoxy-8-(3-morpholin -4- ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl] nicotinamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl}-2,4-dimethyl-l,3-thiazole-5-carboxamide;

N-[7-methoxy-8-(2-morpholin-4-ylethoxy)-2,3-dihydroimidazo[l ,2-c]quinazolin

5-yl]nicotinamide;

rel-6-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]prop oxy}-7-methoxy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)nicotinamide;

rel-2-amino-N-(8-{3-[(2R,6S)-2,6-dimethylmorpholin-4-yl]prop oxy}-7-methoxy- 2,3-dihydroimidazo[l,2-c]quinazolin-5-yl)pyrimidine-5-carbox amide;

2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro imidazo[l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

N-{8-[2-(dimethylamino)ethoxy]-7-methoxy-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl}pyrimidine-5-carboxamide;

N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydroimidazo[ l,2- c]quinazolin-5-yl]pyrimidine-5-carboxamide;

a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.

In another embodiment, said component A is 2-amino-N-[7-methoxy-8-(3-morpholin-4- ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidi ne-5-carboxamide, or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. In another embodiment, said component A is 2-amino-N-[7-methoxy-8-(3-morpholin-4- ylpropoxy)-2,3-dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidi ne-5-carboxamide dihydrochloride. Where there is a discrepancy between the chemical name and the chemical structure depicted, the chemical structure depicted takes precedence over the chemical name given.

Without being bound by theory or mechanism, the compounds of the present invention display surprising activity for the inhibition of phosphatidylinositol-3-kinase and chemical and structural stability over those compounds of the prior art. It is believed that this surprising activity is based on the chemical structure of the compounds, in particular the basicity of the compounds as a result of R 1 being amino optionally substituted with R 5 and R 5' . Further, the appropriate choice of R 3 and R 2 provide the necessary activity against the appropriate isoforms to allow for activity in vivo.

The synthesis of the compounds listed above is described in International Patent Application No. PCT/EP2003/010377, published as WO 2004/029055 Al, and in International Patent Application No. PCT/US2007/024985, published as WO 2008/070150, both of which are hereby incorporated herein in their entirety by reference.

Said component A may be in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially. The components may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.

The PI3K-inhibitors mentioned in the prior art as well as in the lists above have been disclosed for the treatment or prophylaxis of different diseases, especially cancer. The specific compounds of the lists as disclosed above are preferred as being component A of the combination, most preferred is the compound used in the experimental section. The synergistic behavior of a combination of the present invention is demonstrated herein with one of the PI3K inhibitors specifically disclosed in the Examples section as compound A.

In addition, a combination of the present invention comprises compound A as mentioned above and darolutamide or enzalutamide is a preferred aspect of the invention.

In another aspect, a combination of the present invention comprises compound A or a pharmaceutically acceptable salt thereof as mentioned above and darolutamide or enzalutamide.

It is to be understood that the present invention relates also to any combination of the embodiments of component A described above.

Component B of the combination of the present invention

Component B is an androgen receptor antagonist, such as darolutamide or enzalutamide for example.

Darolutamide (ODM-201) is a high-affinity androgen receptor antagonist with low nanomolar binding affinity for the androgen receptor and high inhibition of transactivation function (A.M. Moilanen et al., Sci. Rep., 2015, 5:12007). Darolutamide (ODM-201) was obtained from Orion Pharma, Turku, Finland. Enzalutamide (MDV-3100) is a high-affinity androgen receptor antagonist with nanomolar binding affinity for the androgen receptor and high inhibition of transactivation function (C. Tran et al., Science, 2009, 324:787-790). Enzalutamide (MDV-3100) was purchased from Selleckchem under catalog number S1250.

In accordance with an embodiment, the present invention relates to a combination of any component A mentioned herein with any component B mentioned herein, optionally with any component C mentioned herein.

In one embodiment component A of the combination is the compound used in the experimental section and Component B is an androgen receptor antagonist, such as darolutamide or enzalutamide for example being used in the experimental section.

In a particular embodiment, the present invention relates to a combination of a component A with a component B, optionally with a component C, as mentioned in the Examples Section herein.

Further, the present invention relates to a kit containing :

• a combination of :

o a component A : which is a PI3K kinase inhibitor, such as copanlisib, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a physiologically acceptable salt, or a mixture of same, for example ;

o a component B : which is an androgen receptor antagonist, such as darolutamide or enzalutamide for example ; and, optionally, o one or more pharmaceutical agents C ;

in which optionally either or both of said components A and B are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially. The term "component C" being at least one pharmaceutical agent includes the effective compound itself as well as its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers as well as any composition or pharmaceutical formulation comprising such effective compound or its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers. A list of such readily available agents is being provided further below.

The components may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.

Component A is administered intravenously, intraperitoneally, preferably it is administered orally.

Component B is administered intravenously, intraperitoneally, preferably it is administered orally.

Component C being administered as the case may be.

The term "pharmaceutically acceptable" is used synonymously to the term "physiologically acceptable".

The term "pharmaceutically or physiologically acceptable salt" of component A refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19. Pharmaceutically acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succinic acid and citric acid. Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an appropriate base to form, e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts. Those skilled in the art will further recognize that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.

Representative salts of a component A of this invention include the conventional nontoxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, sulfate, tartrate, thiocyanate, tosylate, and undecanoate.

Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, or butyl chlorides, bromides and iodides; dialkyi sulfates like dimethyl, diethyl, dibutyl sulfate, or diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.

A solvate for the purpose of this invention is a complex of a solvent and a compound of the invention in the solid state. Exemplary solvates would include, but are not limited to, complexes of a compound of the invention with ethanol or methanol. Hydrates are a specific form of solvate wherein the solvent is water. Compositions (formulations) of the present invention

Components of this invention can be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.

Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present.

Components of this invention can also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.

Oily suspensions can be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.

Syrups and elixirs can be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents.

Components of this invention can also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in preferably a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-l,l-dioxolane-4- methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as a soap or a detergent, suspending agent such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants. Illustrative of oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.

The parenteral compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile- lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above H LB or can be a mixture of two or more components having the desired HLB. Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The pharmaceutical compositions can be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate.

The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.

Components of the invention can also be administered in the form of suppositories for rectal administration of the drug. These components can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are, for example, cocoa butter and polyethylene glycol.

Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 11, 1991, incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.

It can be desirable or necessary to introduce a component of the present invention to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,011,472, issued April 30, 1991.

The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M. F. et al, "Compendium of Excipients for Parenteral Formulations" PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311; Strickley, R.G "Parenteral Formulations of Small Molecule Therapeutics Marketed in the U nited States (1999)-Part-1" PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349; and Nema, S. et al, "Excipients and Their Use in Injectable Products" PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171. Commonly used pharmaceutical ingredients that can be used as appropriate to formulate the composition for its intended route of administration include: acidifying agents (examples include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid); alkalinizing agents (examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine); adsorbents (examples include but are not limited to powdered cellulose and activated charcoal); aerosol propellants (examples include but are not limited to carbon dioxide, CCI2F2, F 2 CIC-CCIF 2 and CCIF 3 ) air displacement agents (examples include but are not limited to nitrogen and argon); antifungal preservatives (examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate); antimicrobial preservatives (examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal); antioxidants (examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium meta bisulfite); binding materials (examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene- butadiene copolymers); buffering agents (examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate) carrying agents (examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection) chelating agents (examples include but are not limited to edetate disodium and edetic acid) colorants (examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red); clarifying agents (examples include but are not limited to bentonite); emulsifying agents (examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate); encapsulating agents (examples include but are not limited to gelatin and cellulose acetate phthalate) flavorants (examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin); humectants (examples include but are not limited to glycerol, propylene glycol and sorbitol); levigating agents (examples include but are not limited to mineral oil and glycerin); oils (examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil); ointment bases (examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment); penetration enhancers (transdermal delivery) (examples include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas) plasticizers (examples include but are not limited to diethyl phthalate and glycerol); solvents (examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation); stiffening agents (examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax); suppository bases (examples include but are not limited to cocoa butter and polyethylene glycols (mixtures)); surfactants (examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate); suspending agents (examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum); sweetening agents (examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose); tablet anti-adherents (examples include but are not limited to magnesium stearate and talc); tablet binders (examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, non-crosslinked polyvinyl pyrrolidone, and pregelatinized starch); tablet and capsule diluents (examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch); tablet coating agents (examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac); tablet direct compression excipients (examples include but are not limited to dibasic calcium phosphate); tablet disintegrants (examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, cross- linked polyvinylpyrrolidone, sodium alginate, sodium starch glycollate and starch); tablet glidants (examples include but are not limited to colloidal silica, corn starch and talc); tablet lubricants (examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate); tablet/capsule opaquants (examples include but are not limited to titanium dioxide); tablet polishing agents (examples include but are not limited to carnuba wax and white wax); thickening agents (examples include but are not limited to beeswax, cetyl alcohol and paraffin); tonicity agents (examples include but are not limited to dextrose and sodium chloride); viscosity increasing agents (examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, polyvinyl pyrrolidone, sodium alginate and tragacanth); and wetting agents (examples include but are not limited to heptadecaethylene oxycetanol, lecithins, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).

Pharmaceutical compositions according to the present invention can be illustrated as follows:

Sterile IV Solution: A 5 mg/mL solution of the desired compound of this invention can be made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/mL with sterile 5% dextrose and is administered as an IV infusion over about 60 minutes. Lyophilized powder for IV administration: A sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lyophilized powder, (ii) 32- 327 mg/mL sodium citrate, and (iii) 300 - 3000 mg Dextran 40. The formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/mL, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/mL, and is administered either IV bolus or by IV infusion over 15 - 60 minutes.

Intramuscular suspension: The following solution or suspension can be prepared, for intramuscular injection:

50 mg/mL of the desired, water-insoluble compound of this invention

5 mg/mL sodium carboxymethylcellulose

4 mg/mL TWEEN 80

9 mg/mL sodium chloride

9 mg/mL benzyl alcohol

Hard Shell Capsules: A large number of unit capsules are prepared by filling standard two-piece hard galantine capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose and 6 mg of magnesium stearate.

Soft Gelatin Capsules: A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into molten gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules are washed and dried. The active ingredient can be dissolved in a mixture of polyethylene glycol, glycerin and sorbitol to prepare a water miscible medicine mix.

Tablets: A large number of tablets are prepared by conventional procedures so that the dosage unit is 100 mg of active ingredient, 0.2 mg. of colloidal silicon dioxide, 5 mg of magnesium stearate, 275 mg of microcrystalline cellulose, 11 mg. of starch, and 98.8 mg of lactose. Appropriate aqueous and non-aqueous coatings may be applied to increase palatability, improve elegance and stability or delay absorption.

Immediate Release Tablets/Capsules: These are solid oral dosage forms made by conventional and novel processes. These units are taken orally without water for immediate dissolution and delivery of the medication. The active ingredient is mixed in a liquid containing ingredient such as sugar, gelatin, pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze drying and solid state extraction techniques. The drug compounds may be compressed with viscoelastic and thermoelastic sugars and polymers or effervescent components to produce porous matrices intended for immediate release, without the need of water.

Commercial utility of the present invention Component A

The compounds of formula (A) and (I) and the stereoisomers thereof according to the combination as referred to above are components A. The compounds according to the combination have valuable pharmaceutical properties, which make them commercially utilizable. In particular, they inhibit the PI3K/AKT pathway and exhibit cellular activity. They are expected to be commercially applicable in the therapy of diseases (e.g. diseases dependent on overactivated PI3K/AKT). An abnormal activation of the PI3K/AKT pathway is an essential step towards the initiation and maintenance of human tumors and thus its inhibition, for example with PI3K inhibitors, is understood to be a valid approach for treatment of human tumors. For a recent review see Garcia- Echeverria et al (Oncogene, 2008, 27, 551-5526.

Component B Due to the mechanism as discussed in the introductory section component B is especially suitable to have effects on tumor diseases, especially those developing resistance mechanism via antiapoptotic pathways or cell cycle activation. Combination

The combinations of the present invention thus can be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, such as, for example, haematological tumours and/or metastases thereof, solid tumours, and/or metastases thereof, e.g. leukaemias, multiple myeloma thereof and myelodysplastic syndrome, malignant lymphomas, breast tumours including and bone metastases thereof, tumours of the thorax including non-small cell and small cell lung tumours and bone metastases thereof, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours and bone metastases thereof, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.

One embodiment relates to the use of a combination as defined herein for the preparation of a medicament for the treatment or prophylaxis of a cancer, in particular prostate cancer.

One embodiment relates to the use of a combination as defined herein in the treatment or prophylaxis of a cancer, in particular prostate cancer.

In one embodiment the invention relates to combinations comprising component A or a pharmaceutically acceptable salt thereof and component B being intravenously, intraperitoneal^, preferably it is administered orally. The term "inappropriate" within the context of the present invention, in particular in the context of "inappropriate cellular immune responses, or inappropriate cellular inflammatory responses", as used herein, is to be understood as preferably meaning a response which is less than, or greater than normal, and which is associated with, responsible for, or results in, the pathology of said diseases.

Combinations of the present invention might be utilized to inhibit, block, reduce, decrease, etc., cell proliferation and/or cell division, and/or produce apoptosis.

This invention includes a method comprising administering to a mammal in need thereof, including a human, an amount of a component A and an amount of component B of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof ; etc. which is effective to treat the disorder.

Hyper-proliferative disorders include but are not limited, e.g., psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), as well as malignant neoplasia. Examples of malignant neoplasia treatable with the compounds according to the present invention include solid and hematological tumors. Solid tumors can be exemplified by tumors of the breast, bladder, bone, brain, central and peripheral nervous system, colon, anum, endocrine glands (e.g. thyroid and adrenal cortex), esophagus, endometrium, germ cells, head and neck, kidney, liver, lung, larynx and hypopharynx, mesothelioma, ovary, pancreas, prostate, rectum, renal, small intestine, soft tissue, testis, stomach, skin, ureter, vagina and vulva. Malignant neoplasias include inherited cancers exemplified by Retinoblastoma and Wilms tumor. In addition, malignant neoplasias include primary tumors in said organs and corresponding secondary tumors in distant organs ("tumor metastases"). Hematological tumors can be exemplified by aggressive and indolent forms of leukemia and lymphoma, namely non-Hodgkins disease, chronic and acute myeloid leukemia (CML / AML), acute lymphoblastic leukemia (ALL), Hodgkins disease, multiple myeloma and T-cell lymphoma. Also included are myelodysplastic syndrome, plasma cell neoplasia, paraneoplastic syndromes, and cancers of unknown primary site as well as AIDS related malignancies.

Examples of breast cancer include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ, particularly with bone metastases.

Examples of cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.

Examples of brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.

Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.

Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.

Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.

Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma.

Examples of liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.

Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer. Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell. Lymphomas include, but are not limited to AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.

Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.

Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.

These disorders have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.

The term "treating" or "treatment" as stated throughout this document is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc., of a disease or disorder, such as a carcinoma.

Combinations of the present invention might also be used for treating disorders and diseases associated with excessive and/or abnormal angiogenesis.

Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism. A number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, ischemic retinal- vein occlusion, and retinopathy of prematurity [Aiello et al. New Engl. J. Med. 1994, 331, 1480 ; Peer et al. Lab. Invest. 1995, 72, 638], age-related macular degeneration [AMD ; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855], neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc. In addition, the increased blood supply associated with cancerous and neoplastic tissue, encourages growth, leading to rapid tumor enlargement and metastasis. Moreover, the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer. Thus, combinations of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis disorders, e.g., by inhibiting and/or reducing blood vessel formation ; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apoptosis of such cell types.

Dose and administration of the present invention

Component A and component B

Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of hyper-proliferative disorders and angiogenic disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredients to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular component And dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.

The total amount of the active ingredients to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day. Clinically useful dosing schedules of a compound will range from one to three times a day dosing to once every four weeks dosing. In addition, "drug holidays" in which a patient is not dosed with a drug for a certain period of time, may be beneficial to the overall balance between pharmacological effect and tolerability. A unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg. The average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.

Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compounds employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.

Combinations of the present invention

The combinations of the present invention can be used in particular in therapy and prevention, i.e. prophylaxis, of tumour growth and metastases, including solid and haematological tumours of all indications and stages with or without pre-treatment of the tumour growth.

Methods of testing for a particular pharmacological or pharmaceutical property are well known to persons skilled in the art. The combinations of component A and component B of this invention can be administered as the sole pharmaceutical agent or in combination with one or more further pharmaceutical agents C where the resulting combination of components A, B and C causes no unacceptable adverse effects. For example, the combinations of components A and B of this invention can be combined with component C, i.e. one or more further pharmaceutical agents, such as known anti-angiogenesis, anti-hyper- proliferative, antiinflammatory, analgesic, immunoregulatory, diuretic, antiarrhytmic, anti-hypercholsterolemia, anti-dyslipidemia, anti-diabetic or antiviral agents, and the like, as well as with admixtures and combinations thereof.

Component C, can be one or more pharmaceutical agents such as 1311-chTNT, abarelix, abiraterone, aclarubicin, adalimumab, ado-trastuzumab emtansine, afatinib, aflibercept, aldesleukin, alectinib, alemtuzumab, alendronic acid, alitretinoin, altretamine, amifostine, aminoglutethimide, hexyl aminolevulinate, amrubicin, amsacrine, anastrozole, ancestim, anethole dithiolethione, anetumab ravtansine, angiotensin II, antithrombin III, aprepitant, arcitumomab, arglabin, arsenic trioxide, asparaginase, atezolizumab, axitinib, azacitidine, basiliximab, belotecan, bendamustine, besilesomab, belinostat, bevacizumab, bexarotene, bicalutamide, bisantrene, bleomycin, blinatumomab, bortezomib, buserelin, bosutinib, brentuximab vedotin, busulfan, cabazitaxel, cabozantinib, calcitonine, calcium folinate, calcium levofolinate, capecitabine, capromab, carbamazepine carboplatin, carboquone, carfilzomib, carmofur, carmustine, catumaxomab, celecoxib, celmoleukin, ceritinib, cetuximab, chlorambucil, chlormadinone, chlormethine, cidofovir, cinacalcet, cisplatin, cladribine, clodronic acid, clofarabine, cobimetinib, crisantaspase, crizotinib, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daratumumab, darbepoetin alfa, dabrafenib, dasatinib, daunorubicin, decitabine, degarelix, denileukin diftitox, denosumab, depreotide, deslorelin, dianhydrogalactitol, dexrazoxane, dibrospidium chloride, dianhydrogalactitol, diclofenac, dinutuximab, docetaxel, dolasetron, doxifluridine, doxorubicin, doxorubicin + estrone, dronabinol, eculizumab, edrecolomab, elliptinium acetate, elotuzumab, eltrombopag, endostatin, enocitabine, enzalutamide, epirubicin, epitiostanol, epoetin alfa, epoetin beta, epoetin zeta, eptaplatin, eribulin, erlotinib, esomeprazole, estradiol, estramustine, ethinylestradiol, etoposide, everolimus, exemestane, fadrozole, fentanyl, filgrastim, fluoxymesterone, floxuridine, fludarabine, fluorouracil, flutamide, folinic acid, formestane, fosaprepitant, fotemustine, fulvestrant, gadobutrol, gadoteridol, gadoteric acid meglumine, gadoversetamide, gadoxetic acid, gallium nitrate, ganirelix, gefitinib, gemcitabine, gemtuzumab, Glucarpidase, glutoxim, GM-CSF, goserelin, granisetron, granulocyte colony stimulating factor, histamine dihydrochloride, histrelin, hydroxycarbamide, 1-125 seeds, lansoprazole, ibandronic acid, ibritumomab tiuxetan, ibrutinib, idarubicin, ifosfamide, imatinib, imiquimod, improsulfan, indisetron, incadronic acid, ingenol mebutate, interferon alfa, interferon beta, interferon gamma, iobitridol, iobenguane (1231), iomeprol, ipilimumab, irinotecan, Itraconazole, ixabepilone, ixazomib, lanreotide, lansoprazole, lapatinib, lasocholine, lenalidomide, lenvatinib, lenograstim, lentinan, letrozole, leuprorelin, levamisole, levonorgestrel, levothyroxine sodium, lisuride, lobaplatin, lomustine, lonidamine, masoprocol, medroxyprogesterone, megestrol, melarsoprol, melphalan, mepitiostane, mercaptopurine, mesna, methadone, methotrexate, methoxsalen, methylaminolevulinate, methylprednisolone, methyltestosterone, metirosine, mifamurtide, miltefosine, miriplatin, mitobronitol, mitoguazone, mitolactol, mitomycin, mitotane, mitoxantrone, mogamulizumab, molgramostim, mopidamol, morphine hydrochloride, morphine sulfate, nabilone, nabiximols, nafarelin, naloxone + pentazocine, naltrexone, nartograstim, necitumumab, nedaplatin, nelarabine, neridronic acid, netupitant/palonosetron, nivolumab, pentetreotide, nilotinib, nilutamide, nimorazole, nimotuzumab, nimustine, nintedanib, nitracrine, nivolumab, obinutuzumab, octreotide, ofatumumab, olaparib, olaratumab, omacetaxine mepesuccinate, omeprazole, ondansetron, oprelvekin, orgotein, orilotimod, osimertinib, oxaliplatin, oxycodone, oxymetholone, ozogamicine, p53 gene therapy, paclitaxel, palbociclib, palifermin, palladium-103 seed, palonosetron, pamidronic acid, panitumumab, panobinostat, pantoprazole, pazopanib, pegaspargase, PEG-epoetin beta (methoxy PEG-epoetin beta), pembrolizumab, pegfilgrastim, peginterferon alfa- 2b, pembrolizumab, pemetrexed, pentazocine, pentostatin, peplomycin, Perflubutane, perfosfamide, Pertuzumab, picibanil, pilocarpine, pirarubicin, pixantrone, plerixafor, plicamycin, poliglusam, polyestradiol phosphate, polyvinylpyrrolidone + sodium hyaluronate, polysaccharide-K, pomalidomide, ponatinib, porfimer sodium, pralatrexate, prednimustine, prednisone, procarbazine, procodazole, propranolol, quinagolide, rabeprazole, racotumomab, radium-223 chloride, radotinib, raloxifene, raltitrexed, ramosetron, ramucirumab, ranimustine, rasburicase, razoxane, refametinib , regorafenib, risedronic acid, rhenium-186 etidronate, rituximab, rolapitant, romidepsin, romiplostim, romurtide, roniciclib , rucaparib, samarium (153Sm) lexidronam, sargramostim, satumomab, secretin, siltuximab, sipuleucel-T, sizofiran, sobuzoxane, sodium glycididazole, sonidegib, sorafenib, stanozolol, streptozocin, sunitinib, talaporfin, talimogene laherparepvec, tamibarotene, tamoxifen, tapentadol, tasonermin, teceleukin, technetium (99mTc) nofetumomab merpentan, 99mTc-HYNIC- [Tyr3]-octreotide, tegafur, tegafur + gimeracil + oteracil, temoporfin, temozolomide, temsirolimus, teniposide, testosterone, tetrofosmin, thalidomide, thiotepa, thymalfasin, thyrotropin alfa, tioguanine, tocilizumab, topotecan, toremifene, tositumomab, trabectedin, trametinib, tramadol, trastuzumab, trastuzumab emtansine, treosulfan, tretinoin, trifluridine + tipiracil, trilostane, triptorelin, trametinib, trofosfamide, thrombopoietin, tryptophan, ubenimex, valatinib , valrubicin, vandetanib, vapreotide, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vismodegib, vorinostat, vorozole, yttrium-90 glass microspheres, zinostatin, zinostatin stimalamer, zoledronic acid, zorubicin, or combinations thereof.

Alternatively, said component C can be one or more further pharmaceutical agents selected from gemcitabine, paclitaxel, cisplatin, carboplatin, sodium butyrate, 5-FU, doxirubicin, tamoxifen, etoposide, trastumazab, gefitinib, intron A, rapamycin, 17-AAG, U0126, insulin, an insulin derivative, a PPAR ligand, a sulfonylurea drug, an ct- glucosidase inhibitor, a biguanide, a PTP-IB inhibitor, a DPP-IV inhibitor, a 11-beta-HSD inhibitor, GLP-1, a GLP-1 derivative, GI P, a GIP derivative, PACAP, a PACAP derivative, secretin or a secretin derivative.

Optional anti-hyper-proliferative agents which can be added as component C to the combination of components A and B of the present invention include but are not limited to compounds listed on the cancer chemotherapy drug regimens in the 11 th Edition of the Merck Index, (1996), which is hereby incorporated by reference, such as asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6- mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifen, streptozocin, tamoxifen, thioguanine, topotecan, vinblastine, vincristine, and vindesine.

Other anti-hyper-proliferative agents suitable for use as component C with the combination of components A and B of the present invention include but are not limited to those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., publ. by McGraw-Hill, pages 1225-1287, (1996), which is hereby incorporated by reference, such as aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2',2'- difluorodeoxycytidine, docetaxel, erythrohydroxynonyl adenine, ethinyl estradiol, 5- fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel (when component B is not itself paclitaxel), pentostatin, N-phosphonoacetyl-L- aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine, uridine, and vinorelbine.

Other anti-hyper-proliferative agents suitable for use as component C with the combination of components A and B of the present invention include but are not limited to other anti-cancer agents such as epothilone and its derivatives, irinotecan, raloxifen and topotecan. Generally, the use of cytotoxic and/or cytostatic agents as component C in combination with a combination of components A and B of the present invention will serve to:

(1) yield better efficacy in reducing the growth of a tumor and/or metastasis or even eliminate the tumor and/ or metastasis as compared to administration of either agent alone,

(2) provide for the administration of lesser amounts of the administered chemo- therapeutic agents,

(3) provide for a chemotherapeutic treatment that is well tolerated in the patient with fewer deleterious pharmacological complications than observed with single agent chemotherapies and certain other combined therapies,

(4) provide for treating a broader spectrum of different cancer types in mammals, especially humans,

(5) provide for a higher response rate among treated patients,

(6) provide for a longer survival time among treated patients compared to standard chemotherapy treatments,

(8) provide a longer time for tumor progression, and/or

(9) yield efficacy and tolerability results at least as good as those of the agents used alone, compared to known instances where other cancer agent combinations produce antagonistic effects. EXPERIMENTAL SECTION

Examples demonstrating the synergistic effect of the combinations of components A and B of the present invention

1. Compounds used :

• Component A :

In this Experimental section and in the Figures, the term "compound A" is :

• "copanlisib", which is 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3- dihydroimidazo[l,2-c]quinazolin-5-yl]pyrimidine-5-carboxamid e

dihydrochloride, of structure :

compound A or a solvate, hydrate or stereoisomer thereof.

The synthesis of compound A is described in European patent application number EP 11 161 111.7, and in PCT application number PCT/EP2012/055600 published under WO 2012/136553, both of which are hereby incorporated herein in their entirety by reference. Copanlisib is a pan-class I PI3K inhibitor with subnanomolar activity against PI3K0C and ΡΙ3Κδ (N. Liu et al., Mol. Cancer Ther., 2013, 12:2319-2330). or

• "apitolisib" (GDC-0980), which is a class I PI3K/mTOR kinase inhibitor with low nanomolar activity against PI3K isoforms and mTOR (D.P. Sutherlin et al., J. Med. Chem., 2011, 54:7579-7587). Apitolisib was purchased from Selleckchem. or

• "dactolisib" (NVP-BEZ2359, which is a class I PI3K/mTOR kinase inhibitor with low nanomolar activity against PI3K isoforms and mTOR (S.M. Maira et al., Mol. Cancer Ther., 2008, 7:1851-1863). Dactolisib was purchased from Selleckchem.

• Component B :

In this Experimental Section and in the Figures, the term "compound B" refers to : · "darolutamide" (ODM-201), which is a high-affinity androgen receptor antagonist with low nanomolar binding affinity for the androgen receptor and high inhibition of transactivation function (A.M. Moilanen et al., Sci. Rep., 2015, 5:12007). Darolutamide (ODM-201) is exemplified together with its synthesis in WO2011/051540 as Example 56 (page 66).

• "enzalutamide" (MDV-3100) is a high-affinity androgen receptor antagonist with nanomolar binding affinity for the androgen receptor and high inhibition of transactivation function (C. Tran et al., Science, 2009, 324:787-790). 2. Assay description :

Cell lines derived from human prostate cancer metastasis were used (Table 1). Table 1:

For the determination of antiproliferative activity of single agents, the following cell culture conditions were used:

LAPC-4 human prostate cancer cells were plated in 30 μΙ medium (Roswell Park Memorial Institute 1640, 10% charcoal-stripped fetal bovine serum) at 1,200 cells/well in a 384-well microtiter plate. After one day, the cells were treated with R1881 (10 nM) and 1 nM to 10 μΜ compounds (day 0). Cell number was determined following addition of CellTiter-Glo ¾ (Promega) at day 0 and day 6. Luminescence was determined in a Victor X3 device (PerkinElmer). The inhibition of cell growth was calculated by normalization with respect to the luminescence reading (cell number) measured at the end of the experiment for cells treated with R1881 alone compared to the fluorescence reading (cell number) measured at the end of the experiment for DMSO-treated cells. VCaP human prostate cancer cells were plated in 30 μΙ medium (Roswell Park Memorial Institute 1640, 10% charcoal-stripped fetal bovine serum) at 2,400 cells/well in a 384- well microtiter plate. After one day, the cells were treated with R1881 (0.1 nM) and 1 nM to 10 μΜ compounds (day 0). Cell number was determined following addition of CellTiter-Glo ¾ (Promega) at day 0 and day 6. Luminescence was determined in a Victor X3 device (Promega). The inhibition of cell growth was calculated by normalization with respect to the luminescence reading (cell number) measured at the end of the experiment for cells treated with R1881 alone compared to the fluorescence reading (cell number) measured at the start of the experiment for DMSO-treated cells. For the determination of the combination index, the cells were treated with different ratios of compound A (PI3K inhibitor) and compound B (androgen receptor antagonist), typically between 1/100 to 10/1. The Combination Index (CI) was determined for the two compound ratios closest to the ratio of the respective IC50 values of each compound alone. It was calculated according to Chou and Talalay (Adv. Enzyme Regul., 1984, 22:27-55).

Results :

Table 2 shows the results of the LAPC-4 proliferation assays following treatment with a PI3K inhibitor or an AR antagonist, and the corresponding combination index.

Table 2:

Table 3 shows the results of the VCaP proliferation assays following treatment with PI3K inhibitor or an AR antagonist, and the corresponding combination index. Table 3:

The results show that the combined treatment of a PI3K inhibitor and an androgen receptor antagonist is moderately synergistic in LAPC-4 cells and slightly synergistic in VCaP cells.

In summary, our data indicate synergistic effects of the PI3K inhibitor copanlisib and darolutamide or enzalutamide in inhibiting tumor cell proliferation and warrant further clinical evaluation of this promising combination therapy for the treatment of cancer, in particular prostate cancer.