Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
2-AZA- AND 2-OXABICYCLO[2.1.1]HEXANE DERIVATIVES AS FACTOR XIIA ENZYME INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2024/038282
Kind Code:
A1
Abstract:
The present invention provides compounds of formula (I): Formula (I) compositions comprising such compounds; the use of such compounds in medicine; and methods of treating patients with such compounds; wherein A, W, V, Z, Y and B are as defined herein.

Inventors:
DAVIE REBECCA LOUISE (GB)
EDWARDS HANNAH JOY (GB)
EVANS DAVID MICHAEL (GB)
FROMONT CHRISTOPHE (GB)
GREVES WILLIAM JACK (GB)
NORTH CARL LESLIE (GB)
CAMP NICHOLAS PAUL (GB)
BARTHOLOMEUS JOHAN (CA)
JOHNSTONE SHAWN DONALD (CA)
KEITA MASSABA (CA)
POMEROLE JULIEN (CA)
Application Number:
PCT/GB2023/052166
Publication Date:
February 22, 2024
Filing Date:
August 18, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
KALVISTA PHARMACEUTICALS LTD (GB)
International Classes:
C07D401/14; A61K31/4427; A61K31/444; A61K31/4725; A61P7/00; A61P37/00; C07D405/14; C07D413/14; C07D471/04; C07D491/107
Domestic Patent References:
WO2022118016A22022-06-09
WO2022175675A12022-08-25
WO2019113642A12019-06-20
WO2018093695A12018-05-24
WO2018093716A12018-05-24
WO2017123518A12017-07-20
WO2017205296A12017-11-30
WO2019108565A12019-06-06
WO2012120128A12012-09-13
WO2019211585A12019-11-07
WO2019186164A12019-10-03
Foreign References:
EP2281885A12011-02-09
Other References:
DATABASE Registry [online] 22 April 2021 (2021-04-22), ENAMINE LLC: "1H-Pyrrole-2-carboxamide, N-[1-[[(6-methoxy-4-pyrimidinyl)amino]methyl]-3-methyl-2-oxabicyclo[2.1.1]hex-4-yl]-1-methyl-", XP093084075, Database accession no. 2636674-43-4
INVANOV, BLOOD, vol. 129, no. 11, 16 March 2017 (2017-03-16), pages 1527 - 1537
M. PATHAK ET AL.: "Crystal structures of the recombinant β-factor XIIa protease with bound Thr-Arg and Pro-Arg substrate mimetics", ACTA. CRYST, vol. D75, 2019, pages 1 - 14
A DEMENTIEV ET AL.: "Structures of human plasma β factor Xlla cocrystallized with potent inhibitors", BLOOD ADVANCES, vol. 2, no. 5, 2018, pages 549 - 558, XP055901041, DOI: 10.1182/bloodadvances.2018016337
B. K. HAMAD: "Journal< of Thrombois and Haemostasis", OF THROMBOSIS AND HAEMOSTASIS, vol. 15, pages 1818 - 1828
GOBEL ET AL., PROC NATL ACAD SCI USA., vol. 116, no. 1, 2 January 2019 (2019-01-02), pages 271 - 276
ABDALLAH ET AL., J BIOL CHEM., vol. 285, no. 45, 5 November 2010 (2010-11-05), pages 35206 - 15
KONINGS ET AL., THROMB RES, vol. 136, no. 2, August 2015 (2015-08-01), pages 474 - 80
ABID ET AL., J BIOL CHEM., vol. 284, no. 37, 11 September 2009 (2009-09-11), pages 24715 - 24
KAPLAN ET AL., ADV IMMUNOL, vol. 121, 2014, pages 41 - 89
HOPP ET AL., J NEUROINFLAMMATION, vol. 14, no. 1, 20 February 2017 (2017-02-20), pages 39
CRAIG ET AL., THE LANCET, vol. 401, no. 10382, 2023, pages 1079 - 1090
BJBRKQVIST ET AL., J CLIN INVEST, vol. 125, no. 8, 3 August 2015 (2015-08-03), pages 3132 - 46
DE MAAT ET AL., J ALLERGY CLIN IMMUNOL, vol. 138, no. 5, November 2016 (2016-11-01), pages 1414 - 1423
VERONEZ ET AL., FRONT MED (LAUSANNE, vol. 6, 21 February 2019 (2019-02-21), pages 28
RECKE ET AL., CLIN TRANSL ALLERGY, vol. 9, 14 February 2019 (2019-02-14), pages 9
MANSI ET AL., 2014 THE ASSOCIATION FOR THE PUBLICATION OF THE JOURNAL OF INTERNAL MEDICINE JOURNAL OF INTERNAL MEDICINE, vol. 277, 2015, pages 585 - 593
MAAT ET AL., J THROMB HAEMOST, vol. 17, no. 1, January 2019 (2019-01-01), pages 183 - 194
KEDARISETTY ET AL., OTOLARYNGOL HEAD NECK SURG, 30 April 2019 (2019-04-30), pages 194599819846446
LONG ET AL., NANOTOXICOLOGY, vol. 10, no. 4, 2016, pages 501 - 11
BAS ET AL., N ENGL J MED, 2015
LEIBFRIEDKOVARY, J PHARM PRACT, 2017
VAN DEN ELZEN ET AL., CLINIC REV ALLERG IMMUNOL, 2018
HAN ET AL., JCI, 2002
SIMAO ET AL., BLOOD, vol. 129, no. 16, 20 April 2017 (2017-04-20), pages 2280 - 2290
FROHLICH ET AL., STROKE, 11 June 2019 (2019-06-11), pages STROKEAHA119025260
RATHBUN, OXF MED CASE REPORTS, vol. 2019, no. 1, 24 January 2019 (2019-01-24), pages omy112
LEKOUBOU ET AL., NEUROL RES, vol. 36, no. 7, July 2014 (2014-07-01), pages 687 - 94
HILL ET AL., NEUROLOGY, vol. 60, no. 9, 13 May 2003 (2003-05-13), pages 1525 - 7
STONE ET AL., IMMUNOL ALLERGY CLIN NORTH AM, vol. 37, no. 3, August 2017 (2017-08-01), pages 483 - 495
SCOTT ET AL., CURR DIABETES REV, vol. 14, no. 4, 2018, pages 327 - 333
HERMANRUD ET AL., BMJ CASE REP, vol. 2017, 10 January 2017 (2017-01-10), pages bcr2016217802
KIM ET AL., BASIC CLIN PHARMACOL TOXICOL, vol. 124, no. 1, January 2019 (2019-01-01), pages 115 - 122
REICHMAN ET AL., PHARMACOEPIDEMIOL DRUG SAF, vol. 26, no. 10, October 2017 (2017-10-01), pages 1190 - 1196
DIESTRO ET AL., J STROKE CEREBROVASC DIS, vol. 28, no. 5, May 2019 (2019-05-01), pages e44 - e45
GIARD ET AL., DERMATOLOGY, vol. 225, no. 1, 2012, pages 62 - 9
LIU ET AL., BIOL CHEM, vol. 394, no. 3, March 2013 (2013-03-01), pages 319 - 28
GAO ET AL., NAT MED., vol. 13, no. 2, 28 January 2007 (2007-01-28), pages 181 - 8
GAO ET AL., PROTEOME RES, vol. 7, no. 6, June 2008 (2008-06-01), pages 2516 - 25
KITA ET AL., DIABETES, vol. 64, no. 10, October 2015 (2015-10-01), pages 3588 - 99
CLERMONT ET AL., INVEST OPHTHALMOL VIS SCI, vol. 57, no. 6, 1 May 2016 (2016-05-01), pages 2390 - 9
CLERMONT ET AL., ARVO TALK, 2019
MORRISON ET AL., J EXP MED., vol. 140, no. 3, 1 September 1974 (1974-09-01), pages 797 - 811
ZAMOLODCHIKOV ET AL., PROC NATL ACAD SCI USA., vol. 112, no. 13, 31 March 2015 (2015-03-31), pages 4068 - 73
SIMOES ET AL., J NEUROCHEM, vol. 150, no. 3, August 2019 (2019-08-01), pages 296 - 311
GOBEL ET AL., NAT COMMUN., vol. 7, 18 May 2016 (2016-05-18), pages 11626
BENDER ET AL., FRONT IMMUNOL, vol. 8, 15 September 2017 (2017-09-15), pages 1115
SALA-CUNILL ET AL., J ALLERGY CLIN IMMUNOL, vol. 135, no. 4, April 2015 (2015-04-01), pages 1031 - 43
DAVIE ET AL., SCIENCE, vol. 145, no. 3638, 18 September 1964 (1964-09-18), pages 1310 - 2
RADCLIFFE ET AL., BLOOD, vol. 50, no. 4, October 1977 (1977-10-01), pages 611 - 7
PUY ET AL., J THROMB HAEMOST, vol. 11, no. 7, July 2013 (2013-07-01), pages 1341 - 52
RENNE ET AL., J EXP MED., vol. 202, no. 2, 18 July 2005 (2005-07-18), pages 271 - 81
SIMAO ET AL., FRONT MED (LAUSANNE)., vol. 4, 31 July 2017 (2017-07-31), pages 121
WORM ET AL., ANN TRANSL MED, vol. 3, no. 17, October 2015 (2015-10-01), pages 247
BARBIERI ET AL., J PHARMACOL EXP THER, vol. 360, no. 3, March 2017 (2017-03-01), pages 466 - 475
KRUPKA ET AL., PLOS ONE, vol. 11, no. 1, 27 January 2016 (2016-01-27), pages e0146783
LEUNG ET AL., TRANSL STROKE RES, vol. 3, no. 3, September 2012 (2012-09-01), pages 381 - 9
LIU ET AL., NAT MED., vol. 17, no. 2, February 2011 (2011-02-01), pages 206 - 10
DIDIASOVA ET AL., CELL SIGNAL., vol. 51, November 2018 (2018-11-01), pages 257 - 265
GHEBREHIWET ET AL., IMMUNOL REV, vol. 274, no. 1, November 2016 (2016-11-01), pages 281 - 289
MURUGESAN ET AL., EXP EYE RES, vol. 186, 24 July 2019 (2019-07-24), pages 107744
IRMSCHER ET AL., J INNATE IMMUN, vol. 10, no. 2, 2018, pages 94 - 105
GHEBREHIWET ET AL., J EXP MED., vol. 153, no. 3, 1 March 1981 (1981-03-01), pages 665 - 76
SHATZEL ET AL., RES PRACT THROMB HAEMOST, vol. 4, no. 4, 15 May 2020 (2020-05-15), pages 500 - 505
WYGRECKA ET AL.: "Coagulation factor XII regulates inflammatory responses in human lungs", EUROPEAN RESPIRATORY JOURNAL, vol. 50, 2017, pages PA339
WONG ET AL.: "CSL312, a Novel Anti-FXII Antibody, Blocks FXII-Induced IL-6 Production from Primary Non-Diseased and Idiopathic Pulmonary Fibrosis Fibroblasts", AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE, vol. 201, 2020, pages A6363
GOBEL ET AL.: "The Coagulation Factors Fibrinogen, Thrombin, and Factor XII in Inflammatory Disorders-A Systematic Review", FRONT. IMMUNOL, 26 July 2018 (2018-07-26)
SCHEFFEL ET AL.: "Cold-induced urticarial autoinflammatory syndrome related to factor XII activation", NATURE COMMUNICATIONS, vol. 11, 2020
PEYROU ET AL.: "The kallikrein-kinin pathway as a mechanism for auto-control of brown adipose tissue activity", NATURE COMMUNICATIONS, vol. 11, 2020, pages 2132, Retrieved from the Internet
WYGRECKA ET AL., USE OF A FXLLA-LNHIBITOR IN THE TREATMENT OF RENAL FIBROSIS AND/OR KIDNEY DISEASE
PHILIPPOU ET AL., FACTOR XLLA INHIBITORS
BRESLOW ET AL., AMINOTRIAZOLE IMMUNOMODULATORS FOR TREATING AUTOIMMUNE DISEASES
PONDA, AMINACYLINDAZOLE IMMUNOMODULATORS FOR TREATMENT OF AUTOIMMUNE DISEASES
NOLTE ET AL., FACTOR XII INHIBITORS FOR THE ADMINISTRATION WITH MEDICAL PROCEDURES COMPRISING CONTACT WITH ARTIFICIAL SURFACES
WILBS ET AL., NAT COMMUN, vol. 11, 2020, pages 3890
S. PATEL, RETINA, vol. 29, June 2009 (2009-06-01), pages S45 - 8
ELMAN MAIELLO LBECK R ET AL.: "Randomized trial evaluating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema", OPHTHALMOLOGY, 27 April 2010 (2010-04-27)
STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection and Use", 2002, WILEY-VCH
"The Practice of Medicinal Chemistry", 2003, pages: 561 - 585
F. J. LEINWEBER: "Drug Metab. Res", vol. 18, 1987, pages: 379
ARYLBORONIC ESTERST. ISHIYAMAM. MURATAN. MIYAURA, J. ORG. CHEM., vol. 60, 1995, pages 7508 - 7510
LIANGCHEN, EXPERT OPINION IN THERAPEUTIC PATENTS, vol. 11, no. 6, 2001, pages 981 - 986
H. LIEBERMANL. LACHMAN: "Pharmaceutical Dosage Forms", vol. 1, 1980, MARCEL DEKKER
T. W. GREENEP. G. M. WUTS: "Protective groups in organic chemistry", 2006, JOHN WILEY AND SONS
MAEHR, J. CHEM. ED, vol. 62, 1985, pages 114 - 120
CAS , no. 1522098-73-2
TETRAHEDRON LETTERS, vol. 55, 2014, pages 466 - 468
CHEMISTRYSELECT, vol. 4, 2019, pages 4933 - 493
Attorney, Agent or Firm:
HOGGETT, Christopher James et al. (GB)
Download PDF:
Claims:
Claims

1. A compound of Formula (I),

Formula (I), wherein:

V is selected from -O- and -N(R23)-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or,

V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is selected from -O- and -N(R18)-; wherein R18 or R23 are selected from C(=S)NH-aryl; C(=S)NH-R19; C(=S)N(alkyl)R19; C(=S)NR13R14; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkylb; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one substituent selected from cycloalkyl and heterocycloalkylb, and optionally one or two further substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3;

R19 is selected from (CH2)1-3-cycloalkyl, (CH2)0-3-polycycloalkyl, heterocycloalkyla, (CH2)1-3-heterocycloalkylb, (CH2)0-3-R22, (C(R2O)(R21))-(CH2)0-3-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)1-3-O-(CH2)-phenyl;

R20 and R21 are independently selected from H, alkyl and CF3, or R20 and R21 together with the carbon atom to which they are attached form a carbon-containing 3-, 4-, 5- or 6- membered saturated ring, optionally containing an O ring member, wherein R20 and R21 are not both H; R22 is a fused 6,5- or 6,6- bicyclic ring containing an aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6bicyclic ring is not aryl substituted with ethylenedioxy;

Y is selected from -O- and -N(R12)-;

B is selected from:

(i) heteroaryla; and

(ii) aryl;

AW- is selected from:

-(CH2)O-6-(CHR15)-(CH2)0-6-A, -(CHR12)-A, -O-(CHR12)-A, -(CH2)O.6-A, -(CH2)0-6-O-(CH2)0-6-A, -(CH2)0-6-NH-(CH2)0-6-A, -(CH2)O-6-N(R12)-(CH2)I.6-C(=0)-A, -(CH2)0-6-NH-C(=O)-(CH2)0-6-A,

-C(=0)N(R12)-(CH2)0-6-A, -(CH2)O-6-C(=0)-(CH2)0-6-A, -(CH2)0-6-(phenyl)-(CH2)0-6-A, -NH-SO2-A and -SO2-NH-A;

A is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN, and phenyl; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system; wherein when:

(i) V is -O-, or

(ii) Z is -O-,

A is A', wherein A' is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, wherein the ring system is substituted with phenyl and optionally, where possible, 1, 2, or 3 further substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN, and phenyl; wherein when A' is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A' is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system; alkoxy is OCF3, a linear O-linked hydrocarbon of between 1 and 6 carbon atoms (C1-C6), or a branched O-linked hydrocarbon of between 3 and 6 carbon atoms (C3-C6); alkoxy may optionally be substituted, where possible, with 1 or 2 substituents independently selected from OH, CN, CF3, and fluoro; alkyl is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, -NR13R14, -C(=O)OR13, -C(=O)NR13R14, CN, CF3, halo; alkylb is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkylb may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, CN, CF3, halo; small alkyl is a linear saturated hydrocarbon having up to 4 carbon atoms (C1-C4) or a branched saturated hydrocarbon of between 3 and 4 carbon atoms (C3-C4); small alkyl may optionally be substituted with 1 or 2 substituents independently selected from alkoxy, OH, NR13R14, C(=O)OR13, C(=O)NR13R14, CN, CF3, halo; aryl is phenyl, biphenyl or naphthyl; aryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, methylenedioxy, ethylenedioxy, OH, halo, CN, -(CH2)0-3- O-heteroaryla, arylb, -O-arylb, -(CH2)1-3-arylb, -(CH2)0-3-heteroaryla, -C(=O)OR13, -C(=O)NR13R14, -(CH2)0-3-NR13R14, OCF3 and CF3; a ry I b is phenyl, biphenyl or naphthyl; a ryl b may be optionally substituted with 1, 2 or 3 substituents independently selected from alkylb, alkoxy, OH, halo, CN, and CF3; cycloalkyl is a monocyclic saturated hydrocarbon ring of between 3 and 6 carbon atoms (C3-C6); cycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo; halo is F, Cl, Br, or I; heteroaryl is a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members that are selected from N, NR8, S, and O; heteroaryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3; heteroaryla is a 5-, 6-, 9- or 10- membered mono- or bi-cyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O; heteroaryla may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, arylb, -(CH2)o-3-NR13R14, heteroarylb, -C(=O)OR12, -C(=O)NR13R14 and CF3; heteroarylb is a 5-, 6-, 9- or 10- membered mono- or bi-cyclic aromatic ring, containing, where possible, 1, 2 or 3 ring members independently selected from N, N(R12), S and O; wherein heteroarylb may be optionally substituted with 1, 2 or 3 substituents independently selected from alkylb, alkoxy, OH, halo, CN, arylb, -(CH2)i-3-arylb, and CF3; heterocycloalkyl is a non-aromatic carbon-containing monocyclic ring containing 3, 4, 5, or 6, ring members, wherein at least one ring member is independently selected from N, N(R12), S, and O; heterocycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo; heterocycloalkyla is a 3-10 membered non-aromatic carbon-containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O; heterocycloalkyla may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, aryl, halo, OH, CN, CF3, and heterocycloalkyl; wherein when heterocycloalkyla is a bicyclic ring system, the bicyclic ring system is spiro; heterocycloalkylb is a 3-6 membered non-aromatic carbon-containing monocyclic ring system which may be saturated or, where possible, unsaturated with 1 or 2 double bonds; heterocycloalkylb contains one or two ring members independently selected from N, N(C(=O)CH3) and O; heterocycloalkyla may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, halo, OH, CN, CF3, phenyl and oxo; wherein the point of attachment of heterocycloalkylb is not at a ring nitrogen; polycycloalkyl is a bi- or tri-cyclic saturated hydrocarbon ring system of between 5 and 10 carbon atoms (C5-C10), wherein the bi- or tri-cyclic ring system is bridged or spiro;

R8 is independently selected from H, alkyl, cycloalkyl, and heterocycloalkyl;

R12 is independently selected from H, alkyl, and cycloalkyl;

R13 and R14 are independently selected from H, alkylb, arylb and heteroarylb or R13 and R14 together with the nitrogen atom to which they are attached form a carbon-containing 4-, 5-, 6- or 7-membered heterocyclic ring, optionally containing an additional heteroatom selected from N, N(R12), S, SO, SO2, and O, which may be saturated or unsaturated with 1 or 2 double bonds and which may be optionally mono- or di-substituted with substituents selected from oxo, alkylb, alkoxy, OH, halo and CF3;

R15 is selected from alkyl, halo, CF3, CN, OH, alkoxy, NR13R14, and CONR13R14;

R16 and R17 are independently selected from H and small alkyl; and tautomers, isomers, stereoisomers (including enantiomers, diastereoisomers and racemic and scalemic mixtures thereof), deuterated analogues, and pharmaceutically acceptable salts and/or solvates thereof.

2. A compound of formula (I) according to claim 1, wherein the compound is a compound of formula (la), formula (lb), or formula (Ic),

Formula (la),

Formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

3. A compound according to claim 1, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or,

V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-.

4. A compound of formula (I) according to any of the preceding claims, wherein the compound is a compound of formula (la),

Formula (la), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

5. A compound according to any of the preceding claims, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkylb; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

6. A compound according to claim 5, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- membered heterocycloalkylb ring, optionally substituted as for heterocycloalkylb; a 6- membered carbon-containing aromatic ring containing two N ring members substituted with one

7. A compound according to any of the preceding claims or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is a compound of formula (la), wherein R18 is as described in any of the preceding claims, or a compound of formula (lb) and R23 is C(=O)R19.

8. A compound according to any of claims 3 to 7, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein A is a 6 to 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro.

9. A compound according to claim 8, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A is selected from:

10. A compound according to claim 1 or 2, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

V is -O-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or,

V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is -O-.

11. A compound according to claim 1, 2 or 9, wherein the compound is a compound of formula (Ic),

Formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

12. A compound according any of the preceding claims, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A.

13. A compound according to any of the preceding claims, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

Y is -N(R12)-.

14. A compound according to claim 13, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

Y is -NH-.

15. A compound according to any of the preceding claims, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is heteroaryla.

16. A compound according to claim 15, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is isoquinolinyl, optionally substituted as for heteroaryla.

17. A compound according to claim 16 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is optionally substituted as for heteroaryla.

18. A compound according to claim 16 or 17, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is isoquinolinyl, substituted with NH2 at the 1- position optionally further substituted with 1 or 2 substituents as for heteroaryla.

19. A compound according to claim 18, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is optionally further substituted with 1 or 2 substituents as for heteroaryla.

20. A compound selected from Table 1, Table 2, Table 3, Table 4, and Table 5, or a pharmaceutically acceptable salt, solvate, or solvate of a salt thereof.

21. A pharmaceutical composition comprising: a compound, or a pharmaceutically acceptable salt and/or solvate thereof, according to any of claims 1 to 20, and at least one pharmaceutically acceptable excipient.

22. A compound, or a pharmaceutically acceptable salt and/or solvate thereof, as defined in any of claims 1 to 20, or the pharmaceutical composition according to claim 21, for use in medicine.

23. A compound, or a pharmaceutically acceptable salt and/or solvate thereof, as defined in any of claims 1 to 20, or the pharmaceutical composition according to claim 21, for use in a method of treatment of a disease or condition in which Factor XI la activity is implicated; wherein the disease or condition in which Factor Xlla activity is implicated is a bradykinin-mediated angioedema, wherein the bradykinin-mediated angioedema is hereditary angioedema.

24. A compound, or a pharmaceutically acceptable salt and/or solvate thereof, as defined in any of claims 1 to 20, or the pharmaceutical composition according to claim 21 for use in a method of treatment of a disease or condition in which Factor Xlla activity is implicated; wherein the disease or condition in which Factor Xlla activity is implicated is a bradykinin-mediated angioedema, wherein the bradykinin-mediated angioedema is non hereditary.

25. A compound, or a pharmaceutically acceptable salt and/or solvate thereof, as defined in any of claims 1 to 20, or the pharmaceutical composition according to claim 21, for use in a method of treatment of a disease or condition in which Factor Xlla activity is implicated; wherein the disease or condition in which Factor Xlla activity is implicated is a thrombotic disorder.

Description:
2-AZA- AND 2-OXABICYCLO[2.1.1]HEXANE DERIVATIVES AS FACTOR XIIA ENZYME INHIBITORS

This invention relates to enzyme inhibitors that are inhibitors of Factor XI la (FXIIa), and to pharmaceutical compositions comprising, and uses of, such inhibitors.

Background to the invention

The compounds of the present invention are inhibitors of factor Xlla (FXIIa) and thus have a number of possible therapeutic applications, particularly in the treatment of diseases or conditions in which factor Xlla inhibition is implicated.

FXIIa is a serine protease (EC 3.4.21.38) derived from its zymogen precursor, factor XII (FXII), which is expressed by the F12 gene. Single chain FXII has a low level of amidolytic activity that is increased upon interaction with negatively charged surfaces and has been implicated in its activation (see Invanov et al., Blood. 2017 Mar 16;129(11):1527-1537. doi: 10.1182/blood-2016-10-744110). Proteolytic cleavage of FXII to heavy and light chains of FXIIa dramatically increases catalytic activity. FXIIa that retains its full heavy chain is aFXIIa. FXIIa that retains a small fragment of its heavy chain is PFXIIa. The separate catalytic activities of aFXIIa and PFXIIa contribute to the activation and biochemical functions of FXIIa. Mutations and polymorphisms in the F12 gene can alter the cleavage of FXII and FXIIa.

FXIIa has a unique and specific structure that is different from many other serine proteases. For instance, the Tyr99 in FXIIa points towards the active site, partially blocking the S2 pocket and giving it a closed characteristic. Other serine proteases containing a Tyr99 residue (e.g. FXa, tPA and FIXa) have a more open S2 pocket. Moreover, in several trypsin-like serine proteases the P4 pocket is lined by an "aromatic box" which is responsible for the P4-driven activity and selectivity of the corresponding inhibitors. However, FXIIa has an incomplete "aromatic box" resulting in more open P4 pocket. See e.g. "Crystal structures of the recombinant ^-factor Xlla protease with bound Thr-Arg and Pro-Arg substrate mimetics" M. Pathak et al., Acta. Cryst.2019, D75, 1-14; "Structures of human plasma ^-factor Xlla cocrystallized with potent inhibitors" A Dementiev et al., Blood Advances 2018, 2(5), 549-558; "Design of Small-Molecule Active-Site Inhibitors of theSlA Family Proteases as Procoagulant and Anticoagulant Drugs" P. M. Fischer, J. Med. Chem., 2018, 61(9), 3799-3822; "Assessment of the protein interaction between coagulation factor XII and corn trypsin inhibitor by molecular docking and biochemical validation" B. K. Hamad et al. Journal of Thrombosis and Haemostasis, 15: 1818-1828. FXIIa converts plasma prekallikrein (PK) to plasma kallikrein (PKa), which provides positive feedback activation of FXII to FXIIa. FXII, PK, and high molecular weight kininogen (HK) together represent the contact system. FXIIa mediated conversion of plasma prekallikrein to plasma kallikrein can cause subsequent cleavage of HKto generate bradykinin, a potent inflammatory hormone that can also increase vascular permeability, which has been implicated in disorders such as hereditary angioedema (HAE). The contact system is activated via a number of mechanisms, including interactions with negatively charged surfaces, negatively charged molecules, unfolded proteins, artificial surfaces, foreign tissue (e.g. biological transplants, that include bio-prosthetic heart valves, and organ/tissue transplants), bacteria, and biological surfaces (including endothelium and extracellular matrix) that mediate assembly of contact system components. In addition, the contact system is activated by plasmin, and cleavage of FXII by other enzymes can facilitate its activation.

Activation of the contact system leads to activation of the kallikrein kinin system (KKS), complement system, and intrinsic coagulation pathway (see https://www.genome.jp/kegg- bin/show_pathway?map04610). In addition, FXIIa has additional substrates both directly, and indirectly via PKa, including Proteinase-activated receptors (PARs), plasminogen, and neuropeptide Y (NPY) which can contribute to the biological activity of FXIIa. Inhibition of FXIIa could provide clinical benefits by treating diseases and conditions associated with these systems, pathways, receptors, and hormones.

PKa activation of PAR2 mediates neuroinflammation and may contribute to neuroinflammatory disorders including multiple sclerosis (see Gbbel et al., Proc Natl Acad Sci U S A. 2019 Jan 2;116(1):271-276. doi: 10.1073/pnas.1810020116). PKa activation of PARI and PAR2 on vascular smooth muscle cells has been implicated in vascular hypertrophy and atherosclerosis (see Abdallah et al., J Biol Chem. 2010 Nov 5;285(45):35206-15. doi: 10.1074/jbc.M110.171769). FXIIa activation of plasminogen to plasmin contributes to fibrinolysis (see Konings et al., Thromb Res. 2015 Aug;136(2):474-80. doi: 10.1016/j.thromres.2015.06.028). PKa proteolytically cleaves NPY and thereby alters its binding to NPY receptors (Abid et al., J Biol Chem. 2009 Sep ll;284(37):24715-24. doi: 10.1074/jbc.M109.035253). Inhibition of FXIIa could provide clinical benefits by treating diseases and conditions caused by PAR signaling, NPY metabolism, and plasminogen activation.

FXIIa-mediated activation of the KKS results in the production of bradykinin (BK), which can mediate, for example, angioedema, pain, inflammation, vascular hyperpermeability, and vasodilatation (see Kaplan et al., Adv Immunol. 2014;121:41-89. doi: 10.1016/B978-0-12-800100-4.00002-7; and Hopp et al., J Neuroinflammation. 2017 Feb 20;14(1):39. doi: 10.1186/sl2974-017-0815-8). Garadacimab (CSL-312), a monoclonal antibody inhibitory against FXIIa, recently completed a phase 3 study where monthly prophylactic subcutaneous treatment was reported to reduce attacks in patients with type l/ll hereditary angioedema (HAE), which results in intermittent swelling of face, hands, throat, gastro-intestinal tract and genitals (see https://www.clinicaltrials.gov/ct2/show/NCT04656418 and Craig et al., The Lancet. 2023;401(10382):1079-1090. doi: 10.1016/50140-6736(23)00350-1). Mutations in FXII that facilitate its activation to FXIIa have been identified as a cause of HAE (see Bjbrkqvist et al., J Clin Invest. 2015 Aug 3;125(8):3132-46. doi: 10.1172/JCI77139; and de Maat et al., J Allergy Clin Immunol. 2016 Nov;138(5):1414-1423.e9. doi: 10.1016/j.jaci.2016.02.021). Since FXIIa mediates the generation of PK to PKa, inhibitors of FXIIa could provide protective effects of all form of BK-mediated angioedema, including HAE and non-hereditary bradykinin-mediated angioedema (BK-AEnH).

"Hereditary angioedema" can be defined as any disorder characterised by recurrent episodes of bradykinin-mediated angioedema (e.g. severe swelling) caused by an inherited genetic dysfunction/fault/mutation. There are currently three known categories of HAE: (i) HAE type 1, (ii) HAE type 2, and (iii) normal Cl inhibitor HAE (normal Cl-lnh HAE). However, work on characterizing the etiologies of HAE is ongoing so it is expected that further types of HAE might be defined in the future.

Without wishing to be bound by theory, it is thought that HAE type 1 is caused by mutations in the SERPING1 gene that lead to reduced levels of Cl inhibitor in the blood. Without wishing to be bound by theory, it is thought that HAE type 2 is caused by mutations in the SERPING1 gene that lead to dysfunction of the Cl inhibitor in the blood. Without wishing to be bound by theory, the cause of normal Cl-lnh HAE is less well defined and the underlying genetic dysfunction/fault/mutation can sometimes remain unknown. What is known is that the cause of normal Cl-lnh HAE is not related to reduced levels or dysfunction of the Cl inhibitor (in contrast to HAE types 1 and 2). Normal Cl-lnh HAE can be diagnosed by reviewing the family history and noting that angioedema has been inherited from a previous generation (and thus it is hereditary angioedema). Normal Cl-lnh HAE can also be diagnosed by determining that there is a dysfunction/fault/mutation in a gene other than those related to Cl inhibitor. For example, it has been reported that dysfunction/fault/mutation with plasminogen can cause normal Cl-lnh HAE (see e.g. Veronez et al., Front Med (Lausanne). 2019 Feb 21;6:28. doi: 10.3389/fmed.2019.00028; or Recke et al., Clin Transl Allergy. 2019 Feb 14;9:9. doi: 10.1186/sl3601-019- 0247-x.). It has also been reported that dysfunction/fault/mutation with Factor XII can cause normal Cl- lnh HAE (see e.g. Mansi et al. 2014 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine, 2015, 277; 585-593; or Maat et al. J Thromb Haemost. 2019 Jan;17(1):183- 194. doi: 10.1111/jth.14325). However, angioedemas are not necessarily inherited. Indeed, another class of angioedema is bradykinin mediated angioedema non-hereditary (BK-AEnH), which is not caused by an inherited genetic dysfunction/fault/mutation. Often the underlying cause of BK-AEnH is unknown and/or undefined. However, the signs and symptoms of BK-AEnH are similar to those of HAE, which, without being bound by theory, is thought to be on account of the shared bradykinin mediated pathway between HAE and BK-AEnH. Specifically, BK-AEnH is characterised by recurrent acute attacks where fluids accumulate outside of the blood vessels, blocking the normal flow of blood or lymphatic fluid and causing rapid swelling of tissues such as in the hands, feet, limbs, face, intestinal tract, airway or genitals.

Specific types of BK-AEnH include: non hereditary angioedema with normal Cl Inhibitor (AE-nCl Inh), which can be environmental, hormonal, or drug induced; acquired angioedema; anaphylaxis associated angioedema; angiotensin converting enzyme (ACE) inhibitor induced angioedema; dipeptidyl peptidase 4 inhibitor induced angioedema; and tPA induced angioedema (tissue plasminogen activator induced angioedema). However, reasons why these factors and conditions cause angioedema in only a relatively small proportion of individuals are unknown.

Environmental factors that can induce AE-nCl Inh include air pollution (Kedarisetty et al, Otolaryngol Head Neck Surg. 2019 Apr 30:194599819846446. doi: 10.1177/0194599819846446) and silver nanoparticles such as those used as antibacterial components in healthcare, biomedical and consumer products (Long et al., Nanotoxicology. 2016;10(4):501-ll. doi: 10.3109/17435390.2015.1088589).

Various publications suggest a link between the bradykinin and contact system pathways and BK-AEnHs, and also the potential efficacy of treatments, see e.g.: Bas et al. (N Engl J Med 2015; Leibfried and Kovary. J Pharm Pract 2017); van den Elzen et al. (Clinic Rev Allerg Immunol 2018); Han et al (JCI 2002).

For instance, BK-medicated AE can be caused by thrombolytic therapy. For example, tPA induced angioedema is discussed in various publications as being a potentially life threatening complication following thrombolytic therapy in acute stroke victims (see e.g. Simao et al., Blood. 2017 Apr 20;129(16):2280-2290. doi: 10.1182/blood-2016-09-740670; Frohlich et al., Stroke. 2019 Jun 11:STROKEAHA119025260. doi: 10.1161/STROKEAHA.119.025260; Rathbun, Oxf Med Case Reports. 2019 Jan 24;2019(1):omyll2. doi: 10.1093/omcr/omyll2; Lekoubou et al., Neurol Res. 2014 Jul;36(7):687-94. doi: 10.1179/1743132813Y.0000000302; Hill et al., Neurology. 2003 May 13;60(9):1525-7).

Stone et al. (Immunol Allergy Clin North Am. 2017 Aug;37(3):483-495.) reports that certain drugs can cause angioedema. Scott et al. (Curr Diabetes Rev. 2018;14(4):327-333. doi: 10.2174/1573399813666170214113856) reports cases of dipeptidyl Peptidase-4 Inhibitor induced angioedema.

Hermanrud et al., (BMJ Case Rep. 2017 Jan 10;2017. pii: bcr2016217802) reports recurrent angioedema associated with pharmacological inhibition of dipeptidyl peptidase IV and also discusses acquired angioedema related to angiotensin-converting enzyme inhibitors (ACEI-AAE). Kim et al. (Basic Clin Pharmacol Toxicol. 2019 Jan;124(1):115-122. doi: 10.1111/bcpt.l3097) reports angiotensin II receptor blocker (ARB)-related angioedema. Reichman et al., (Pharmacoepidemiol Drug Saf. 2017 Oct;26(10):1190- 1196. doi: 10.1002/pds.4260) also reports angioedema risk for patients taking ACE inhibitors, ARB inhibitors and beta blockers. Diestro et al. (J Stroke Cerebrovasc Dis. 2019 May;28(5):e44-e45. doi: 10.1016/j.jstrokecerebrovasdis.2019.01.030) also reports a possible association between certain angioedemas and ARBs.

Giard et al. (Dermatology. 2012;225(1):62-9. doi: 10.1159/000340029) reports that bradykinin mediated angioedema can be precipitated by estrogen contraception, so called "oestrogen associated angioedema".

Contact system mediated activation of the KKS has also been implicated in retinal edema and diabetic retinopathy (see Liu et al., Biol Chem. 2013 Mar;394(3):319-28. doi: 10.1515/hsz-2012-0316). FXIIa concentrations are increased in the vitreous fluid from patients with advance diabetic retinopathy and in Diabetic Macular Edema (DME) (see Gao et al., Nat Med. 2007 Feb;13(2):181-8. Epub 2007 Jan 28 and Gao et al., J Proteome Res. 2008 Jun;7(6):2516-25. doi: 10.1021/pr800112g). FXIIa has been implicated in mediating both vascular endothelial growth factor (VEGF) independent DME (see Kita et al., Diabetes. 2015 Oct;64(10):3588-99. doi: 10.2337/dbl5-0317) and VEGF mediated DME (see Clermont et al., Invest Ophthalmol Vis Sci. 2016 May l;57(6):2390-9. doi: 10.1167/iovs.15-18272). FXII deficiency is protective against VEGF induced retinal edema in mice (Clermont et al., ARVO talk 2019). Therefore, it has been proposed that FXIIa inhibition will provide therapeutic effects for diabetic retinopathy and retinal edema caused by retinal vascular hyperpermeability, including DME, retinal vein occlusion, age-related macular degeneration (AMD).

As noted above, the contact system can be activated by interaction with bacteria, and therefore FXIIa has been implicated in the treatment of sepsis and bacterial sepsis (see Morrison et al., J Exp Med. 1974 Sep l;140(3):797-811). Therefore, FXIIa inhibitors could provide therapeutic benefits in treating sepsis, bacterial sepsis and disseminated intravascular coagulation (DIC). FXIIa mediated activation of the KKS and production of BK have been implicated in neurodegenerative diseases including Alzheimer's disease, multiple sclerosis, epilepsy and migraine (see Zamolodchikov et al., Proc Natl Acad Sci U S A. 2015 Mar 31;112(13):4068-73. doi: 10.1073/pnas.l423764112; Simoes et al., J Neurochem. 2019 Aug;150(3):296-311. doi: 10.1111/jnc.14793; Gobel et al., Nat Commun. 2016 May 18;7:11626. doi: 10.1038/ncommsll626; and https://clinicaltrials.gov/ct2/show/NCT03108469). Therefore, FXIIa inhibitors could provide therapeutic benefits in reducing the progression and clinical symptoms of these neurodegenerative diseases.

FXIIa has also been implicated in anaphylaxis (see Bender et al., Front Immunol. 2017 Sep 15;8:1115. doi: 10.3389/fimmu.2017.01115; and Sala-Cunill et al., J Allergy Clin Immunol. 2015 Apr;135(4):1031-43.e6. doi: 10.1016/j.jaci.2014.07.057). Therefore, FXIIa inhibitors could provide therapeutic benefits in reducing the clinical severity and incidence of anaphylactic reactions.

The role of FXIIa in coagulation was identified over 50 years ago, and has been extensively documented in publications using biochemical, pharmacological, genetic and molecular studies (see Davie et al., Science. 1964 Sep 18;145(3638):1310-2). FXIIa mediated activation of factor XI (FXI) triggers the intrinsic coagulation pathway. In addition, FXIIa can increase coagulation in a FXI independent manner (see Radcliffe et al., Blood. 1977 Oct;50(4):611-7; and Puy et al., J Thromb Haemost. 2013 Jul;ll(7):1341-52. doi: 10.1111/jth.12295). Studies on both humans and experimental animal models have demonstrated that FXII deficiency prolongs activated partial prothrombin time (APTT) without adversely affecting hemostasis (see Renne et al., J Exp Med. 2005 Jul 18;202(2):271-81; and Simao et al., Front Med (Lausanne). 2017 Jul 31;4:121. doi: 10.3389/fmed.2017.00121). Pharmacological inhibition of FXIIa also prolongs APTT without increasing bleeding (see Worm et al., Ann Transl Med. 2015 Oct;3(17):247. doi: 10.3978/j.issn.2305-5839.2015.09.07). These data suggest that inhibition of FXIIa could provide therapeutic effects against thrombosis without inhibiting bleeding. Therefore, FXIIa inhibitors could be used to treat a spectrum of prothrombotic conditions including venous thromboembolism (VTE); cancer associated thrombosis; complications caused by mechanical and bioprosthetic heart valves, catheters, extracorporeal membrane oxygenation (ECMO), left ventricular assisted devices (LVAD), dialysis, cardiopulmonary bypass (CPB); sickle cell disease, joint arthroplasty, thrombosis induced by tPA, Paget-Schroetter syndrome and Budd-Chari syndrome. FXIIa inhibitor could be used for the treatment and/or prevention of thrombosis, edema, and inflammation associated with these conditions.

Surfaces of medical devices that come into contact with blood can cause thrombosis. FXIIa inhibitors may also be useful for treating or preventing thromboembolism by lowering the propensity of devices that come into contact with blood to clot blood. Examples of devices that come into contact with blood include vascular grafts, stents, in-dwelling catheters, external catheters, orthopedic prosthesis, cardiac prosthesis, and extracorporeal circulation systems.

Preclinical studies have shown that FXIIa has been shown to contribute to stroke and its complications following both ischemic stroke, and hemorrhagic accidents (see Barbieri et al., J Pharmacol Exp Ther. 2017 Mar;360(3):466-475. doi: 10.1124/jpet.ll6.238493; Krupka et al., PLoS One. 2016 Jan 27;ll(1):e0146783. doi: 10.1371/journal.pone.0146783; Leung et al., Transl Stroke Res. 2012 Sep;3(3):381-9. doi: 10.1007/S12975-012-0186-5; Simao et al., Blood. 2017 Apr 20;129(16):2280-2290. doi: 10.1182/blood- 2016-09-740670; and Liu et al., Nat Med. 2011 Feb;17(2):206-10. doi: 10.1038/nm.2295). Therefore, FXIIa inhibition may improve clinical neurological outcomes in the treatment of patients with stroke.

FXII deficiency has been shown to reduce the formation of atherosclerotic lesions in Apoe _/_ mice (Didiasova et al., Cell Signal. 2018 Nov;51:257-265. doi: 10.1016/j.cellsig.2018.08.006). Therefore, FXIIa inhibitors could be used in the treatment of atherosclerosis.

FXIIa, either directly, or indirectly via PKa, has been shown to activate the complement system (Ghebrehiwet et al., Immunol Rev. 2016 Nov;274(1):281-289. doi: 10.1111/imr.l2469). BK increases complement C3 in the retina, and an in vitreous increase in complement C3 is associated with DME (Murugesan et al., Exp Eye Res. 2019 Jul 24;186:107744. doi: 10.1016/j.exer.2019.107744). Both FXIIa and PKa activate the complement system (see Irmscher et al., J Innate Immun. 2018;10(2):94-105. doi: 10.1159/000484257; and Ghebrehiwet et al., J Exp Med. 1981 Mar l;153(3):665-76).

A phase 2 study to assess the safety and efficacy of CSL312, a FXIIa inhibitor, in the treatment of COVID-19 has been assigned clinicaltrials.gov identifier NCT04409509. Shatzel et al. (Res Pract Thromb Haemost, 2020 May 15;4(4):500-505. doi: 10.1002/rth2.12349) also relates to investigating the contact system's role in COVID-19.

Wygrecka et al. ("Coagulation factor XII regulates inflammatory responses in human lungs", European Respiratory Journal 2017 50: PA339; DOI: 10.1183/1393003. congress-2017. PA339) relates to the effect of an accumulation of FXII in acute respiratory distress syndrome (ARDS) lungs.

Wong et al. ("CSL312, a Novel Anti-FXII Antibody, Blocks FXII-lnduced IL-6 Production from Primary Non- Diseased and Idiopathic Pulmonary Fibrosis Fibroblasts", American Journal of Respiratory and Critical Care Medicine 2020;201:A6363) reports that activated FXII may contribute to lung fibrosis (e.g. idiopathic

Pulmonary Fibrosis) through direct stimulation of fibroblasts to produce pro-fibrotic cytokine IL-6.

Gbbel et al. (The Coagulation Factors Fibrinogen, Thrombin, and Factor XII in Inflammatory Disorders— A Systematic Review, Front. Immunol., 26 July 20181 https://doi.org/10.3389/fimmu.2018.01731) relates to FXII's role in the rheumatoid arthritis (RA).

Scheffel et al. (Cold-induced urticarial autoinflammatory syndrome related to factor XII activation, Nature Communications volume 11, Article number: 179 (2020)) reports that there is a link between contact system activation and cytokine-mediated inflammation, such as cold-induced urticarial autoinflammatory syndrome.

Peyrou et al. (The kallikrein— kinin pathway as a mechanism for auto-control of brown adipose tissue activity, NATURE COMMUNICATIONS, (2020) 11:2132, https://doi.org/10.1038/s41467-020-16009-x), reports a pathway for controlling brown adipose tissue (BAT) thermogenic activity mediated by the kallikrein— kinin system, which may contribute to expanding the range of potential pharmacological candidates in therapeutic strategies against obesity and associated diseases designed to improve energy expenditure and remove excess blood metabolites through activation of BAT. Impaired BAT activity is associated with obesity and insulin resistance.

Wygrecka et al. ("Use of a FXIIa-lnhibitor in the treatment of renal fibrosis and/or kidney disease" WO2019/113642) relates to an inhibitor of Factor XII for use in treating or preventing kidney disease, renal fibrosis, glomerulosclerosis, renal scarring, ischemia/reperfusion injury in native or transplant kidneys and/or acute kidney injury.

Compounds that are said to be FXIIa inhibitors have been described by Rao et al. ("Factor XI la Inhibitors" WO2018/093695), Hicks et al. ("Factor XI la Inhibitors" WO2018/093716), Breslow et al. ("Aminotriazole immunomodulators for treating autoimmune diseases" WO2017/123518) and Ponda et al. ("Aminacylindazole immunomodulators for treatment of autoimmune diseases" WO2017/205296 and "Pyranopyrazole and pyrazolopyridine immunomodulators for treatment of autoimmune diseases" WO2019/108565). FXII/FXIIa inhibitors are said to have been described by Nolte et al. ("Factor XII inhibitors for the administration with medical procedures comprising contact with artificial surfaces" W02012/120128). Compounds that are said to be modulators of FXIIa have been described by Philippou et al. ("Factor XI la Inhibitors" WO 2019/211585 and WO 2019/186164). Macrocylic peptides that are said to be inhibitors of FXIIa have been described by Wilbs et al. (Nat Commun 11, 3890 (2020). Doi: 10.1038/s41467-020-17648- w).

To date, no FXIIa inhibitors have been approved for medical use, and there are no small molecule FXIIa inhibitors in clinical development. Although certain known compounds are said to be modulators or inhibitors of FXIIa, these compounds can suffer from limitations such as being non-reversible or covalent binders, being poorly selective for FXIIa over other related enzymes, or not having pharmacokinetic properties suitable for oral therapy. For example, compounds with acylating reactivity e.g. acylated aminotriazoles, are typically non-reversible covalent binders, and can sometimes also be unstable in water and/or blood plasma due to their inherent reactivity. Poor selectivity for FXIIa over other serine proteases (such as thrombin, FXa, FXIa, KLK1, plasmin, trypsin) increases the risk of off-target effects, which can be made even worse (i.e. there is typically a higher likelihood of poor selectivity and off-target effects) if the inhibitor is a covalent binder. Therefore, there remains a need to develop new FXIIa inhibitors that are not covalent inhibitors and/or are highly selective for FXIIa in order to e.g. mitigate the risks of non- selectivity and cytotoxicity. There is a particular need to develop a small molecule FXIIa inhibitors, particularly as an oral therapy.

In view of the above, there also remains a need to develop new FXIIa inhibitors that will have utility to treat a wide range of disorders, in particular angioedema; HAE, including : (i) HAE type 1, (ii) HAE type 2, and (iii) normal Cl inhibitor HAE (normal Cl-lnh HAE); BK-AEnH, including AE-nCl Inh, ACE and tPA induced angioedema; vascular hyperpermeability; stroke including ischemic stroke and haemorrhagic accidents; retinal edema; diabetic retinopathy; impaired visual acuity; DME; retinal vein occlusion; AMD; neuroinflammation; neuroinflammatory/neurodegenerative disorders such as MS (multiple sclerosis); other neurodegenerative diseases such as Alzheimer's disease, epilepsy and migraine; sepsis; bacterial sepsis; inflammation; anaphylaxis; thrombosis; thromboembolism caused by increased propensity of medical devices that come into contact with blood to clot blood; prothrombotic conditions including disseminated intravascular coagulation (DIC), venous thromboembolism (VTE), cancer associated thrombosis, complications caused by mechanical and bioprosthetic heart valves, complications caused by catheters, complications caused by ECMO, complications caused by LVAD, complications caused by dialysis, complications caused by CPB, sickle cell disease, joint arthroplasty, thrombosis induced to tPA, Paget-Schroetter syndrome and Budd-Chari syndrome; atherosclerosis; COVID-19; acute respiratory distress syndrome (ARDS); idiopathic pulmonary fibrosis (IPF); rheumatoid arthritis (RA); cold-induced urticarial autoinflammatory syndrome; obesity; diabetes; kidney disease; renal fibrosis; glomerulosclerosis; renal scarring; ischemia/reperfusion injury in native or transplant kidneys; and acute kidney injury. In particular, there remains a need to develop new FXIIa inhibitors.

Description of the Invention

The present invention relates to a series of inhibitors of Factor Xlla (FXIIa). The compounds of the invention are potentially useful in the treatment of diseases or conditions in which factor Xlla inhibition is implicated. The invention further relates to pharmaceutical compositions of the inhibitors, to the use of the compositions as therapeutic agents, and to methods of treatment using these compositions.

Specifically, the invention provides compounds of Formula (I),

Formula (I), wherein:

V is selected from -O- and -N(R23)-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or,

V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is selected from -O- and -N(R18)-; wherein R18 or R23 are selected from C(=S)NH-aryl; C(=S)NH-R19; C(=S)N(alkyl)R19; C(=S)NR13R14; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one substituent selected from cycloalkyl and heterocycloalkyl b , and optionally one or two further substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3; R19 is selected from (CH 2 )i. 3 -cycloalkyl, (CH 2 )o- 3 -polycycloalkyl, heterocycloalkyl a , (CH 2 )i- 3 -heterocycloalkyl b , (CH 2 ) 0-3 -R22, (C(R2O)(R21))-(CH 2 ) o.3 -phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )i. 3 -O-(CH 2 )-phenyl;

R20 and R21 are independently selected from H, alkyl and CF 3 , or R20 and R21 together with the carbon atom to which they are attached form a carbon-containing 3-, 4-, 5- or 6- membered saturated ring, optionally containing an O ring member, wherein R20 and R21 are not both H;

R22 is a fused 6,5- or 6,6- bicyclic ring containing an aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkyl, alkoxy, OH, CN, CF 3 , halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6bicyclic ring is not aryl substituted with ethylenedioxy;

Y is selected from -O- and -N(R12)-;

B is selected from:

(i) heteroaryl a ; and

(ii) aryl;

AW- is selected from:

-(CH 2 )O-6-(CHR15)-(CH 2 ) 0-6 -A, -(CHR12)-A, -O-(CHR12)-A, -(CH 2 ) O.6 -A, -(CH 2 ) 0-6 -O-(CH 2 ) 0-6 -A, -(CH 2 ) 0-6 -NH-(CH 2 ) 0-6 -A, -(CH 2 )O-6-N(R12)-(CH 2 )I. 6 -C(=0)-A, -(CH 2 ) 0-6 -NH-C(=O)-(CH 2 ) 0-6 -A,

-C(=0)N(R12)-(CH 2 ) 0-6 -A, -(CH 2 )O-6-C(=0)-(CH 2 ) 0-6 -A, -(CH 2 ) 0-6 -(phenyl)-(CH 2 ) 0-6 -A, -NH-SO 2 -A and -SO 2 -NH-A;

A is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN, and phenyl; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system; wherein when:

(i) V is -O-, or

(ii) Z is -O-,

A is A', wherein A' is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, wherein the ring system is substituted with phenyl and optionally, where possible, 1, 2, or 3 further substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN, and phenyl; wherein when A' is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A' is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system; alkoxy is OCF3, a linear O-linked hydrocarbon of between 1 and 6 carbon atoms (C1-C6), or a branched O-linked hydrocarbon of between 3 and 6 carbon atoms (C3-C6); alkoxy may optionally be substituted, where possible, with 1 or 2 substituents independently selected from OH, CN, CF3, and fluoro; alkyl is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, -NR13R14, -C(=O)OR13, -C(=O)NR13R14, CN, CF 3 , halo; alkyl b is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl b may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, CN, CF3, halo; small alkyl is a linear saturated hydrocarbon having up to 4 carbon atoms (C1-C4) or a branched saturated hydrocarbon of between 3 and 4 carbon atoms (C3-C4); small alkyl may optionally be substituted with 1 or 2 substituents independently selected from alkoxy, OH, NR13R14, C(=O)OR13, C(=O)NR13R14, CN, CF 3 , halo; aryl is phenyl, biphenyl or naphthyl; aryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, methylenedioxy, ethylenedioxy, OH, halo, CN, -(CH2)0-3- O-heteroaryl a , aryl b , -O-aryl b , -(CH2)1-3-aryl b , -(CH2)0-3-heteroaryl a , -C(=O)OR13, -C(=O)NR13R14, -(CH 2 )0-3-NR13R14, OCF 3 and CF 3 ; a ry I b is phenyl, biphenyl or naphthyl; a ryl b may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl b , alkoxy, OH, halo, CN, and CF3; cycloalkyl is a monocyclic saturated hydrocarbon ring of between 3 and 6 carbon atoms (C 3 -C 6 ); cycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo; halo is F, Cl, Br, or I; heteroaryl is a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members that are selected from N, NR8, S, and O; heteroaryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3; heteroaryl a is a 5-, 6-, 9- or 10- membered mono- or bi-cyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O; heteroaryl a may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF 3 , halo, CN, aryl b , -(CH 2 )o- 3 -NR13R14, heteroaryl”, -C(=O)OR12, -C(=O)NR13R14 and CF 3 ; heteroaryl b is a 5-, 6-, 9- or 10- membered mono- or bi-cyclic aromatic ring, containing, where possible, 1, 2 or 3 ring members independently selected from N, N(R12), S and O; wherein heteroaryl b may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl b , alkoxy, OH, halo, CN, aryl b , -(CH2)1-3-aryl b , and CF3; heterocycloalkyl is a non-aromatic carbon-containing monocyclic ring containing 3, 4, 5, or 6, ring members, wherein at least one ring member is independently selected from N, N(R12), S, and O; heterocycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo; heterocycloalkyl a is a 3-10 membered non-aromatic carbon-containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O; heterocycloalkyl a may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, aryl, halo, OH, CN, CF3, and heterocycloalkyl; wherein when heterocycloalkyl a is a bicyclic ring system, the bicyclic ring system is spiro; heterocycloalkyl b is a 3-6 membered non-aromatic carbon-containing monocyclic ring system which may be saturated or, where possible, unsaturated with 1 or 2 double bonds; heterocycloalkyl b contains one or two ring members independently selected from N, N(C(=O)CH3) and O; heterocycloalkyl b may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, halo, OH, CN, CF3, phenyl and oxo; wherein the point of attachment of heterocycloalkyl b is not at a ring nitrogen; polycycloalkyl is a bi- or tri-cyclic saturated hydrocarbon ring system of between 5 and 10 carbon atoms (C5-C10), wherein the bi- or tri-cyclic ring system is bridged or spiro;

R8 is independently selected from H, alkyl, cycloalkyl, and heterocycloalkyl;

R12 is independently selected from H, alkyl, and cycloalkyl;

R13 and R14 are independently selected from H, alkyl b , aryl b and heteroaryl b or R13 and R14 together with the nitrogen atom to which they are attached form a carbon-containing 4-, 5-, 6- or 7-membered heterocyclic ring, optionally containing an additional heteroatom selected from N, N(R12), S, SO, SO2, and O, which may be saturated or unsaturated with 1 or 2 double bonds and which may be optionally mono- or di-substituted with substituents selected from oxo, alkyl b , alkoxy, OH, halo and CF3;

R15 is selected from alkyl, halo, CF 3 , CN, OH, alkoxy, NR13R14, and CONR13R14;

R16 and R17 are independently selected from H and small alkyl; and tautomers, isomers, stereoisomers (including enantiomers, diastereoisomers and racemic and scalemic mixtures thereof), deuterated analogues, and pharmaceutically acceptable salts and/or solvates thereof. The compounds of formula (I) have been developed to be inhibitors of FXIIa, which as noted above, has a unique and specific binding site. The compounds of formula (I) aim to address the need for small molecule FXIIa inhibitors, particularly with pharmacokinetic properties suitable for use as oral therapies. The compounds of formula (I) can also avoid including groups associated with covalent binding properties e.g. groups with acylating reactivity such as acylated aminotriazoles, and thus can provide compounds that are reversible inhibitors, to further reduce the risk of off-target effects and cytotoxicity.

The present invention also provides a prodrug of a compound as herein defined, or a pharmaceutically acceptable salt and/or solvate thereof.

The present invention also provides an N-oxide of a compound as herein defined, or a prodrug or pharmaceutically acceptable salt and/or solvate thereof.

It will be understood that "methylenedioxy" refers to -O-CH2-O- only, wherein "methylenedioxy" cannot be substituted.

It will be understood that "ethylenedioxy" refers to -O-CH2-CH2-O- only, wherein "ethylenedioxy" cannot be substituted.

It will be understood that "aryl substituted with methylenedioxy" refers to aryl as defined above, wherein aryl is substituted with -O-CH2-O-. For example, .

It will be understood that "aryl substituted with ethylenedioxy" refers to aryl as defined above, wherein aryl is substituted with -O-CH2-CH2-O-. For example,

It will be understood that, as noted above R22 is not aryl substituted with methylenedioxy, which as mentioned above excludes ; and R22 is not aryl substituted with ethylenedioxy which as mentioned above excludes R22 can include fused 6,5- or 6,6- bicyclic rings containing an aryl ring fused to a non-aromatic ring, wherein the non-aromatic ring contains two O ring members, positioned as for but wherein the non-aromatic ring is substituted as for

R22. For example, R22 can be

It will be understand that A and A', as defined above, contain at least one N ring member. Other than optional non-carbon ring members, it will be understood that the remaining ring members are carbon.

It will be understood that "pharmaceutically acceptable salts and/or solvates thereof" means "pharmaceutically acceptable salts thereof", "pharmaceutically acceptable solvates thereof", and "pharmaceutically acceptable solvates of salts thereof".

The compounds of the present invention can be provided as mixtures of more than one stereoisomer. When provided as a mixture of stereoisomers, one stereoisomer can be present at a purity >90 % relative to the remaining stereoisomers. More specifically, when provided as a mixture of stereoisomers, one stereoisomer can be present at a purity >95 % relative to the remaining stereoisomers.

It will be understood that substituents may be named as its free unbonded structure (e.g. piperidine) or by its bonded structure (e.g. piperidinyl). No difference is intended.

It will be understood that the compounds of the invention comprise several substituents. When any of these substituents is defined more specifically herein, the substituents/optional substituents to these groups described above also apply, unless stated otherwise. For example, B can be heteroaryl a , which more specifically can be isoquinolinyl. In this case, isoquinolinyl can be optionally substituted in the same manner as "heteroaryl a ".

It will be understood that the term "where possible" means that the group, atom, or substituent in question may be present if it is chemically possible to do so, e.g. does not exceed the valencies of chemically stable compounds. It will be understood that a fused ring system refers to a ring system where two rings in the ring system share two adjacent atoms (i.e., one common covalent bond). For example, is a fused ring system (specifically a fused bicyclic ring system) which can be considered as a benzene ring and a piperidine ring sharing a common bond.

It will be understood that a bridged ring system refers to a ring system having two rings sharing three or more atoms. For example, is a bridged ring system (specifically a bridged bicyclic ring system) which can be considered as a cyclopentane ring and a second cyclopentane ring joined at a bridge and sharing three common atoms.

It will be understood that a spiro ring system refers to a ring system where two rings in the ring system share one common atom. For example, is a spiro ring system (specifically a spiro bicyclic ring system) which can be considered as a cyclobutane ring and an azetidine ring sharing a common carbon atom.

It will be understood that the ring system A, as defined in formula (I), can be fully saturated, or have any degree of unsaturation. For example, the ring system can be fully saturated, partially unsaturated, aromatic, non-aromatic, or have an aromatic ring bridged, fused or spiro to a non-aromatic ring.

It will be understood that ring system A can contain non-carbon ring members, and that these non-carbon ring members can, where possible, be optionally substituted themselves (as well, or as opposed to the carbon ring members), with the optional substituents included in the definition of A.

It will be understood that when any variable (e.g. alkyl) occurs more than once, its definition on each occurrence is independent of every other occurrence.

It will be understood that combinations of substituents and variables are permissible only if such combinations result in stable compounds. As used herein the term "bradykinin-mediated angioedema" means hereditary angioedema, and any non- hereditary bradykinin-mediated angioedema. For example, "bradykinin-mediated angioedema" encompasses hereditary angioedema and acute bradykinin-mediated angioedema of unknown origin.

As used herein, the term "hereditary angioedema" means any bradykinin-mediated angioedema caused by an inherited genetic dysfunction, fault, or mutation. As a result, the term "HAE" includes at least HAE type 1, HAE type 2, and normal Cl inhibitor HAE (normal Cl-lnh HAE).

More specifically, the invention provides compounds of formula (I) wherein the compound is a compound of formula (la), formula (lb), or formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

The invention provides compounds of formula (I) wherein the compound is a compound of formula (la), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

The invention provides compounds of formula (I) wherein the compound is a compound of formula (lb), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

The invention provides compounds of formula (I) wherein the compound is a compound of formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

As noted above, B can be selected from:

(i) heteroaryl a ; and

(ii) aryl.

B can be aryl; or B can be a 6-membered monocyclic or 9- or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

B can be aryl; or B can be 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a . B can be aryl; or B can be 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

B can be phenyl substituted as for aryl; or B can be a 6-membered monocyclic or 9- or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

B can be phenyl substituted as for aryl; or B can be a 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

B can be phenyl substituted as for aryl; or B can be a 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

B can be phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl; or B can be a 6- membered monocyclic or 9- or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

B can be phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl; or B can be a 6- membered monocyclic or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

B can be phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl; or B can be a 6- membered monocyclic or 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

B can be phenyl optionally substituted as for aryl, or B can be pyridyl or isoquinolinyl, optionally substituted as for heteroaryl a .

B can be phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl, or B can be pyridyl or isoquinolinyl, optionally substituted as for heteroaryl a . or optionally substituted as for heteroaryl a , or B can be phenyl optionally substituted as for aryl. B can be optionally substituted as for heteroaryl a . B can be optionally substituted as for heteroaryl a . B can be phenyl optionally substituted as for aryl.

B can be or optionally substituted as for heteroaryl a , or B can be phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl. B can be phenyl substituted with

CN and optionally 1 or 2 further substituents as for aryl.

B can be unsubstituted; or B can be aryl, wherein B can be substituted with CN and optionally 1 or 2 further substituents as for aryl; or B can be heteroaryl a , wherein B can be substituted with alkyl, halo, OH, or NH2, and optionally 1 or 2 further substituents as for heteroaryl a .

B can be selected from optionally further substituted with 1 or 2 substituents as for heteroaryl a ; or each optionally further substituted with 1 or 2 substituents as for heteroaryl a ; with no substituents; or no further substituents. B can be , optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a .

B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be

. B can be

B can be selected from the following groups: optionally further substituted with 1 or 2 substituents as for heteroaryl a ; or B can be with no substituents; or B can be with no further substituents. B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be , optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a . B can be with no substituents. B can be with no further substituents.

Preferably, B is heteroaryl a .

B can be heteroaryl a and Y can be attached to B at a carbon atom on the heteroaryl a ring.

B can be heteroaryl a and Y can be attached to B at a carbon atom on the heteroaryl a ring, and the two ring atoms adjacent to the carbon atom on the heteroaryl a ring to which Y attaches are both carbon.

It will be understood that, in the instance when Y is attached to B at a carbon atom on the heteroaryl a ring, the attachment of Y to B can be at any carbon on the heteroaryl a ring, so long as the remainder of the ring is still a heteroaryl ring.

It will be understood that, in the instance when Y is attached to B at a carbon atom on the heteroaryl a ring, and the two ring atoms adjacent to the carbon atom on the heteroaryl a ring to which Y attaches are both carbon, these adjacent ring atoms can be, where possible, substituted or unsubstituted as defined in the embodiment or claim. Further, for example, if B is isoquinolinyl, the attachment to Y can be at any of the following ring atoms:

When B is heteroaryl a , B can be a 9 or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a . B can be a 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a . B can be a 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

When B is heteroaryl a , B is preferably isoquinolinyl, optionally substituted as for heteroaryl a .

When B is heteroaryl a , B is preferably isoquinolinyl, wherein Y is attached to B at a carbon atom on the heteroaryl a ring, and the two ring atoms adjacent to the carbon atom on the heteroaryl a ring to which Y attaches are both carbon.

When B is isoquinolinyl, B is preferably optionally substituted as for heteroaryl a .

When B is heteroaryl a , B is preferably substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a .

When B is isoquinolinyl, B is preferably substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a . When B is isoquinolinyl, B can be substituted with NH 2 at the 1- position and optionally further substituted with 1 or 2 substituents as for heteroaryl a .

When B is isoquinolinyl, B can be optionally further substituted with 1 or 2 substituents as for heteroaryl a .

Preferably, B is

Y can be as defined above. Preferably, Y is -N(R12)-. Preferably, Y is -NH- or -N(CH3)-. Yet more preferably,

Y is -NH-.

Preferably, B is heteroaryl a and Y is -NH-.

Preferably, B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; and Y is -NH-.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; and Y is -NH-.

Preferably, B is and Y is -NH-.

AW- can be as defined above. AW- can be selected from

-(CH 2 )0-3-(CHR15)-(CH 2 )0-3-A, -(CHR12)-A, -O-(CHR12)-A, -(CH 2 ) O .3-A, -(CH 2 )0-3-0-(CH 2 )O.3-A,

-(CH 2 )0-3-NH-(CH 2 )0-3-A, -(CH 2 )0-3-N(R12)-(CH 2 )I.3-C(=0)-A, -(CH 2 )0-3-NH-C(=0)-(CH 2 )0-3-A,

-C(=0)N(R12)-(CH 2 )0-3-A, -(CH 2 )0-3-C(=0)-(CH 2 )0-3-A, -(CH 2 ) 0 -3-(phenyl)-(CH 2 ) 0 -3-A, -NH-SO 2 -A and -SO 2 -NH-A. AW- can be selected from -(CH 2 )0-3-A, and -(CH2)0-3-0-(CH2)0-3-A.

AW- can be selected from -(CH2)0-1-A, and -(CH2)0-1-O-A.

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A.

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; and Y is -NH-.

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; and B is heteroaryl a .

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; and B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a .

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; and B is isoquinolinyl, optionally substituted as for heteroaryl a .

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; and B is

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; B is heteroaryl a ; and Y is -NH-.

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; and Y is -NH-.

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; B is isoquinolinyl, optionally substituted as for heteroaryl a ; and Y is -NH-.

Preferably, AW- is selected from -(CH2)0-1-A, and -(CH2)-O-A; B is and Y is -NH-. The invention provides compounds of formula (I), wherein

V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-.

More specifically, the invention provides compounds of formula (I) wherein the compound is a compound of formula (la) or formula (lb), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

R16 can be H. R16 and R17 can both be H.

Preferably, R16 and R17 are both H; and B is heteroaryl a .

Preferably, R16 and R17 are both H; and B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a .

Preferably, R16 and R17 are both H; and B is isoquinolinyl, optionally substituted as for heteroaryl a .

Preferably, R16 and R17 are both H; and B is

Preferably, R16 and R17 are both H; and Y is -NH-. Preferably, R16 and R17 are both H; B is heteroaryl a ; and Y is -NH-.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; and Y is -NH-.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; and Y is - NH-.

Preferably, R16 and R17 are both H; B is ; and Y is -NH-.

R18 or R23 can be selected from C(=S)NH-aryl; C(=S)NH-R19; C(=S)N(alkyl)R19; C(=S)NR13R14; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one substituent selected from cycloalkyl and heterocycloalkyl b , and optionally one or two further substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3; wherein R13 and R14 together with the nitrogen atom to which they are attached form a carbon- containing 4-, 5-, 6- or 7-membered heterocyclic ring, optionally containing an additional heteroatom selected from N, N(R12), S, SO, SO 2 , and O, which may be saturated or unsaturated with 1 or 2 double bonds and which may be optionally mono- or di-substituted with substituents selected from oxo, alkyl b , alkoxy, OH, halo and CF3.

R18 or R23 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic one cycloalkyl substituent;

R18 or R23 can be selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 , heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

Preferably, R18 or R23 can be selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19);

C(=O)NH2; heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

Yet more preferably, R18 or R23 can be selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- or 6- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

Yet more preferably, R18 or R23 can be selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

Yet more preferably, R18 or R23 can be selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituent;

Yet more preferably, R18 or R23 can be selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing two N ring members substituted with one substituent; and

R18 can be selected from:

R18 can be selected from:

R18 can be selected from:

R18 can be selected from:

Preferably, R18 is selected from:

Preferably, R18 is selected from:

The compound of the invention can be a compound of formula (la), wherein R18 is as described as above, or a compound of formula (lb) and R23 is C(=O)R19.

The compound of the invention can be a compound of formula (la), wherein R18 is as described as above, or a compound of formula (lb) and R23 is C(=O)(CH2)1-3cycloalkyl.

The compound of the invention can be a compound of formula (la), wherein R18 is as described above, or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

The compound of the invention can be a compound of formula (la), wherein R18 is as described above, or a compound of formula (lb) and R23 is selected from

Preferably, R16 and R17 are both H; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, B is heteroaryl a ; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl. Preferably, B is and the compound of the invention is a compound formula (la), wherein

R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, B is heteroaryl a ; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl. preferably, B is Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl. Preferably, R16 and R17 are both H; B is heteroaryl a ; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with

1 or 2 further substituents as for heteroaryl a ; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon- containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both H; B is and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19);

C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; and ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both H; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl. Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with

1 or 2 further substituents as for heteroaryl a ; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; and the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

Preferably, R16 and R17 are both the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19;

C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

R19 can be selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22,

(C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, R19 can be selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, and

(CH2)3-O-(CH 2 )-phenyl.

Heterocycloalkyl a can be a 4-10 membered non-aromatic carbon-containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O; heterocycloalkyl a may optionally be substituted with 1 or 2 substituents independently selected from alkyl, aryl, halo, and heterocycloalkyl; wherein when heterocycloalkyl a is a bicyclic ring system, the bicyclic ring system is spiro. Heterocycloalkyl a can be selected from:

Heterocycloalkyl a can be selected from:

Heterocycloalkyl b can be a 4-6 membered non-aromatic carbon-containing monocyclic ring system which may be saturated or, where possible, unsaturated with 1 or 2 double bonds; heterocycloalkyl b contains one or two ring members independently selected from N, N(C(=O)CH3) and O; heterocycloalkyl b may optionally be substituted with 1 or 2 substituents independently selected from alkyl, phenyl and oxo; wherein the point of attachment of heterocycloalkyl b is not at a ring nitrogen.

Heterocycloalkyl b can be selected from:

Heterocycloalkyl b can be selected from:

Polycycloalkyl can be selected from:

R22 can be a fused 6,5- or 6,6- bicyclic ring containing an aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkoxy, halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6- bicyclic ring is not aryl substituted with ethylenedioxy.

R22 can be a fused 6,5- or 6,6- bicyclic ring containing a 6-membered aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkoxy, halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6- bicyclic ring is not aryl substituted with ethylenedioxy. R22 can be selected from:

Preferably, R22 is selected from:

R20 and R21 together with the carbon atom to which they are attached can form a carbon-containing 3-, 4- or 6-membered saturated ring, optionally containing an O ring member, wherein R20 and R21 are not both H.

Alternatively, R20 or R21 can be independently selected from H, (Ci-sjalkyl and CF3, wherein R20 and R21 are not both H.

More specifically, R20 or R21 can be independently selected from H, methyl and CF3, wherein R20 and R21 are not both H.

Preferably, R16 and R17 are both H; and R19 is selected from (CH 2 h-z-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH2)0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl. Preferably, Y is -NH-; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O- phenyl, and (CH2)3-O-(CH2)-phenyl.

Preferably, B is heteroaryl a ; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH2)0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a ; Y is -NH-; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1- polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl. preferably, selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a ; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 ) O.2 -R22, (C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and

(CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both selected from (CH 2 )1-2-cycloalkyl,

(CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; Y is -NH-; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 ) 0.2 -R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22,

(C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both selected from (CH 2 )I. 2 - cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl. Preferably, R16 and R17 are both H; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

0 v . ... TH n

° ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

0 v A L.

X LM if

0 ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl. preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic tuent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon- containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with

1 or 2 further substituents as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; and ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH- aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl.

Preferably, R16 and R17 are both the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; and R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; AW- can be -(CH 2 )-O-A.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; AW- can be -(CH 2 )-O-A.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; A can be a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , and CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; A can be a 6 to 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; A can be a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; A can be substituted, where possible, with 1, 2 or 3 substituents independently selected from alkyl and oxo.

When V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-; A can be selected from:

Preferably, R16 and R17 are both H; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a ; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22,

(C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a ; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-0-A. Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A. Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A. Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-0-A.

Preferably, R16 and R17 are both membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-0-A.

Preferably, R16 and R17 are both H; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A. Preferably, Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted one cycloalkyl substituent;

0 v . ... TH n

° ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and

AW- is -(CH 2 )-O-A.

Preferably, B is heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A. Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

0 v A L

X LM if

° ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A. Preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

0 A JL 'h f if c ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14,

NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and

AW- is -(CH 2 )-O-A. preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring substituted with one cycloalkyl substituent; and 'YHT ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14,

NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and

AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19);

C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; and ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon- containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

NH, , 1 ' QQ

Preferably, R16 and R17 are both H; B is ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=0)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19); C(=0)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with

1 or 2 further substituents as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19; C(=0)N(alkyl)(R19);

C(=0)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A. Preferably, R16 and R17 are both H; B is ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=0)R19; C(=0)NHR19;

C(=0)N(alkyl)(R19); C(=0)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-0-A.

Preferably, Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22,

(C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=0)R12, C(=0)0R13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-0-A. Preferably, B is heteroaryl a ; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH2)0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a substituted with NH 2 , and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH2)0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

Preferably, B is R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a ; Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a substituted with NH2, optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2) 3 -O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A. Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and

Preferably, B is Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl,

(C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A. Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )O-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and

(CH2)3-O-(CH2)-phenyl; A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is R19 is selected from (CH2)1-2-cycloalkyl,

(CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1- polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22,

(C(R20)(R21))-O-phenyl, and (CH2)3-O-(CH2)-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 ) 0-2 -heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is ; Y is -NH-; R19 is selected from (CH 2 )1-2-cycloalkyl,

(CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A. Preferably, R16 and R17 are both H; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; . and or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from

(CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19;

C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; and ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13,

C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, B is the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from

(CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring substituted with one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A. preferably, B is Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; and or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring substituted with one cycloalkyl substituent; and ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A. Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon- containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19);

C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; and or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from

(CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic one cycloalkyl substituent; or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R20)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from

(CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with

1 or 2 further substituents as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon- containing aromatic ring is substituted with one cycloalkyl substituent; and ; or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl; R19 is selected from (CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A. Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from

C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; and or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from

(CH 2 )1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22,

(C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 ) 3 -O-(CH 2 )-phenyl; and

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13,

C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH 2 )-O-A.

Preferably, R16 and R17 are both H; B is Y is -NH-; the compound of the invention is a compound formula (la), wherein R18 can be selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19;

C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent; and or a compound of formula (lb) and R23 is C(=O)(CH 2 )1-2cycloalkyl; R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH2)1-2-heterocycloalkyl b , (CH2)0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl; and A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF3, CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused; and AW- is -(CH2)-O-A.

The invention also provides compounds wherein A is A'; wherein

V is -O-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -O-.

More specifically, the invention also provides compounds wherein A is A', wherein the compound is a compound of formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

When V is -O-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or, V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is -O-; wherein A is A'; R16 can be H. Preferably, R16 and R17 can both be H.

When A is A', A' can be a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl and optionally, where possible, 1, 2, or 3 further substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN, and phenyl.

Preferably, A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl. More preferably, A' is selected from:

More preferably, A' is selected from:

When AW- is A'W-, A'W- can be -(CH 2 )0-3-A'.

More specifically, A'W- can be -(CH2)0-1-A'.

Preferably, A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, B is heteroaryl a ; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl. Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; A'W- can be -(CH 2 )0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, B is A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, B is heteroaryl a ; Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl. preferably, B is Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl. Preferably, R16 and R17 are both H; B is heteroaryl a ; A'W- can be -(CH 2 )0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; B is A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a ; Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5- membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; B is heteroaryl a substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a ; Y is -NH-; A'W- can be -(CH2)0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl. Preferably, R16 and R17 are both H; B is isoquinolinyl, optionally substituted as for heteroaryl a ; Y is -NH-; and A'W- can be -(CH 2 )0-1-A'; and A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

Preferably, R16 and R17 are both H; B is Y is -NH-; A'W- can be -(CH 2 )0-1-A'; and A' is a 5- membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

More specifically, the invention provides compounds of formula (I), selected from Table 1, Table 2, Table 3, Table 4, and Table 5, or a pharmaceutically acceptable salt, solvate, or solvate of a salt thereof.

More specifically, the invention provides compounds of formula (I), selected from Table 1; Table 2; compounds 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, and 92 of Table 3; compounds 93, 94, 95, 96, 97, 98, 99, and 101 of Table 4; and Table 5, or a pharmaceutically acceptable salt, solvate, or solvate of a salt thereof.

For the compounds provided in Table 1, Table 2, Table 3, Table 4, and Table 5 below, where stereochemistry is indicated, the compound is intended to cover all possible stereoisomers thereof.

The present invention therefore provides the compounds below in Table 1, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of the compounds below in Table 1, and pharmaceutically acceptable salts and/or solvates thereof.

The present invention therefore provides the compounds below in Table 2, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of the compounds below in Table 2, and pharmaceutically acceptable salts and/or solvates thereof.

The present invention therefore provides the compounds below in Table 3, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of the compounds below in Table 3, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore provides compounds 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, and 92 below in Table 3, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of compounds 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, and 92 below in Table 3, and pharmaceutically acceptable salts and/or solvates thereof.

The present invention therefore provides the compounds below in Table 4, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of the compounds below in Table 4, and pharmaceutically acceptable salts and/or solvates thereof.

The present invention therefore provides compounds 93, 94, 95, 96, 97, 98, 99, and 101 below in Table 4, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of compounds 93, 94, 95, 96, 97, 98, 99, and 101 below in Table 4, and pharmaceutically acceptable salts and/or solvates thereof.

The present invention therefore provides the compounds below in Table 5, and pharmaceutically acceptable salts and/or solvates thereof. The present invention therefore also provides stereoisomers of the compounds below in Table 5, and pharmaceutically acceptable salts and/or solvates thereof.

It will be understood that, when reading the compounds in Table 1, Table 2, Table 3, Table 4, and Table 5 below, the substituents are to be read from left to right. For example, example compound 93 in Table 4 has a Q1 group: and a Q.2 group "OCH2". Therefore, the Qi group is attached to the

O" of the "OCH2" of the Q.2 group, as follows: Table 1 Table 2 

Table 3 Table 4

Table 5

Preferably, the compound of formula (I) is a compound selected from example numbers: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 , 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 57, 59, 60, 61, 62, 63, 64, 65, 66, 69, 70, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 97, 102, 103, 104, or 105, and pharmaceutically acceptable salts and/or solvates thereof. Preferably, the compound of formula (I) is a compound selected from example numbers: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 , 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 57, 59, 60, 61, 62, 63, 64, 65, 66, 69, 70, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 97, 102, or 103, and pharmaceutically acceptable salts and/or solvates thereof. More preferably, the compound of formula (I) is a compound selected from example numbers: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 , 28, 29, 30, 31, 32, 33, 34, 35,

36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 57, 59, 60, 61, 62, 63, 64, 65, 66,

69, 70, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 97, 102,

104, or 105, and pharmaceutically acceptable salts and/or solvates thereof. More preferably, the compound of formula (I) is a compound selected from example numbers: 3, 4, 5, 6,

7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 , 28, 29, 30, 31, 32, 33, 34, 35,

36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 57, 59, 60, 61, 62, 63, 64, 65, 66,

69, 70, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 97, or

102, and pharmaceutically acceptable salts and/or solvates thereof.

Even more preferably, the compound of formula (I) is a compound selected from example numbers: 3, 5, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 38, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 57, 59, 60, 61, 62, 63, 64, 65, 66, 69, 70, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 97, 104, or 105, and pharmaceutically acceptable salts and/or solvates thereof.

Even more preferably, the compound of formula (I) is a compound selected from example numbers: 3, 5, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 38, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 57, 59, 60, 61, 62, 63, 64, 65, 66, 69, 70, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, or 97, and pharmaceutically acceptable salts and/or solvates thereof.

Yet more preferably, the compound of formula (I) is a compound selected from example numbers: 3, 15, 17, 30, 38, 43, 44, 45, 52, 57, 59, 60, 62, 65, 66, 69, 90, or 105, and pharmaceutically acceptable salts and/or solvates thereof.

Yet more preferably, the compound of formula (I) is a compound selected from example numbers: 3, 15, 17, 30, 38, 43, 44, 45, 52, 57, 59, 60, 62, 65, 66, 69, or 90, and pharmaceutically acceptable salts and/or solvates thereof. Preferably, the compound of formula (I) is a compound selected from example numbers 104 or 105, and pharmaceutically acceptable salts and/or solvates thereof.

Therapeutic Applications

As noted above, the compounds (or pharmaceutically acceptable salts and/or solvates thereof), and pharmaceutical compositions comprising the compounds (or pharmaceutically acceptable salts and/or solvates thereof) of the present invention are inhibitors of FXIIa. They are therefore useful in the treatment of disease conditions for which FXIIa is a causative factor. Accordingly, the present invention provides a compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof), or a pharmaceutical composition comprising a compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof), for use in medicine.

The present invention also provides for the use of a compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof), or a pharmaceutical composition comprising the compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof), in the manufacture of a medicament for the treatment or prevention of a disease or condition in which FXIIa activity is implicated.

The present invention also provides a method of treatment of a disease or condition in which FXIIa activity is implicated comprising administration to a subject in need thereof a therapeutically effective amount of a compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof), or a pharmaceutical composition comprising the compound of the invention (ora pharmaceutically acceptable salt and/or solvate thereof).

As discussed above, FXIIa can mediate the conversion of plasma kallikrein from plasma prekallikrein. Plasma kallikrein can then cause the cleavage of high molecular weight kininogen to generate bradykinin, which is a potent inflammatory hormone. Inhibiting FXIIa has the potential to inhibit (or even prevent) plasma kallikrein production. Thus, the disease or condition in which FXIIa activity is implicated can be a bradykinin-mediated angioedema.

The bradykinin-mediated angioedema can be non-hereditary. For example, the non-hereditary bradykinin-mediated angioedema can be selected from non-hereditary angioedema with normal Cl Inhibitor (AE-nCl Inh), which can be environmental, hormonal, or drug-induced; acquired angioedema; anaphylaxis associated angioedema; angiotensin converting enzyme (ACE or ace) inhibitor-induced angioedema; dipeptidyl peptidase-4 inhibitor-induced angioedema; and tPA-induced angioedema (tissue plasminogen activator-induced angioedema).

Alternatively, and preferably, the bradykinin-mediated angioedema can be hereditary angioedema (HAE), which is angioedema caused by an inherited dysfunction/fault/mutation. Types of HAE that can be treated with compounds according to the invention include HAE type 1, HAE type 2, and normal Cl inhibitor HAE (normal Cl Inh HAE).

The disease or condition in which FXIIa activity is implicated can be selected from vascular hyperpermeability, stroke including ischemic stroke and haemorrhagic accidents; retinal edema; diabetic retinopathy; impaired visual acuity; DME; retinal vein occlusion; and AMD. These conditions can also be bradykinin-mediated.

As discussed above, FXIIa can activate FXIa to cause a coagulation cascade. Thrombotic disorders are linked to this cascade. Thus, the disease or condition in which FXIIa activity is implicated can be a thrombotic disorder. More specifically, the thrombotic disorder can be thrombosis; thromboembolism caused by increased propensity of medical devices that come into contact with blood to clot blood; prothrombotic conditions such as disseminated intravascular coagulation (DIC), Venous thromboembolism (VTE), cancer associated thrombosis, complications caused by mechanical and bioprosthetic heart valves, complications caused by catheters, complications caused by ECMO, complications caused by LVAD, complications caused by dialysis, complications caused by CPB, sickle cell disease, joint arthroplasty, thrombosis induced to tPA, Paget-Schroetter syndrome and Budd-Chari syndrome; atherosclerosis; COVID-19; acute respiratory distress syndrome (ARDS); idiopathic pulmonary fibrosis (IPF); rheumatoid arthritis (RA); and cold-induced urticarial autoinflammatory syndrome.

Surfaces of medical devices that come into contact with blood can cause thrombosis. The compounds (or pharmaceutically acceptable salts and/or solvates thereof) and pharmaceutical compositions of the present invention can be coated on the surfaces of devices that come into contact with blood to mitigate the risk of the device causing thrombosis. For instance, they can lower the propensity these devices to clot blood and therefore cause thrombosis. Examples of devices that come into contact with blood include vascular grafts, stents, in dwelling catheters, external catheters, orthopedic prosthesis, cardiac prosthesis, and extracorporeal circulation systems.

Other disease conditions for which FXIIa is a causative factor include: neuroinflammation; neuroinflammatory/neurodegenerative disorders such as MS (multiple sclerosis); other neurodegenerative diseases such as Alzheimer's disease, epilepsy and migraine; sepsis; bacterial sepsis; inflammation; vascular hyperpermeability; and anaphylaxis.

Brown adipose tissue (BAT) thermogenic activity can be mediated by the kallikrein-kinin system, and impaired BAT activity is associated with obesity and insulin resistance. Inhibiting FXIIa has the potential to inhibit (or even prevent) BAT activity mediated by the kallikrein-kinin system. The compounds or pharmaceutically acceptable salts and/or solvates thereof) and pharmaceutical compositions of the invention can therefore treat disease conditions such as obesity and diabetes. Factor XII inhibition is further implicated in the treatment of disease conditions such as kidney disease, renal fibrosis, glomerulosclerosis, renal scarring, ischemia/reperfusion injury in native or transplant kidneys, and acute kidney injury.

Combination Therapy

The compounds of the present invention (or pharmaceutically acceptable salts and/or solvates thereof) may be administered in combination with other therapeutic agents. Suitable combination therapies include any compound of the present invention (or a pharmaceutically acceptable salt and/or solvate thereof) combined with one or more agents selected from agents that inhibit platelet-derived growth factor (PDGF), endothelial growth factor (VEGF), integrin alpha5betal, steroids, other agents that inhibit FXIIa and other inhibitors of inflammation.

Some specific examples of therapeutic agents that may be combined with the compounds of the present invention include those disclosed in EP2281885A1 and by S. Patel in Retina, 2009 Jun;29(6 Suppl):S45-8.

Other suitable combination therapies include a compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof) combined with one or more agents selected from agents that treat HAE (as defined generally herein), for example bradykinin B2 antagonists such icatibant (Firazyr®); plasma kallikrein inhibitors such as ecallantide (Kalbitor®), lanadelumab (Takhzyro®) and berotralstat (ORLADEYO™); or Cl esterase inhibitor such as Cinryze® and Haegarda® and Berinert® and Ruconest®.

Other suitable combination therapies include a compound of the invention (or a pharmaceutically acceptable salt and/or solvate thereof) combined with one or more agents selected from agents that are antithrombotics (as outlined above), for example other Factor Xlla inhibitors, thrombin receptor antagonists, thrombin inhibitors, factor Vila inhibitors, factor Xa inhibitors, factor Xia inhibitors, factor IXa inhibitors, adenosine diphosphate antiplatelet agents (e.g., P2Y12 antagonists), fibrinogen receptor antagonists (e.g. to treat or prevent unstable angina or to prevent reocclusion after angioplasty and restenosis) and aspirin) and platelet aggregation inhibitors.

When combination therapy is employed, the compounds of the present invention and said combination agents may exist in the same or different pharmaceutical compositions, and may be administered separately, sequentially or simultaneously.

The compounds of the present invention can be administered in combination with laser treatment of the retina. The combination of laser therapy with intravitreal injection of an inhibitor of VEGF for the treatment of diabetic macular edema is known (Elman M, Aiello L, Beck R, et al. "Randomized trial evaluating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema" Ophthalmology. 27 April 2010).

Definitions

As noted above, the term "alkyl" is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy,

OH, -NR13R14, -C(=O)OR13, -C(=O)NR13R14, CN, CF 3 , halo. As noted above "alkyl b " is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl b may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, CN, CF3, halo. Examples of such alkyl or alkyl b groups include, but are not limited, to C1 - methyl, C2 - ethyl, C3 - propyl and C4-n-butyl, C3 - iso-propyl, C4 - sec-butyl, C4 - iso- butyl, C4 - tert-butyl and C 5 - neo-pentyl, optionally substituted as noted above. More specifically, "alkyl" or "alkyl b " can be a linear saturated hydrocarbon having up to 6 carbon atoms (C1-C6) or a branched saturated hydrocarbon of between 3 and 6 carbon atoms (C3-C6), optionally substituted as noted above. Even more specifically, "alkyl" or "alkyl b " can be a linear saturated hydrocarbon having up to 4 carbon atoms (C1-C4) or a branched saturated hydrocarbon of between 3 and 4 carbon atoms (C3-C4), optionally substituted as noted above, which is herein called "small alkyl" or "small alkyl b ", respectively. Preferably, "alkyl" or "alkyl b " can be defined as a "small alkyl" or "small alkyl b ".

"Aryl" and "aryl b " are as defined above. Typically, "aryl" or "aryl b " will be optionally substituted with 1, 2 or 3 substituents. Optional substituents are selected from those stated above. Examples of suitable aryl or aryl b groups include phenyl, biphenyl and naphthyl (each optionally substituted as stated above). Preferably "aryl" is selected from phenyl, substituted phenyl (wherein said substituents are selected from those stated above) and naphthyl. Most preferably "aryl" is selected from phenyl and substituted phenyl (wherein said substituents are selected from those stated above).

As noted above, the term "cycloalkyl" is a monocyclic saturated hydrocarbon ring of between 3 and 6 carbon atoms (C3-C6); cycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo. Examples of suitable monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, optionally substituted as noted above. More specifically, "cycloalkyl" can be a monocyclic saturated hydrocarbon ring of between 3 and 5 carbon atoms, more specifically, between 3 and 4 carbon atoms, optionally substituted as noted above.

As noted above, the term "alkoxy" is -OCF3, a linear O-linked hydrocarbon of between 1 and 6 carbon atoms (C1-C6), or a branched O-linked hydrocarbon of between 3 and 6 carbon atoms (C3-C6); alkoxy may optionally be substituted, where possible, with 1 or 2 substituents independently selected from OH, CN, CF3, and fluoro. Examples of such alkoxy groups include, but are not limited to, C1 - methoxy, C2 - ethoxy, C3 - n-propoxy and C4 - n-butoxy for linear alkoxy, and C3 - iso-propoxy, and C4 - sec-butoxy and tert-butoxy for branched alkoxy, optionally substituted as noted above. More specifically, "alkoxy" can be linear groups of between 1 and 4 carbon atoms (C1-C4), more specifically, between 1 and 3 carbon atoms (C1- C3). More specifically, "alkoxy" can be branched groups of between 3 and 4 carbon atoms (C3-C4), optionally substituted as noted above.

"Alkoxy" can be defined as above. Specifically, "alkoxy" is a linear O-linked hydrocarbon of between 1 and 6 carbon atoms (C1-C6), or a branched O-linked hydrocarbon of between 3 and 6 carbon atoms (C3-C6); alkoxy may optionally be substituted with 1 or 2 substituents independently selected from OH, CN, CF3, and fluoro. Examples of such alkoxy groups include, but are not limited to, C1 - methoxy, C2 - ethoxy, C3 - n-propoxy and C4 - n-butoxy for linear alkoxy, and C3 - iso-propoxy, and C4 - sec-butoxy and tert-butoxy for branched alkoxy, optionally substituted as noted above. More specifically, "alkoxy" can be linear groups of between 1 and 4 carbon atoms (C1-C4), more specifically, between 1 and 3 carbon atoms (C1- C3). More specifically, "alkoxy" can be branched groups of between 3 and 4 carbon atoms (C3-C4), optionally substituted as noted above.

"Halo" can be selected from Cl, F, Br and I. More specifically, halo can be selected from Cl and F.

As noted above, "heteroaryl" is a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members that are selected from N, NR8, S, and O; heteroaryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3. For example, heteroaryl can be selected from thiophene, furan, pyrrole, pyrazole, imidazole, oxazole, isoxazole, thiazole, isothiazole, triazole, oxadiazole, thiadiazole, pyridine, pyridazine, pyrimidine, and pyrazine, optionally substituted as noted above.

"Heteroaryl a " and "heteroaryl b " are as defined above. Typically, "heteroaryl a " or "heteroaryl b " will be optionally substituted with 1, 2 or 3 substituents. Optional substituents are selected from those stated above. Examples of suitable heteroaryl a or heteroaryl b groups include thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolyl, benzimidazolyl, benzotriazolyl, quinolinyl, isoquinolinyl, 5-azathianaphthenyl, indolizinyl, isoindolyl, indazolyl, benzothiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1,8-napthyridinyl and phthalazinyl (optionally substituted as stated above). More specifically, "heteroaryl a " or "heteroaryl b " can be a 9- or 10- membered bi-cyclic ring as defined, and optionally substituted as stated above. Examples of suitable 9- or 10- membered heteroaryl a or heteroaryl b groups include indolyl, benzimidazolyl, benzotriazolyl, quinolinyl, isoquinolinyl, 5-azathianaphthenyl, indolizinyl, isoindolyl, indazolyl, benzothiazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, 1,8-napthyridinyl and phthalazinyl.

As noted above, "heterocycloalkyl" is a non-aromatic carbon-containing monocyclic ring containing 3, 4, 5, or 6, ring members, wherein at least one ring member is independently selected from N, N(R12), S, and O; heterocycloalkyl may be optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo. More specifically, "heterocycloalkyl" can be a non-aromatic carbon- containing monocyclic ring containing 3, 4, 5, or 6, ring members, wherein at least one ring member is independently selected from N(R12), and O; heterocycloalkyl may be optionally substituted with 1 or 2 substituents independently selected from alkyl alkoxy, OH, CN, CF3, halo.

As noted above, "polycycloalkyl" is a bi- or tri-cyclic saturated hydrocarbon ring system of between 5 and 10 carbon atoms (C5-C10), wherein the bi- or tri-cyclic ring system is bridged or spiro. Examples of such polycycloalkyls include, but are not limited to, adamantane, spiro[3.3]heptane, bicyclo[2.2.2]octane, bicyclo[2.2.1]heptane, and bicyclo[l.l.l]pentane.

As noted above, "heterocycloalkyl a " is a 3-10 membered non-aromatic carbon-containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O; heterocycloalkyl a may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, aryl, halo, OH, CN, CF3, and heterocycloalkyl; wherein when heterocycloalkyl a is a bicyclic ring system, the bicyclic ring system is spiro. For example, "heterocycloalkyl a " can be morpholine, tetrahydrofuran, piperidine, 2-azaspiro[3.3]heptane, and 2-azaspiro[4.4]nonane optionally substituted as noted above. More specifically, "heterocycloalkyl a " can be a 4-10 membered non-aromatic carbon- containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O. More specifically, heterocycloalkyl a may optionally be substituted with 1 or 2 substituents independently selected from alkyl, aryl, halo, and heterocycloalkyl. As noted above, "heterocycloalkyl b " is a 3-6 membered non-aromatic carbon-containing monocyclic ring system which may be saturated or, where possible, unsaturated with 1 or 2 double bonds; heterocycloalkyl b contains one or two ring members independently selected from N, N(C(=O)CH3) and O; heterocycloalkyl b may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, halo, OH, CN, CF3, phenyl and oxo; wherein the point of attachment of heterocycloalkyl b is not at a ring nitrogen. For example, "heterocycloalkyl b " can be tetrahydro-2H-pyran, tetrahydrofuran, or oxetane, optionally substituted as noted above. Further, for example, "heterocycloalkyl b " can be piperidine, wherein the point of attachment is not at the ring nitrogen. For example, if heterocycloalkyl b is piperidine, the attachment of Y to the rest of compound of formula (I) can be at any of the following ring atoms: refers to the point at which heterocycloalkyl b is attached to the rest of the compound of formula (I), it does not refer to the position of any optional substituents. "Heterocycloalkyl b " can be substituted at a ring nitrogen. For example, heterocycloalkyl b can be More specifically, "heterocycloalkyl b " is a

4-6 membered non-aromatic carbon-containing monocyclic ring system. More specifically, "heterocycloalkyl b " may optionally be substituted with 1 or 2 substituents independently selected from alkyl, phenyl and oxo.

The term "O-linked", such as in "O-linked hydrocarbon residue", means that the hydrocarbon residue is joined to the remainder of the molecule via an oxygen atom.

In groups such as -(CH2)0-6-A, denotes the point of attachment of the substituent group to the remainder of the molecule.

As is clear from the definitions above, and for the avoidance of any doubt, it will be understood that "Y" is defined above, and does not encompass Yttrium.

As is clear from the definitions above, and for the avoidance of any doubt, it will be understood that "B" is defined above, and does not encompass Boron. As is clear from the definitions above, and for the avoidance of any doubt, it will be understood that "W" is defined above, and does not encompass Tungsten.

As is clear from the definitions above, and for the avoidance of any doubt, it will be understood that "V" is defined above, and does not encompass Vanadium.

"Salt", as used herein (including "pharmaceutically acceptable salt") means a physiologically or toxicologically tolerable salt and includes, when appropriate, pharmaceutically acceptable base addition salts and pharmaceutically acceptable acid addition salts. For example (i) where a compound of the invention contains one or more acidic groups, for example carboxy groups, base addition salts (including pharmaceutically acceptable base addition salts) that can be formed include sodium, potassium, calcium, magnesium and ammonium salts, or salts with organic amines, such as, diethylamine, A/-methyl- glucamine, diethanolamine or amino acids (e.g. lysine) and the like; (ii) where a compound of the invention contains a basic group, such as an amino group, acid addition salts (including pharmaceutically acceptable acid addition salts) that can be formed include hydrochlorides, hydrobromides, sulfates, phosphates, acetates, citrates, lactates, tartrates, mesylates, succinates, oxalates, phosphates, esylates, tosylates, benzenesulfonates, naphthalenedisulphonates, maleates, adipates, fumarates, hippurates, camphorates, xinafoates, p-acetamidobenzoates, dihydroxybenzoates, hydroxynaphthoates, succinates, ascorbates, oleates, bisulfates, trifluoroacetates and the like.

Hemisalts of acids and bases can also be formed, for example, hemisulfate and hemicalcium salts.

For a review of suitable salts, see "Handbook of Pharmaceutical Salts: Properties, Selection and Use" by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).

"Prodrug" refers to a compound which is convertible in vivo by metabolic means (e.g. by hydrolysis, reduction or oxidation) to a compound of the invention. Suitable groups for forming prodrugs are described in 'The Practice of Medicinal Chemistry, 2 nd Ed. pp561-585 (2003) and in F. J. Leinweber, Drug Metab. Res., 1987, 18, 379.

The compounds of the invention can exist in both unsolvated and solvated forms. The term 'solvate 1 is used herein to describe a molecular complex comprising the compound of the invention and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term 'hydrate' is employed when the solvent is water. Where compounds of the invention exist in one or more geometric, optical, enantiomeric, diastereomeric and tautomeric forms, including but not limited to cis- and trans-forms, E- and Z-forms, R-, S- and meso- forms, keto-, and enol-forms. Unless otherwise stated a reference to a particular compound includes all such isomeric forms, including racemic and other mixtures thereof. Where appropriate such isomers can be separated from their mixtures by the application or adaptation of known methods (e.g. chromatographic techniques and recrystallisation techniques). Where appropriate such isomers can be prepared by the application or adaptation of known methods (e.g. asymmetric synthesis). For example, where compounds of the invention exist as a mixture of stereoisomers, one stereoisomer can be present at a purity of >90% relative to the remaining stereoisomers, or more specifically at a purity of >95% relative to the remaining stereoisomers, or yet more specifically at a purity of >99% relative to the remaining stereoisomers. For example, where compounds of the invention exists in enantiomeric forms, the compound can be >90% enantiomeric excess (ee), or more specifically >95% enantiomeric excess (ee), or yet more specifically, >99% ee.

Unless otherwise stated, the compounds of the invention include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds wherein hydrogen is replaced by deuterium or tritium, or wherein carbon is replaced by 13 C or 14 C, are within the scope of the present invention. Such compounds are useful, for example, as analytical tools or probes in biological assays.

Methods for the preparation of deuterated analogues will be readily apparent to those skilled in the art. For example, methods may include the use of deuterated starting materials in the synthesis of the compounds described herein. Deuterated starting materials will be readily available to the skilled person, from standard commercial sources. Methods for making deuterated analogues and deuterated starting materials may also include deuterium exchange. For example, deuterium exchange may be achieved by mixing the compounds with D2O.

In the context of the present invention, references herein to "treatment" include references to curative, palliative and prophylactic treatment. For instance, treatment includes preventing the symptoms of the disease conditions for which FXIIa is a causative factor.

Methods The compounds of the invention may be administered alone or in combination with one or more other compounds of the invention or in combination with one or more other drugs (or as any combination thereof). Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable excipients. The term 'excipient' is used herein to describe any ingredient other than the compound(s) of the invention which may impart either a functional (i.e., drug release rate controlling) and/or a non-functional (i.e., processing aid or diluent) characteristic to the formulations. The choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.

Compounds of the invention intended for pharmaceutical use may be administered as a solid or liquid, such as a tablet, capsule or solution. Pharmaceutical compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995).

Accordingly, the present invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier, diluent or excipient.

For the treatment of conditions such as retinal vascular permeability associated with diabetic retinopathy and diabetic macular edema, the compounds of the invention may be administered in a form suitable for injection into the ocular region of a patient, in particular, in a form suitable for intra-vitreal injection. It is envisaged that formulations suitable for such use will take the form of sterile solutions of a compound of the invention in a suitable aqueous vehicle. The compositions may be administered to the patient under the supervision of the attending physician.

The compounds of the invention may also be administered directly into the blood stream, into subcutaneous tissue, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.

Parenteral formulations are typically aqueous or oily solutions. Where the solution is aqueous, excipients such as sugars (including but not restricted to glucose, manitol, sorbitol, etc.), salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.

Parenteral formulations may include implants derived from degradable polymers such as polyesters (i.e., polylactic acid, polylactide, polylactide-co-glycolide, polycapro-lactone, polyhydroxybutyrate), polyorthoesters and polyanhydrides. These formulations may be administered via surgical incision into the subcutaneous tissue, muscular tissue or directly into specific organs.

The preparation of parenteral formulations under sterile conditions, for example, by lyophilisation, may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.

The solubility of compounds of the invention used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of co-solvents and/or solubility-enhancing agents such as surfactants, micelle structures and cyclodextrins.

The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual, or sublingual administration by which the compound enters the blood stream directly from the mouth.

Formulations suitable for oral administration include solid plugs, solid microparticulates, semi-solids and liquids (including multiple phases or dispersed systems). Exemplary formulations suitable for oral administration include tablets; soft or hard capsules containing multi- or nano-particulates, liquids, emulsions or powders; lozenges (including liquid-filled); chews; gels; fast dispersing dosage forms; films; ovules; sprays; and buccal/mucoadhesive patches.

Liquid (including multiple phases and dispersed systems) formulations include emulsions, solutions, syrups and elixirs. Such formulations may be presented as fillers in soft or hard capsules (made, for example, from gelatin or hydroxypropylmethylcellulose) and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.

The compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Liang and Chen, Expert Opinion in Therapeutic Patents, 2001, 11 (6), 981-986. The formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets, Vol. 1, by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).

For administration to human patients, the total daily dose of the compounds of the invention is typically in the range 0.1 mg and 10,000 mg, or between 1 mg and 5000 mg, or between 10 mg and 1000 mg depending, of course, on the mode of administration.

The total dose may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein. These dosages are based on an average human subject having a weight of about 60kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.

Numbered Embodiments:

1. A compound of Formula (I),

Formula (I), wherein:

V is selected from -O- and -N(R23)-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-;

V is selected from -CH2-C(R16)(R17)- and -C(R16)(R17)-, and Z is selected from -O- and -N(R18)-; wherein R18 or R23 are selected from C(=S)NH-aryl; C(=S)NH-R19; C(=S)N(alkyl)R19; C(=S)NR13R14; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one substituent selected from cycloalkyl and heterocycloalkyl b , and optionally one or two further substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3;

R19 is selected from (CH 2 )1-3-cycloalkyl, (CH 2 )0-3-polycycloalkyl, heterocycloalkyl a , (CH 2 )i- 3 -heterocycloalkyl b , (CH 2 )0-3-R22, (C(R2O)(R21))-(CH 2 ) o.3 -phenyl, (C(R20)(R21))-R22, (C(R20)(R21))-O-phenyl, and (CH 2 )i- 3 -O-(CH 2 )-phenyl;

R20 and R21 are independently selected from H, alkyl and CF3, or R20 and R21 together with the carbon atom to which they are attached form a carbon-containing 3-, 4-, 5- or 6- membered saturated ring, optionally containing an O ring member, wherein R20 and R21 are not both H;

R22 is a fused 6,5- or 6,6- bicyclic ring containing an aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6bicyclic ring is not aryl substituted with ethylenedioxy;

Y is selected from -O- and -N(R12)-;

B is selected from:

(i) heteroaryl a ; and

(ii) aryl;

AW- is selected from:

-(CH 2 )O-6-(CHR15)-(CH 2 ) 0-6 -A, -(CHR12)-A, -O-(CHR12)-A, -(CH 2 ) 0-6 -A, -(CH 2 ) 0-6 -O-(CH 2 ) 0-6 -A, -(CH 2 ) 0-6 -NH-(CH 2 ) 0-6 -A, -(CH 2 )O-6-N(R12)-(CH 2 )I. 6 -C(=0)-A, -(CH 2 ) 0-6 -NH-C(=O)-(CH 2 ) 0-6 -A,

-C(=0)N(R12)-(CH 2 ) 0-6 -A, -(CH 2 )O-6-C(=0)-(CH 2 ) 0-6 -A, -(CH 2 ) 0-6 -(phenyl)-(CH 2 ) 0-6 -A, -NH-SO 2 -A and -SO 2 -NH-A; A is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN, and phenyl; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system; wherein when:

(i) V is -O-, or

(ii) Z is -O-,

A is A', wherein A' is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, wherein the ring system is substituted with phenyl and optionally, where possible, 1, 2, or 3 further substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN, and phenyl; wherein when A' is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A' is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system; alkoxy is OCF 3 , a linear O-linked hydrocarbon of between 1 and 6 carbon atoms (C1-C6), or a branched O-linked hydrocarbon of between 3 and 6 carbon atoms (C3-C6); alkoxy may optionally be substituted, where possible, with 1 or 2 substituents independently selected from OH, CN, CF3, and fluoro; alkyl is a linear saturated hydrocarbon having up to 10 carbon atoms (C1-C10) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, -NR13R14, -C(=O)OR13, -C(=O)NR13R14, CN, CF 3 , halo; alkyl b is a linear saturated hydrocarbon having up to 10 carbon atoms (Ci-Cio) or a branched saturated hydrocarbon of between 3 and 10 carbon atoms (C3-C10); alkyl b may optionally be substituted with 1, 2 or 3 substituents independently selected from alkoxy, OH, CN, CF3, halo; small alkyl is a linear saturated hydrocarbon having up to 4 carbon atoms (C1-C4) or a branched saturated hydrocarbon of between 3 and 4 carbon atoms (C3-C4); small alkyl may optionally be substituted with 1 or 2 substituents independently selected from alkoxy, OH, NR13R14, C(=O)OR13, C(=O)NR13R14, CN, CF 3 , halo; aryl is phenyl, biphenyl or naphthyl; aryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, methylenedioxy, ethylenedioxy, OH, halo, CN, -(CH2)0-3- O-heteroaryl a , aryl b , -O-aryl b , -(CH2)1-3-aryl b , -(CH2)0-3-heteroaryl a , -C(=O)OR13, -C(=O)NR13R14, -(CH 2 )0-3-NR13R14, OCF 3 and CF 3 ; a ry I b is phenyl, biphenyl or naphthyl; a ryl b may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl b , alkoxy, OH, halo, CN, and CF3; cycloalkyl is a monocyclic saturated hydrocarbon ring of between 3 and 6 carbon atoms (C 3 -C 6 ); cycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo; halo is F, Cl, Br, or I; heteroaryl is a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members that are selected from N, NR8, S, and O; heteroaryl may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF3; heteroaryl a is a 5-, 6-, 9- or 10- membered mono- or bi-cyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O; heteroaryl a may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl, alkoxy, OH, OCF 3 , halo, CN, aryl b , -(CH 2 )o- 3 -NR13R14, heteroaryl”, -C(=O)OR12, -C(=O)NR13R14 and CF 3 ; heteroaryl b is a 5-, 6-, 9- or 10- membered mono- or bi-cyclic aromatic ring, containing, where possible, 1, 2 or 3 ring members independently selected from N, N(R12), S and O; wherein heteroaryl b may be optionally substituted with 1, 2 or 3 substituents independently selected from alkyl b , alkoxy, OH, halo, CN, aryl b , -(CH2)1-3-aryl b , and CF3; heterocycloalkyl is a non-aromatic carbon-containing monocyclic ring containing 3, 4, 5, or 6, ring members, wherein at least one ring member is independently selected from N, N(R12), S, and O; heterocycloalkyl may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, OH, CN, CF3, halo; heterocycloalkyl a is a 3-10 membered non-aromatic carbon-containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O; heterocycloalkyl a may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, aryl, halo, OH, CN, CF3, and heterocycloalkyl; wherein when heterocycloalkyl a is a bicyclic ring system, the bicyclic ring system is spiro; heterocycloalkyl b is a 3-6 membered non-aromatic carbon-containing monocyclic ring system which may be saturated or, where possible, unsaturated with 1 or 2 double bonds; heterocycloalkyl b contains one or two ring members independently selected from N, N(C(=O)CH3) and O; heterocycloalkyl b may optionally be substituted with 1 or 2 substituents independently selected from alkyl, alkoxy, halo, OH, CN, CF3, phenyl and oxo; wherein the point of attachment of heterocycloalkyl b is not at a ring nitrogen; polycycloalkyl is a bi- or tri-cyclic saturated hydrocarbon ring system of between 5 and 10 carbon atoms (C5-C10), wherein the bi- or tri-cyclic ring system is bridged or spiro;

R8 is independently selected from H, alkyl, cycloalkyl, and heterocycloalkyl;

R12 is independently selected from H, alkyl, and cycloalkyl;

R13 and R14 are independently selected from H, alkyl b , aryl b and heteroaryl b or R13 and R14 together with the nitrogen atom to which they are attached form a carbon-containing 4-, 5-, 6- or 7-membered heterocyclic ring, optionally containing an additional heteroatom selected from N, N(R12), S, SO, SO2, and O, which may be saturated or unsaturated with 1 or 2 double bonds and which may be optionally mono- or di-substituted with substituents selected from oxo, alkyl b , alkoxy, OH, halo and CF3; R15 is selected from alkyl, halo, CF 3 , CN, OH, alkoxy, NR13R14, and CONR13R14;

R16 and R17 are independently selected from H and small alkyl; and tautomers, isomers, stereoisomers (including enantiomers, diastereoisomers and racemic and scalemic mixtures thereof), deuterated analogues, and pharmaceutically acceptable salts and/or solvates thereof.

2. A compound of formula (I) according to numbered embodiment 1, wherein the compound is a compound of formula (la), formula (lb), or formula (Ic),

Formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

3. A compound according to numbered embodiment 1, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

V is -N(R23)-, and Z is selected from -C(R16)(R17)-CH 2 - and -C(R16)(R17)-; or, V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -N(R18)-.

4. A compound of formula (I) according to any of the preceding numbered embodiments, wherein the compound is a compound of formula (la) or formula (lb), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

5. A compound of formula (I) according to any of the preceding numbered embodiments, wherein the compound is a compound of formula (la), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

6. A compound according to numbered embodiment 3, 4 or 5, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A is a 4- to 15- membered mono-, bi-, or tri- cyclic ring system, containing one N ring member and optionally one, two or three further ring members independently selected from N, O and S, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , and CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro; wherein when A is a tricyclic ring system, each of the three rings in the tricyclic ring system is either fused, bridged or spiro to at least one of the other rings in the tricyclic ring system.

7. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-aryl; C(=S)NH-R19; C(=S)N(alkyl)R19;

C(=S)NR13R14; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; heterocycloalkyl b ; a 5- or

6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one substituent selected from cycloalkyl and heterocycloalkyl b , and optionally one or two further substituents independently selected from alkyl, alkoxy, OH, OCF3, halo, CN, and CF 3 ; and wherein R13 and R14 together with the nitrogen atom to which they are attached form a carbon- containing 4-, 5-, 6- or 7-membered heterocyclic ring, optionally containing an additional heteroatom selected from N, N(R12), S, SO, SO 2 , and O, which may be saturated or unsaturated with 1 or 2 double bonds and which may be optionally mono- or di-substituted with substituents selected from oxo, alkyl b , alkoxy, OH, halo and CF3. 8. A compound according to any of numbered embodiments 1 to 6, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 or R23 are selected from C(=S)NH-aryl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

9. A compound according to numbered embodiment 8, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2, heterocycloalkyl b ; a 5- or 6- membered carbon-containing aromatic ring containing one, two or three ring members independently selected from N and, where possible, O, wherein the carbon-containing aromatic ring is substituted with one cycloalkyl substituent;

10. A compound according to numbered embodiment 9, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=0)NH2; heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

11. A compound according to numbered embodiment 10, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- or 6- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

12. A compound according to numbered embodiment 11, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing one or two N ring members substituted with one cycloalkyl substituent;

13. A compound according to numbered embodiment 12, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH2; 5- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a

6- membered carbon-containing aromatic ring containing one or two N ring members substituted

14. A compound according numbered embodiment 13, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R18 or R23 are selected from C(=S)NH-phenyl; C(=O)R19; C(=O)NHR19; C(=O)N(alkyl)(R19); C(=O)NH 2 ; 5- membered heterocycloalkyl b ring, optionally substituted as for heterocycloalkyl b ; a 6- membered carbon-containing aromatic ring containing two N ring members substituted with one

15. A compound according to any of the preceding numbered embodiments or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is a compound of formula (la), wherein R18 is as described in any of the preceding numbered embodiments, or a compound of formula (lb) and R23 is C(=O)R19.

16. A compound according to numbered embodiment 15, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is a compound of formula (la), wherein R18 is as described in any of the preceding numbered embodiments, or a compound of formula (lb) and R23 is C(=O)(CH2)1-3Cycloalkyl.

17. A compound according to numbered embodiment 16, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is a compound of formula (la), wherein R18 is as described in any of the preceding numbered embodiments, or a compound of formula (lb) and R23 is C(=O)(CH2)1-2cycloalkyl.

18. A compound according to numbered embodiment 17, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is a compound of formula (la), wherein R18 is as described in any of the preceding numbered embodiments, or a compound of formula (lb) and R23 is selected from

19. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R16 is H.

20. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R16 and R17 are both H.

21. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein:

R19 is selected from (CH2)1-2-cycloalkyl, (CH2)0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22,

(C(R20)(R21))-O-phenyl, and (CH 2 )3-O-(CH 2 )-phenyl.

22. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein:

R19 is selected from (CH2)1-2-cycloalkyl, (CH 2 )0-1-polycycloalkyl, heterocycloalkyl a , (CH 2 )1-2-heterocycloalkyl b , (CH 2 )0-2-R22, (C(R2O)(R21))-(CH 2 )0-1-phenyl, (C(R20)(R21))-R22, and (CH 2 ) 3 -O-(CH 2 )-phenyl. 23. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein heterocycloalkyl a is a 4-10 membered non-aromatic carbon-containing mono- or bi- cyclic ring system containing one or two ring members independently selected from N and O; heterocycloalkyl a may optionally be substituted with 1 or 2 substituents independently selected from alkyl, aryl, halo, and heterocycloalkyl; wherein when heterocycloalkyl a is a bicyclic ring system, the bicyclic ring system is spiro.

24. A compound according to numbered embodiment 23, or a tautomer, isomer, stereoisomer

(including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein heterocycloalkyl a is selected from:

25. A compound according to numbered embodiment 24, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein heterocycloalkyl a is selected from:

26. A compound according to any of numbered embodiments 1 to 22, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein heterocycloalkyl b is a 4-6 membered non-aromatic carbon-containing monocyclic ring system which may be saturated or, where possible, unsaturated with 1 or 2 double bonds; heterocycloalkyl b contains one or two ring members independently selected from N, N(C(=O)CH3) and O; heterocycloalkyl b may optionally be substituted with 1 or 2 substituents independently selected from alkyl, phenyl and oxo; wherein the point of attachment of heterocycloalkyl b is not at a ring nitrogen. 27. A compound according to numbered embodiment 26, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein heterocycloalkyl b is selected from:

28. A compound according to numbered embodiment 27, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein heterocycloalkyl b is selected from:

29. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein polycycloalkyl is selected from:

30. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein:

R22 is a fused 6,5- or 6,6- bicyclic ring containing an aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkoxy, halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6- bicyclic ring is not aryl substituted with ethylenedioxy.

31. A compound according to numbered embodiment 30, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein:

R22 is a fused 6,5- or 6,6- bicyclic ring containing a 6-membered aromatic ring fused to a non-aromatic ring, wherein the bicyclic ring optionally contains one or two ring members selected from N and O, wherein the fused 6,5- or 6,6- bicyclic ring may be optionally substituted with one or two substituents independently selected from alkoxy, halo, and oxo, wherein the 6,5- bicyclic ring may be attached via the 6- or 5- membered ring, wherein the 6,5-bicyclic ring is not aryl substituted with methylenedioxy, and wherein the 6,6- bicyclic ring is not aryl substituted with ethylenedioxy.

32. A compound according to numbered embodiment 31, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R22 is selected from: 33. A compound according to numbered embodiment 31, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein R22 is selected from:

34. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R20 and R21 together with the carbon atom to which they are attached form a carbon-containing 3-, 4- or 6-membered saturated ring, optionally containing an O ring member, wherein R20 and R21 are not both H.

35. A compound according to any of numbered embodiments 1 to 33, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R20 or R21 are independently selected from H, (C1-3)alkyl and CF3, wherein R20 and R21 are not both H.

36. A compound according to numbered embodiment 35, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R20 or R21 are independently selected from H, methyl and CF3, wherein R20 and R21 are not both H. 37. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 can be selected from:

38. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 can be selected from:

 39. A compound according to numbered embodiment 37, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 is selected from:

A compound according to numbered embodiment 39, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 is selected from:

41. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 is selected from:

42. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 is selected from:

43. A compound according to numbered embodiment 41, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 is selected from:

 and

44. A compound according to numbered embodiment 43, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R18 is selected from:

45. A compound according to numbered embodiment 44, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

46. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A is a 6 to 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused, bridged or spiro.

47. A compound according to numbered embodiment 46, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A is a 6 or 9 membered mono or bi- cyclic ring system containing one N ring member, optionally wherein the ring system is substituted, where possible, with 1, 2, 3 or 4 substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH2)0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN; wherein when A is a bicyclic ring system, the bicyclic ring system is fused.

48. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A is substituted, where possible, with 1, 2 or 3 substituents independently selected from alkyl and oxo.

49. A compound according to numbered embodiments 46 to 48, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A is selected from: 50. A compound according to numbered embodiment 1, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

V is -O-, and Z is selected from -C(R16)(R17)-CH2- and -C(R16)(R17)-; or,

V is selected from -CH 2 -C(R16)(R17)- and -C(R16)(R17)-, and Z is -O-.

51. A compound of formula (I) according to numbered embodiment 1, 2 or 50, wherein the compound is a compound of formula (Ic),

Formula (Ic), or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof.

52. A compound according to numbered embodiment 50 or 51, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R16 is H.

53. A compound according to any of numbered embodiments 50 to 52, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

R16 and R17 are both H.

54. A compound according to any of numbered embodiments 1, 2 or 50 to 53, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl and optionally, where possible, 1, 2, or 3 further substituents independently selected from halo, alkyl, OH, oxo, cycloalkyl, alkoxy, -(CH 2 )0-2-heteroaryl, heterocycloalkyl, C(=O)R12, C(=O)OR13, C(=O)NR13R14, NR13R14, CF 3 , CN, and phenyl.

55. A compound of formula (I) according to numbered embodiment 54 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated isotope, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A' is a 5-membered monocyclic ring system containing one N ring member and optionally one further ring member selected from N and O, wherein the ring system is substituted with phenyl.

56. A compound of formula (I) according to numbered embodiment 55 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A' is selected from

57. A compound of formula (I) according to numbered embodiment 56 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A' is selected from: 58. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

AW- is selected from

-(CH 2 )0-3-(CHR15)-(CH 2 )0-3-A, -(CHR12)-A, -O-(CHR12)-A, -(CH 2 )0-3-A, -(CH 2 )0-3-0-(CH 2 )0-3-A, -(CH 2 )0-3-NH-(CH 2 )0-3-A, -(CH 2 )0-3-N(R12)-(CH 2 )I.3-C(=0)-A, -(CH 2 )0-3-NH-C(=0)-(CH 2 )0-3-A,

-C(=0)N(R12)-(CH 2 )0-3-A, -(CH 2 )0-3-C(=0)-(CH 2 )0-3-A, -(CH 2 )0-3-(phenyl)-(CH 2 )0-3-A, -NH-SO 2 -A and -SO 2 -NH-A.

59. A compound according to numbered embodiment 58, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

AW- is selected from -(CH 2 )0-3-A, and -(CH 2 )0-3-0-(CH 2 )0-3-A .

60. A compound according to numbered embodiment 59, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

AW- is selected from -(CH 2 )0-1-A, and -(CH 2 )0-1-0-A

61. A compound according to numbered embodiment 60, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

AW- is selected from -(CH 2 )0-1-A, and -(CH 2 )-O-A.

62. A compound according to any of numbered embodiments 1 to 49, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

AW- is -(CH 2 )0-1-O-A. 63. A compound according to numbered embodiment 62, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

AW- is -(CH 2 )-O-A.

64. A compound according to any of numbered embodiments 1, 2, or 50 to 57, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein AW- is A'W- and A'W- is selected from -(CH 2 )0-3-A'.

65. A compound according to numbered embodiment 64, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

A'W- is -(CH 2 )0-1-A'.

66. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

Y is -N(R12)-.

67. A compound according to numbered embodiment 66, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

Y is -NH- or N(CH 3 ).

68. A compound according to any one of numbered embodiments 66 or 67, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

Y is -NH-. 69. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is aryl; or B is a 6-membered monocyclic or 9- or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

70. A compound according to numbered embodiment 69, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is aryl; or B is a 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

71. A compound according to numbered embodiment 70, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is aryl; or B is a 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

72. A compound according to numbered embodiment 71, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is phenyl substituted as for aryl; or B is a 6-membered monocyclic or 9- or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

73. A compound according to numbered embodiment 72, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is phenyl substituted as for aryl; or B is a 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

74. A compound according to numbered embodiment 73, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is phenyl substituted as for aryl; or B is a 6-membered monocyclic or 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

75. A compound according to numbered embodiment 74, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl; or B is a 6- membered monocyclic or 9- or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

76. A compound according to numbered embodiment 75, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl; or B is a 6- membered monocyclic or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

77. A compound according to numbered embodiment 76, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl; or B is a 6- membered monocyclic or 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

78. A compound according to numbered embodiment 74 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is phenyl optionally substituted as for aryl, or B is pyridyl or isoquinolinyl, optionally substituted as for heteroaryl a .

79. A compound according to numbered embodiment 78 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl, or B is pyridyl or isoquinolinyl, optionally substituted as for heteroaryl a .

80. A compound according to numbered embodiment 78 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein optionally substituted as for heteroaryl a , or B is phenyl optionally substituted as for aryl.

81. A compound according to numbered embodiment 80 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein optionally substituted as for heteroaryl a , or B is phenyl substituted with CN and optionally 1 or 2 further substituents as for aryl. 82. A compound according to any of the preceding numbered embodiments or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is unsubstituted; or wherein B is aryl, B is substituted with CN and optionally 1 or 2 further substituents as for aryl; or wherein B is heteroaryl a , B is substituted with alkyl, halo, OH, or NH2, and optionally 1 or 2 further substituents as for heteroaryl a .

83. A compound according to numbered embodiment 78, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein

B is selected from optionally further substituted with 1 or 2 substituents as for heteroaryl a ; or each optionally further substituted with 1 or 2 substituents as for heteroaryl a ; with no substituents ; or with no further substituents.

84. A compound according to numbered embodiment 83, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is selected from the following groups: optionally further substituted with 1 or 2 substituents as for heteroaryl a ; or B is with no substituents; or B is with no further substituents.

85. A compound according to numbered embodiment 84, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is selected from

86. A compound according to any of numbered embodiments 1-68, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is heteroaryl a .

87. A compound according to numbered embodiment 86 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein Y is attached to B at a carbon atom on the heteroaryl a ring.

88. A compound according to numbered embodiment 86 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein Y is attached to B at a carbon atom on the heteroaryl a ring, and the two ring atoms adjacent to the carbon atom on the heteroaryl a ring to which Y attaches are both carbon. 89. A compound according to any of numbered embodiments 86 to 88, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is a 9 or 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

90. A compound according to numbered embodiment 89, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is a 10-membered bicyclic aromatic ring, containing, where possible, 1, 2, 3 or 4 ring members independently selected from N, N(R12), S and O, optionally substituted as for heteroaryl a .

91. A compound according to numbered embodiment 90, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is a 10-membered bicyclic aromatic ring, containing 1 or 2 ring members independently selected from N or N(R12), optionally substituted as for heteroaryl a .

92. A compound according to numbered embodiment 91 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is isoquinolinyl, optionally substituted as for heteroaryl a .

93. A compound according to numbered embodiment 92 or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein optionally substituted as for heteroaryl a .

94. A compound according to any of numbered embodiments 86 to 93, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is substituted with NH2, and optionally substituted with 1 or 2 further substituents as for heteroaryl a .

95. A compound according to numbered embodiment 94, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is isoquinolinyl, substituted with NH2 at the 1- position optionally further substituted with 1 or 2 substituents as for heteroaryl a .

96. A compound according to numbered embodiment 95, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein B is optionally further substituted with 1 or 2 substituents as for heteroaryl a .

97. A compound according to any of the preceding numbered embodiments, or a tautomer, isomer, stereoisomer (including an enantiomer, a diastereoisomer and a racemic and scalemic mixture thereof), a deuterated analogue, and a pharmaceutically acceptable salt and/or solvate thereof, wherein alkoxy is a linear O-linked hydrocarbon of between 1 and 6 carbon atoms (C1-C6), or a branched O-linked hydrocarbon of between 3 and 6 carbon atoms (C3-C6); alkoxy may optionally be substituted with 1 or 2 substituents independently selected from OH, CN, CF3, and fluoro.

98. A compound selected from Table 1, Table 2, Table 3, Table 4, and Table 5, or a pharmaceutically acceptable salt, solvate, or solvate of a salt thereof.

99. A compound selected from Table 1; Table 2; compounds 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, and 92 of Table 3; compounds 93, 94, 95, 96, 97, 98, 99, and 101 of Table 4; and Table 5, or a pharmaceutically acceptable salt, solvate, or solvate of a salt thereof.

100. A pharmaceutical composition comprising: a compound, or a pharmaceutically acceptable salt and/or solvate thereof, according to any of numbered embodiments 1 to 99, and at least one pharmaceutically acceptable excipient.

101. A compound, or a pharmaceutically acceptable salt and/or solvate thereof, as defined in any of numbered embodiments 1 to 99, or the pharmaceutical composition according to numbered embodiment 100, for use in medicine.

102. A compound, or a pharmaceutically acceptable salt and/or solvate thereof, as defined in any of numbered embodiments 1 to 99, or the pharmaceutical composition according to numbered embodiment 100, for use in a method of treatment of a disease or condition in which Factor XI la activity is implicated.

103. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102, wherein the disease or condition in which Factor Xlla activity is implicated is a bradykinin-mediated angioedema, wherein the bradykinin-mediated angioedema is hereditary angioedema.

104. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102, wherein the disease or condition in which Factor Xlla activity is implicated is a bradykinin-mediated angioedema, wherein the bradykinin-mediated angioedema is non hereditary.

105. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102, wherein the disease or condition in which Factor Xlla activity is implicated is selected from vascular hyperpermeability, stroke including ischemic stroke and haemorrhagic accidents; retinal edema; diabetic retinopathy; impaired visual acuity; DME; retinal vein occlusion; and AMD.

106. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102 wherein the disease or condition in which Factor Xlla activity is implicated is a thrombotic disorder. 107. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 106, wherein the thrombotic disorder is thrombosis; thromboembolism caused by increased propensity of medical devices that come into contact with blood to clot blood; prothrombotic conditions such as disseminated intravascular coagulation (DIC), Venous thromboembolism (VTE), cancer associated thrombosis, complications caused by mechanical and bioprosthetic heart valves, complications caused by catheters, complications caused by ECMO, complications caused by LVAD, complications caused by dialysis, complications caused by CPB, sickle cell disease, joint arthroplasty, thrombosis induced to tPA, Paget-Schroetter syndrome and Budd-Chari syndrome; atherosclerosis; COVID-19; acute respiratory distress syndrome (ARDS); idiopathic pulmonary fibrosis (IPF); rheumatoid arthritis (RA); and cold-induced urticarial autoinflammatory syndrome.

108. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102 wherein the disease or condition in which Factor Xlla activity is implicated is neuroinflammation; neuroinflammatory/neurodegenerative disorders such as MS (multiple sclerosis); other neurodegenerative diseases such as Alzheimer's disease, epilepsy and migraine; sepsis; bacterial sepsis; inflammation; vascular hyperpermeability; and anaphylaxis.

109. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102, wherein the disease or condition in which Factor Xlla activity is implicated is obesity or diabetes.

110. A compound, a pharmaceutically acceptable salt and/or solvate thereof, or a pharmaceutical composition for use as defined in numbered embodiment 102, wherein the disease or condition in which Factor Xlla activity is implicated is kidney disease, renal fibrosis, glomerulosclerosis, renal scarring, ischemia/reperfusion injury in native or transplant kidneys, and acute kidney injury. Synthetic Methods

The compounds of the present invention can be prepared according to the procedures of the following schemes and examples, using appropriate materials, and are further exemplified by the specific examples provided herein below. Moreover, by utilising the procedures described herein, one of ordinary skill in the art can readily prepare additional compounds that fall within the scope of the present invention claimed herein. The compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention. Those skilled in the art will readily understand that known variations of the conditions, processes and order in which the synthetic steps are performed in the following preparative procedures can be used to prepare these compounds.

The compounds and intermediates of the invention may be isolated in the form of their pharmaceutically acceptable salts, such as those described previously herein above. The interconversion between free form and salt form would be readily known to those skilled in the art.

It may be necessary to protect reactive functional groups (e.g. hydroxy, amino, thio or carboxy) in intermediates used in the preparation of compounds of the invention to avoid their unwanted participation in a reaction leading to the formation of the compounds. Conventional protecting groups, for example those described by T. W. Greene and P. G. M. Wuts in "Protective groups in organic chemistry" John Wiley and Sons, 4 th Edition, 2006, may be used. For example, a common amino protecting group suitable for use herein is tert-butoxy carbonyl (boc), which is readily removed by treatment with an acid such as trifluoroacetic acid or hydrogen chloride in an organic solvent such as dichloromethane. Alternatively, the amino protecting group may be a benzyloxycarbonyl (Cbz or Z) group which can be removed by hydrogenation with a palladium catalyst under a hydrogen atmosphere or 9- fluorenylmethyloxycarbonyl (Fmoc) group which can be removed by solutions of secondary organic amines such as diethylamine or piperidine in an organic solvent. Carboxyl groups are typically protected as esters such as methyl, ethyl, benzyl or tert-butyl which can all be removed by hydrolysis in the presence of bases such as lithium or sodium hydroxide. Benzyl protecting groups can also be removed by hydrogenation with a palladium catalyst under a hydrogen atmosphere whilst tert-butyl groups can also be removed by trifluoroacetic acid. Alternatively, a trichloroethyl ester protecting group is removed with zinc in acetic acid. A common hydroxy protecting group suitable for use herein is a methyl ether, deprotection conditions comprise refluxing in 48% aqueous HBr, or by stirring with borane tribromide in an organic solvent such as DCM. Alternatively, where a hydroxy group is protected as a benzyl ether, deprotection conditions comprise hydrogenation with a palladium catalyst under a hydrogen atmosphere. Palladium catalysed cross coupling reactions to form carbon-carbon, carbon-nitrogen and carbon-oxygen bonds typically require a palladium catalyst and ligand capable of forming an in situ catalytically active LnPd (0) species. More recently, precatalysts have been identified that contain the desired ligand and a labile group capable of eliminating to rapidly generate the active L n Pd (0) catalyst species in the reaction mixture (see, for example http://dx.doi.org/10.1039/c2sc20903a, http://dx.doi.org/10.1002/ange.201207750, http://dx.doi.org/10.1021/jo500355k, http://dx.doi.org/10.1021/ol401208t, http://dx.doi.org/10.1021/acs.orglett.8b00325). The choice of catalyst and ligand or precatalyst can be determined by literature or catalyst screening, by the skilled person.

The graphic representations of racemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are taken from Maehr J. Chem. Ed. 62, 114-120 (1985): solid wedges and broken wedges ( are used to denote the absolute configuration of a chiral element; solid bold lines and broken bold lines are geometric descriptors indicating the relative configuration shown, but denoting racemic character;

As used herein, a depiction including wedges or broken lines (eg. or indicates that the structure encompasses purity of that relative or absolute configuration of at least 80% ee, preferably >90% ee.

As used herein, when a compound possesses a centre of asymmetry, its depiction with simple lines (eg. ) indicates that the structure includes any and all stereoisomers, without regard to enantiomeric purity. The invention is illustrated by the following non-limiting examples in which the following abbreviations and definitions are used:

All reactions were carried out under an atmosphere of nitrogen unless specified otherwise. Hydrogenations were typically carried out using an H-Cube® reactor (manufactured by Thalesnano, Inc, Hungary).

References to the use of microwave, a microwave reactor, microwave heating and microwave irradiation all refer to the use of a CEM Discover Microwave Reactor.

References to the use of a phase separator refer to columns fitted with a selectively permeable, optimized frit material that separates aqueous phase from an organic phase under gravity.

1 H NMR spectra were recorded using instrumentation selected from

- Bruker (500MHz or 400MHz) spectrometer with a Bruker Avance II or Avance III console

- Oxford (400 MHz) AS400 magnet with a Inova console

- 300 MHz Varian Mercury+ NMR System and reported as chemical shift (ppm). It will be understood that, where exchangeable protons are present in any compound, the number of protons in the 1 H NMR spectra may not exactly correspond to the number of protons in the structure of any compound synthesised herein.

Molecular ions were obtained using LCMS with appropriate conditions selected from

- Chromolith Speedrod RP-18e column, 50 x 4.6 mm, with a linear gradient 10% to 90% 0.1% HCO2H/MeCN into 0.1% HCO2H/H2O over 13 min, flow rate 1.5 mL/min;

- Agilent, X-Select, acidic, 5-95% MeCN/water over 4 min. Data was collected using a Thermofinnigan Surveyor MSQ mass spectrometer with electrospray ionisation in conjunction with a Thermofinnigan Surveyor LC system;

- LCMS (Waters Acquity UPLC, C18, Waters X-Bridge UPLC C18, 1.7 pm, 2.1x30mm, Basic (0.1% Ammonium Bicarbonate) 3 min method;

- LCMS (Agilent, X-Select, Waters X-Select C18, 2.5 pm, 4.6x30 mm, Acidic 4 min method, 95-5 MeCN/water);

- LCMS (Agilent, Basic, Waters X-Bridge C18, 2.5 pm, 4.6x30 mm, Basic 4 min method, 5-95 MeCN/water;

- Acquity UPLC BEH C18 1.7 pM column, 50 x 2.1 mm, with a linear gradient 10% to 90% 0.1% HCO2H/MeCN into 0.1% HCO2H/H2O over 3 min, flow rate 1 mL/min. Data was collected using a Waters Acquity UPLC mass spectrometer with quadropole dalton, photodiode array and electrospray ionisation detectors; - Agilent 1100 LC/MSD with Kinetex® 5 pm EVO C18 100 A LC 50 x 4.6 mm and Gemini® 5 pm NX- C18 110 A LC 50 x 4.6 mm columns. Acidic mobile phases used a linear gradient of 5-95% 10 mM aq. NH 4 HCO 2 /MeCN and basic mobile phases used a linear gradient of 5-95% 10 mM aq. NI-UHCOa/MeCN. Samples were run over 3 minutes, using a flow rate of 2.2 mL/min, and a pressure range of 0-200 bar. Data was collected using a Waters 3100 Mass Detector with single high resolution quadrupole and photomultiplier detectors and High Performance ZsprayTM dual- orthoganal API sources for standard ESI, or multimode ESI/APCI/ESCi®.

- UPLC (CSH C18 Column, 130A, 1.7 pm, 2.1 mm x 30 mm, 3 min method, 0.1% Formic acid, 2-100% MeCN/water)

- LCMS (Cortecs C18+, 90A, 2.7 pm, 2.1 mm x 30 mm, 3 min method, 0.1% Formic acid, 5-100% MeCN/water)

- UPLC (BEH C18 Column, 130A, 1.7 pm, 2.1 mm x 30 mm, 3 min method, 0.1% Ammonium Hydroxide, 2-100% MeCN/water)

- LCMS (Kinetex Evo C18, 130A, 2.5 pm, 2.1 mm x 30 mm, 3 min method, 0.1% Ammonium Hydroxide, 5-100% MeCN/water)

Flash chromatography was typically carried out over 'silica' (silica gel for chromatography, 0.035 to 0.070 mm (220 to 440 mesh) (e.g. Merck silica gel 60)), and an applied pressure of nitrogen up to 10 p.s.i accelerated column elution. Alternatively, pre-prepared cartridges of silica gel were used, for example pre-packed SiliaSep™ columns from Silicycle or Sfar C18 D - Duo 100 A 30 pm columns from Biotage. Typical conditions included, for example, flow rate range of 18-200 mL/min, with an applied pressure range of 0-225 PSI. Hexanes, EtOAc, DCM, MeOH, were used as mobile phases for normal-phase chromatography purifications. MeOH, MeCN, lOmM Ammonium Formate pH 4 in H2O and lOmM Ammonium Bicarbonate pH 10 in H2O buffers were used as mobile phases for reverse-phase chromatography. It will be understood that alternative conditions (such as flow rate ranges, applied pressures, solvents and pH) may be used for flash chromatography in order to separate and purify compounds synthesised herein.

The term "prep HPLC" refers to reverse phase preparative HPLC purifications. Typical instrumentation and conditions include, for example, Waters Fraction Lynx Prep-HPLC with MWD/DAD & MSD using

- 5-95% acetonitrile/methanol with 0.1% NH4OH pH 10 in H2O as a basic mobile phase

- 5-95% acetonitrile/methanol with 0.1% HCO2H pH 3 in H2O as an acidic mobile phase It will be understood that alternative conditions (such as choice of column, flow rate ranges, solvents, modifiers and pH) may be used for prep HPLC in order to separate and purify compounds synthesised herein.

The procedure of lyophilisation (or freeze drying) is generally well known in the art. Typically the substance is taken up in water, if necessary, with the addition of a minimum amount of MeCN to aid dissolution, and frozen, typically by rapid cooling in a cold bath at -78 °C. The resulting frozen solid mixture is evaporated to dryness in vacuo.

The term "concentrated" refers to evaporation of solvent under reduced pressure using a rotary evaporator, heating where necessary.

All solvents and commercial reagents were used as received.

IUPAC chemical names were generated using automated software such as Dotmatics Studies Notebook or ChemDraw (PerkinElmer). Compounds produced by the methods below may be isolated in salt forms. However, compound naming used herein typically refers to the compound without any salt counter ion.

The example compounds described herein can be prepared using conventional synthetic methods for example, but not limited to, the routes outlined in the General Schemes below, using, for example, the General Methods below.

General Methods

1. General Method 1: (GM1): mesylation and chlorination a. General Method la (GMla): mesylation

A solution of alcohol (1.0 eq) in DCM was cooled in an ice/water bath and MsCI (1.2 eq) was added dropwise followed by TEA (1.4 eq) maintaining cooling. The reaction was stirred at rt for 2-18 h. The reaction was diluted with DCM and washed with water. The aqueous layer was extracted with DCM (x 3) and the combined organics were washed with brine, dried (Na2SO4), filtered and concentrated. The crude product was used without purification or purified by flash chromatography. b. General Method lb (GMlb): chlorination via a mesylate

MsCI (2.5 eq) was added to a solution of TEA (2.8 eq) and alcohol (1.0 eq) in DCM while cooling in an ice/water bath. The reaction was stirred at rt for 18 h. The reaction was diluted with DCM and washed with sat. NaHCO3 (aq). The aqueous layer was extracted with DCM (x 3) and the combined organics were washed with brine, dried (Na2SO4), filtered and concentrated. The crude product was purified by flash chromatography. c. General Method lc (GMlc): chlorination via NCS

A solution of indole or azaindole (1.0 eq) in DCM was protected from light and treated with NCS (3.75 eq) at rt for 12 - 48 h. The mixture was treated with IM HCI (aq) and the phases separated. The organic phase was washed with brine, dried ( Na2SO4), filtered, concentrated and purified by flash chromatography.

2. General Method 2 (GM2): SN2 alkylation (O and N) a. General Method 2a (GM2a): SN2 alkylation: NaH

To a suspension of NaH (60% wt. on mineral oil) (1.1 eq) in DMF in an ice/water bath was added a solution of alcohol, pyrrole or indole (1.0 eq) in DMF dropwise over 2 min. The mixture was allowed to warm to rt for 5 min before cooling again in an ice/water bath and treating with a solution of the alkylhalide or mesylate (1.0 eq) in DMF over 2 min. The mixture was maintained in an ice/water bath for 1 h before being allowed to warm to rt, or heated at elevated temperature and stirred for 2-18 h. Sat. NH4CI (aq) or sat. NaHCO3 (aq) was added and extracted with EtOAc (x 3). The organic phases were combined, dried (MgSO4), filtered and concentrated. The crude product was purified by flash chromatography. b. General Method 2b (GM2b): SN2 alkylation; CS2CO3 or K2CO3

A solution of alkyl halide or mesylate (1-2 eq), amine (1.0 eq), and base such as K2CO3, or CS2CO3 (2.5 eq), in a solvent such as DMF, DMSO or MeCN, was stirred at 80 °C for 2-18 h. MeOH (5 mL) was added, and the mixture was diluted with water. The product was extracted into EtOAc (x 2) and washed with brine. The organic layer was dried (Na2SO4), filtered and concentrated. The product was either used directly or purified by flash chromatography.

3. General Method 3 (GM3): Reduction a. General Method 3a (GM3a): nitrile reduction; H-Cube® with Pd/C or Raney Ni cartridge

The nitrile was dissolved in a 0.5M NH3/MeOH solution and passed through an H-Cube® reactor (Pd/C or Raney Ni cartridge) at 50 °C, 'full' hydrogen delivery mode (50 bar), flow rate: 1 mL/min. The reaction was concentrated to afford the product which was used without further purification. b. General Method 3b (GM3b): nitrile, amide and ester reduction; LiAIH4 in THF

To a solution of amide, nitrile, or ester (1.0 eq) in THF in an ice/water bath was added LiAl H4 (2M in THF) (2.0 eq) dropwise and the reaction mixture was allowed to warm to rt then stirred for 4-18 h. The reaction mixture was cooled in an ice/water bath, treated portionwise with Na2SO4.10H2O (3.5 eq) and stirred for 30 min before being dried (MgSO4), filtering and washing with THF. The filtrate was concentrated to afford the crude product which was used without purification or purified by flash chromatography. c. General Method 3c: borane-THF reduction

A solution of nitrile (1.0 eq) in THF was cooled in an ice/water bath before borane (IM in THF, 2.0 eq) was added dropwise. The reaction was allowed to warm to rt then heated to 60 °C for 16-96 h. MeOH was added and heating continued at 60 °C for 24 h before cooling to rt and concentrating. The product was isolated and purified using one of the following methods: i) The crude product was loaded onto an SCX in MeOH and washed with MeOH. The product was eluted with 7M NH3 in MeOH and the eluent concentrated. ii) The crude product was purified by flash chromatography iii) BOC2O (1.2 eq) was added to the crude reaction mixture and stirred overnight. The solvent was concentrated. The product was taken up in DCM, washed with water and brine, dried ( Na2SO4 , filtered and concentrated. The boc-protected amine was either used without further purification or purified by flash chromatography d. General Method 3d: NiCl2 reduction

A solution of nitrile (1.0 eq), NiCl.6H2O (1.0 eq) and BOC2O (3.0 eq) in MeOH was cooled in an ice/water bath and sodium borohydride (NaBH4) (5.0 eq) added portionwise. The reaction was allowed to warm to rt and stirred for 18 h. Water was added, and the reaction mixture filtered, washed with THF and concentrated. The crude product was purified by flash chromatography. e. General Method 3e: hydrogenation; Pd/C

To a solution of nitrile (1.0 eq) in MeOH or EtOH under an inert atmosphere was added 10% Pd/C (0.1-0.2 eq). Additives such as HCI, sulfuric acid, or BOC2O may optionally be added. The reaction was stirred under an atmosphere of H2 (g) for 2-72h. The catalyst was removed by filtration over Celite®, which was washed with EtOH. The product was isolated following concentration of the filtrate and used directly or purified by flash chromatography. f. General Method 3f: aromatic ring reduction

A biaryl ring (1.0 eq) was dissolved in EtOH and subjected to hydrogenation in the H-Cube® at 70 °C, 50 bar, 1 mL/min using a 10% Pd/C CatCart, recirculating when necessary. The solvent was removed in vacuo to afford the product which was used without purification.

4. General Method 4 (GM4): Buchwald coupling A suspension of amine or alcohol (1.0 eq), aryl halide (1.1 eq) and a base such as CS2CO3, NaOtBu (2.0 eq) or LiHMDS (2.0 eq) in a degassed solvent such as THF or 1,4-dioxane was purged with N2 (g). A Buchwald palladium precatalyst, such as BrettPhos Pd G3, (0.11 eq) was added and the mixture degassed and purged with N2 (g) for 5 min. The reaction was heated in a sealed vial at rt - 60°C for 30 min - 3 days as required. The product was isolated and purified using one of the following methods: i) The reaction was quenched with AcOH (2.0 eq) and concentrated. The crude was purified by SCX, loading and washing with MeOH then eluting the product with NH3 in MeOH followed by purification by flash chromatography or prep HPLC. ii) The reaction was quenched with AcOH (2.0 eq), filtered through Celite®, washing with EtOAc and the filtrate concentrated. The crude product was purified by flash chromatography. iii) The reaction mixture was acidified with AcOH (2.0 eq) and stirred for 5 min, IM NH3 in MeOH was added and the reaction mixture was concentrated on to silica and purified by flash chromatography. iv) The reaction mixture was dry loaded on to silica and purified by flash chromatography. v) The reaction was concentrated, and the product taken forward to deprotection without purification.

5. General Method 5 (GM5): boc deprotection; HCI or TFA a. General Method 5a (GM5a): boc deprotection; HCI/dioxane

A suspension of boc protected amine (1.0 eq) in 1,4-dioxane was treated with 4M HCI in dioxane (10.0 eq) and the reaction stirred at rt for 2-24 h. The product was isolated and purified using one of the following methods: i) The reaction mixture was concentrated, optionally azeotroping with Et20 or toluene to afford the product as a hydrochloride salt. ii) The reaction mixture was concentrated, and the product converted to free base using a bicarbonate cartridge, PL-HCO3 MP SPE (Agilent), loading in MeOH. The filtrate was concentrated and triturated with Et20 to afford the product. b. General Method 5b (GM5b): boc deprotection; TFA

A mixture of boc protected amine (1.0 eq) in DCM was treated with TFA (10.0 eq) and stirred at rt for 2 h. The product was isolated and purified using one of the following methods: i) The mixture was passed directly through an SCX column and washed with MeOH. The product was eluted with a solution of 7M NH3 in MeOH and concentrated. The crude product was purified by flash chromatography or prep HPLC. ii) The mixture was concentrated and loaded onto a PL-HCO3 MP SPE cartridge eluting with a mixture of MeCN and MeOH (1:1 v/v, 3 x 2 mL). The crude product was purified by flash chromatography or prep HPLC. iii) The mixture was diluted with aq. NaOH (2M, 10 mL) and a mixture of DCM and MeOH (9:1 v/v, 20 mL). The phases were separated and the aq. Phase was extracted with a mixture of DCM and MeOH (9:1 v/v). The combined organic phases were dried and concentrated. The residue was purified by flash chromatography or prep HPLC iv) The mixture was diluted with aq. NaOH (2M, 10 mL) and a mixture of DCM and MeOH (9:1 v/v, 20 mL). The phases were separated, and the aqueous phase was extracted with a mixture of DCM and MeOH (9:1 v/v). The combined organic layers were dried (NazSO^, filtered, and concentrated. Product was purified by PL-HCO3 MP SPE cartridge eluting with a mixture of MeCN and MeOH (1:1 v/v), followed by flash chromatography or prep HPLC.

6. General Method 6 (GM6): pyridone chlorination

Pyridone (1.0 eq) was suspended in phosphorus oxychloride (large excess) and heated at reflux for 4 h. The reaction mixture was evaporated then azeotroped with toluene (x 2). The residue was used immediately in the next step, taking care to exclude moisture.

7. General Method 7 (GM7): SNAr Alkylation (O and N) a. General Method 7a (GM7a): SNAr O-alkylation using NaH

To a suspension of NaH (60% wt. on mineral oil) (1.04 eq) in DMF in an ice/water bath was added a solution of alcohol (1.02 eq) in DMF dropwise over 2 min. The mixture was allowed to warm to rt for 5 min before cooling again in an ice/water bath and treating with pyridyl halide or aryl halide (1.0 eq). The reaction mixture was maintained in an ice/water bath for 1 h then warmed to rt for 18 h. The reaction mixture was cooled in an ice/water bath and sat. NajCOs (aq) was added followed by water. This was extracted with EtOAc (x 3) and the organic phases were combined, washed with 1:1 water/brine and brine. The organic phase was dried (MgSO4), filtered and concentrated. The crude product was purified by flash chromatography. b. General Method 7b (GM7b): SNAr O-alkylation using CS2CO3

To a solution of alcohol (1.0 eq) and pyridyl halide (1.0 eq) in MeCN was added CS2CO3 (2.0 eq) and the mixture was stirred in a sealed vial at 50 °C for 18-72 h. The product was isolated and purified using one of the following methods i) The reaction mixture was cooled to rt and diluted with water (10 mL). The crude product was extracted into DCM, dried (MgSO4), filtered and concentrated. The residue was purified by flash chromatography ii) The reaction mixture was filtered through Celite® and the filtrate was concentrated to yield the crude product which was either used without further purification or purified by flash chromatography iii) The crude reaction mixture was passed directly through an SCX column, loading in MeOH. The SCX was washed with MeOH and the product was eluted with 7M NH3 in MeOH. The crude product was purified by flash chromatography. c. General Method 7c (GM7c): SNAr O-alkylation using NaO l Bu

A solution of alcohol (1.0 eq), aryl halide (1.0 eq) and NaO l Bu (3.0 eq) in NMP was stirred in the microwave at 140 °C for 4 h. The crude reaction mixture was loaded onto an SCX column in MeOH and washed with MeOH and the product was eluted with 7M NH3 in MeOH (50 mL). The product was concentrated and purified by flash chromatography or prep HPLC. d. General Method 7d (GM7d): SNAr N-alkylation

Amine (1.0 eq) and halopyridine (1.0 eq) were dissolved in MeCN. K2CO3 (3.0 eq) was added and the reaction was stirred at 60 - 120 °C for 60 - 90 min under thermal heating or microwave irradiation. The reaction was diluted with water and extracted with IPA/CHCI3 (1:10) (x 3). The combined organics were washed with brine, dried (MgSO4) and concentrated. The product was isolated and used directly or purified by flash chromatography.

8. General Method 8 (GM8): 2,4-dimethoxybenzyl deprotection

A solution of 2,4-dimethyoxybenzyl protected amine (1.0 eq) in TFA (10 eq.) was stirred at rt - 50 °C for 60 min.

The product was isolated and purified using one of the following methods: i) The reaction mixture was concentrated. The resulting residue was suspended in MeOH (2 mL) and loaded onto SCX, which was flushed with MeOH. The product was eluted with a solution of 7M NH3 in MeOH. The solvent was concentrated and the product was purified by flash chromatography or prep HPLC. ii) The mixture was concentrated and loaded onto a PL-HCO3 MP SPE cartridge eluting with a mixture of MeCN and MeOH (1:1 v/v, 3 x 2 mL). The crude product was purified by flash chromatography or prep HPLC. iii) The mixture was diluted with NaOH (aq) (2M, 10 mL) and a mixture of DCM and MeOH (9:1 v/v, 20 mL). The phases were separated, and the aqueous phase was extracted with a mixture of DCM and MeOH (9:1 v/v). The combined organic phases were dried and concentrated. The residue was purified by flash chromatography or prep HPLC iv) The mixture was diluted with NaOH (aq) (2M, 10 mL) and a mixture of DCM and MeOH (9:1 v/v, 20 mL). The phases were separated, and the aqueous phase was extracted with a mixture of DCM and MeOH (9:1 v/v). The combined organic layers were dried (NazSO^, filtered, and concentrated. Product was purified by PL-HCO3 MP SPE cartridge eluting with a mixture of MeCN and MeOH (1:1 v/v), followed by flash chromatography or prep HPLC.

9. General Method 9 (GM9): carbamate protection

To a solution of aminopyridine (1.0 eq) and TEA (2.0 eq) in DCM in an ice/water bath was added methylchloroformate (3.0 eq) and the reaction was stirred at rt for 48 h. The reaction mixture was diluted with DCM and washed with water. The aqueous was extracted with DCM (x 3) and the combined organics dried (Na2SO4 , filtered and concentrated. The crude product was triturated with EtOAc.

10. General Method 10 (GM10): carbamate deprotection a. General Method 10a: KOH

A mixture of methyl carbamate (1 eq) and KOH (6 eq) in MeOH was stirred at 60 °C for 12 - 48 h. The product was isolated and purified using one of the following methods: i) The reaction was quenched with AcOH (6.0 eq) and the mixture was left to stir for 5 min before being concentrated. The residue was passed directly through an SCX in MeOH. The SCX was washed with MeOH and the product was eluted with 7M NH3 in MeOH and lyophilised. ii) The reaction was quenched with AcOH (6.0 eq) and the mixture was left to stir for 5 min before being concentrated. The residue was passed directly through an SCX in MeOH. The SCX was washed with MeOH and the product was eluted with 7M NH3 in MeOH. The product was purified by flash chromatography or prep HPLC iii) The reaction was quenched with AcOH (6.0 eq), concentrated, and purified by prep HPLC. b. General Method 10b: LiOH

To a solution of methyl carbamate (1 eq) in THF/water (10:1) was added lithium hydroxide monohydrate (3-5 eq) and the reaction stirred at 60 °C for 18 h - 4 days. The mixture was cooled to rt and concentrated. The crude residue was purified via flash chromatography or prep HPLC

11. General Method 11: SEM deprotection

TFA (10 eq) was added dropwise to a rapidly stirred solution of indole or azaindole (1.0 eq) in DCM. The mixture was stirred at rt for 18 h. The reaction mixture was passed directly through an SCX in MeOH. The SCX was washed with MeOH and the product was eluted with 2.5M NH3 in MeOH and concentrated. The crude product was purified by flash chromatography or prep HPLC.

12. General Method 12: Alkylation and cyclisation

To a solution of the amino substituted heteroaryl (e.g. pyridine) (1 eq) in EtOH (3 mL) were added NaHCO3 (2 eq) and the chloromethyl aldehyde or chloromethyl ketone as applicable (2 eq). The mixture was stirred for 20 h at 75 °C. The mixture was then cooled to rt, filtered through Celite® washing with EtOAc (50 mL) and concentrated. The product was purified by flash chromatography.

13. General Method 13: Hydroxylation

A solution of the aryl bromide or heteroaryl bromide (1.0 eq), B2Pin2 (2.0 eq) and Pd(dppf)CI2 (0.1 eq) in anhydrous, degassed 1,4-dioxane was purged with N2 (g) for 5 min. KOAc (3.0 eq) was added and the reaction stirred at 90 °C for 2 - 18 h. AcOH (2.0 eq) and water (1 mL) were added and the reaction stirred at rt for 15 min - 18 h. A solution of H2O2 in water (30% w/w, 2 eq) was added and the reaction mixture stirred for 1 - 18 h. i) Typically for basic compounds: solid Na2S2O3 was added and the reaction was stirred at rt for 5 min then diluted with EtOAc, filtered through Celite ® and concentrated. The residue was purified by SCX and flash chromatography. ii) Typically for non basic compounds: sat. Na2S2O3 (aq) and sat. NaHCO3 (aq) were added and the aqueous was extracted with EtOAc. The combined organics were washed with brine, dried (MgSO4), concentrated, and purified by flash chromatography General Method 14: Esterification

To a stirred suspension of carboxylic acid (1.0 eq) in MeOH in an ice/water bath was added thionyl chloride (1.1 eq) dropwise. The reaction was warmed to rt and stirred for 20 h. The solvent was concentrated and trituration with Et20 afforded the product. General Method 15: Mitsunobu

To a mixture of alcohol (1.0 eq) and phenol (1.1 eq) in anhydrous THF at 0 °C was added PPh3 (1.5 eq) followed by DIAD (1.5 eq). The mixture was stirred at 0 °C for 10 min then warmed to rt and stirred for 24 h and then concentrated. The residue was diluted with NaOH (aq) (IM) and DCM. The phases were separated, and the aqueous phase was extracted with DCM (x 2). The combined organic phases were washed with sat. NaHCO3 (aq) and brine, dried (Na2SO4), filtered and concentrated. Purification by flash column chromatography afforded the product. The crude product was either used without further purification or purified by flash chromatography or prep HPLC. General Method 16: Oxidation

To a solution of alcohol (1.0 eq) in DCM at 0 °C was added Dess-Martin periodinane (2.0 eq). The mixture was stirred at rt for 18 h. A further portion of Dess-Martin periodinane (0.2 eq) was added to the solution at 0 °C. The mixture was stirred at rt for 4 h and diluted with sat. NaHCO3 (aq) (50 mL) and sat. Na2S2O3 (aq) (50 mL) at rt. The mixture was stirred for 15 min at rt and the phases were separated. The aqueous phase was extracted with DCM (x 2) and the combined organic phases were washed with sat. Na2S2O3 (aq), sat. NaHCO3 (aq), brine, dried (Na2SO4), filtered and concentrated to afford the product. General Method 17 (GM17): Reductive amination

To a solution of aldehyde (1.0 eq) and amine (1.10 eq) in DCE was added AcOH (1.15 eq) and the mixture stirred for 30 min then STAB (2.0 eq) was added. The mixture was stirred for 18 - 72 h at rt before quenching with sat. NaHCO3 (aq) and extracting with DCM (x 3). The combined organic layers were dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography afforded the product. General Method 18 (GM18): Amide coupling a. General Method 18a (GM18a): Amide coupling using HATU

To a solution of carboxylic acid (1.2 eq) in anhydrous DMF was added DIPEA (3 eq) and HATU (1.05 eq). The reaction was stirred at rt for 40 min then amine (1.0 eq) was added and stirring continued for 60 min. Concentration afforded the product which was either used without further purification or purified by flash chromatography or prep HPLC b. General Method 18b (GM18b): Amide coupling using PyBop

To a mixture of carboxylic acid (1.0 eq) in DCM was added amine (1.0 eq), PyBOP (1.2 eq) and DIPEA (3.0 eq). The mixture was stirred at rt for 16 h and then concentrated. The residue was partitioned between sat. NaHCO3 (aq) and EtOAc. The aqueous was washed with EtOAc (x 3). The combined organic layers were dried (Na2SO4), filtered and concentrated. Purification by flash chromatography or prep HPLC afforded the product. General Method 19 (GM19): Sulfonamide formation

To a solution of amine (1.0 eq) and TEA (2.3 eq) in anhydrous DCM (2 mL), cooled in an ice/water bath, was added sulfonyl chloride (2.0 eq). The mixture was stirred in an ice/water bath for 2 h. Concentration afford the product. The crude product was either used without further purification or purified by flash chromatography or prep HPLC. General Method 20 (GM20): Carbamate formation a. General Method 20a (GM20a): Carbamate formation using CDI

To a solution of CDI (1.5 eq) in anhydrous MeCN at rt was added alcohol (1.0 eq). The mixture was stirred for 2h and then concentrated. The residue was partitioned between H2O and DCM and the aqueous washed with DCM (x 2). Combined organics were dried (Na2SO4), filtered and concentrated. The intermediate (2.0 eq) was dissolved in anhydrous THF and amine (1.0 eq) added followed by DBU (0.3 eq). The mixture was heated to 65 °C for 18h - 4 days then cooled and concentrated. The residue was partitioned between H2O and DCM and the aqueous washed with DCM (x 2). Combined organics were dired (Na2SO4), filtered and concentrated. Purification by flash chromatography or prep HPLC afforded the product. b. General Method 20b (GM20b): Carbamate formation using phosgene

To a stirred solution of alcohol (1.06 eq) and TEA (1.56 eq) in anhydrous DCM at rt was added phosgene (20% in toluene, 1.02 eq) and the reaction stirred for 1 h. A solution of amine (1.0 eq) and TEA (1.55 eq) in anhydrous DCM was added and stirring continued for 24 h. The mixture was quenched with 1 M NaOH and brine (1:1), and the layers were separated. The aqueous layer was extracted with DCM (x 2), and the combined organic layers were dried (Na2SO4), filtered, and concentrated. The crude product was purified by flash chromatography. General Method 21 (GM21): Urea formation a. General Method 21a (GM21a): Urea formation using CDI

To a solution of amine (1.6 eq) in DMF was added CDI (1.5 eq) and DIPEA (3.6 eq). The mixture was stirred at rt for 2 h then a solution of scaffold amine (1.0 eq) in DMF was added. The mixture was stirred at rt for 72 h and then concentrated. The crude product was either used without further purification or purified by flash chromatography or prep HPLC. b. General Method 21b (GM21b): Urea formation using triphosgene

Part 1: A mixture of amine (1.6 eq) in anhydrous THF was added to a mixture of triphosgene (0.65 eq) and pyridine (2.2 eq) in anhydrous THF at rt. The mixture was stirred for 18-72 h.

Part 2: To the reaction mixture was added a mixture of scaffold amine (1.0 eq) and TEA (3 eq) in anhydrous DMSO. The reaction was stirred for 1 - 36 h. The product was isolated and purified using one of the following methods: i) The reaction mixture was concentrated and purified by flash chromatography. ii) The reaction was diluted with brine and extracted with a mixture of DCM and MeOH (9:1 v/v). The combined organic phases were washed with brine, dried (Na2SO4), filtered and concentrated. The crude product was either used without further purification or purified by flash chromatography or prep HPLC c. General Method 21c (GM21c): Urea formation using isocyanates

To a solution of amine (1.0 eq) and TEA (3.0 eq) in DMSO was added isocyanate (1.1 eq), and the mixture was stirred for 16 h. The mixture was quenched with MeOH, filtered and purified by automated prep HPLC.

General Schemes

General Schemes 1-6 outline synthetic routes for certain example compounds and RgA, RgB, RgC and RgD refer to various substituents as required by the examples. For the sake of clarity General Schemes 1-6 are drawn with an oxabicyclohexyl central core group. Similar chemistry can be applied to examples with different core groups such as bicyclopentyl, azabicyclohexyl, and oxabicycloheptyl.

Alcohols such as la are typically reacted to form a suitable leaving group, such as halide or mesylate and can be generated using conditions well known in the art such as, for example; chlorination via a mesylate, bromination with PBr3, or bromination with CBr 4 and PPh3, using a suitable solvent such as DCM, THF or CCL (General Method 1), to give compound 2a. Compound 2a is subsequently reacted with an alcohol or amine 3 under typical alkylation conditions (General Method 2, eg KCtBu or NaH in DMF or CS2CO3 or K2CO3 in MeCN or DMSO, with heating as necessary). The nitrile 4 can be reduced to amine 5a under a variety of standard literature conditions well known in the art (General Method 3); for example under hydrogenation in the presence of Raney Ni, alternatively hydrogenation in the presence of Pd/C, or alternatively with NiCL and sodium borohydride (NaBH4) in the presence of BOC2O, or alternatively with borane. The amine 5a is reacted with aryl bromide or chloride 6 under Buchwald coupling conditions (General Method 4). This Buchwald coupling is carried out for example using a Buchwald pre catalyst, such as BrettPhos Pd G3, BrettPhos Pd G4 or Pd-PEPPSI-lpent, in the presence of a base such a sodium tert- butoxide, CS2CO3, or lithium hexamethyldisilazide (LiHMDS), in a solvent such as 1,4-dioxane or THF. The aryl bromide or chloride 6 can be prepared from readily available starting materials using methods known in the art, or as described herein. Depending on the identity of RgB, a deprotection step (detailed below) may be required to obtain the example compound.

Alternatively, for example as shown in General Scheme 2, where material is available as the amine, for example compound lb, a similar synthetic sequence can be applied.

The amine is protected, for example with a tert-butoxy carbamate protecting group, using standard conditions such as TEA and BOC2O in DCM or THF. Following formation of a leaving group and alkylation, the tert-butoxy carbamate protecting group is removed from compound 8a using standard conditions such as TFA, or HCI in 1,4-dioxane (General Method 5). Finally, Buchwald coupling (General Method 4) completes the route.

The order of steps can also be reversed, for example as shown in General Scheme 3.

An amine such as lb can undergo a Buchwald (General Method 4), followed by formation of a leaving group (General Method 1) and finally alkylation with compound 3 (General Method 2).

Furthermore, the Buchwald coupling (General Method 4), is also possible with alcohols. This requires a suitable protecting group strategy as shown in General Schemes 4 and 5.

In General Scheme 4, a protected alcohol is in the form of an ester such as Id. Following the usual sequence of steps, formation of a leaving group and alkylation, the ester 8b is reduced using standard conditions such as LiAIH4 (General Method 3) to reveal the alcohol. Finally, Buchwald coupling (General Method 4) of alcohol 5b completes the route. This typically requires an elevated temperature, for example 90 °C or above and in some cases alternative catalysts, such as RockPhos Pd G3, are more suitable.

Again, the order of steps can also be reversed, for example as shown in General Scheme 5.

The alcohol such as Id can undergo a Buchwald (General Method 4), followed by reduction of the ester, formation of a leaving group (General Method 1) and finally alkylation with compound 3 (General Method 2).

In examples, where RgA contains a bicyclic aromatic, it can be partially saturated during the synthesis, as shown in General Scheme 6.

Typically the aromatic ring such as 12 is subjected to hydrogenation at elevated temperature (General Method 3) using H2, 10% Pd/C to give compound 13. This transformation can take place on the free or protected amine.

In example compounds described herein containing a primary or secondary amine, a protecting group strategy may be required. Alternative protecting groups can be used with different deprotection conditions, that is, an orthogonal protecting group strategy can be applied. General Schemes 7-12 outline possible protecting group strategies that may be used for the synthesis of the examples.

For example, for compounds defined herein containing a 6,6 ring system, as shown in General Scheme 7, a protected amine can be installed. Initially the pyridone 14 may be chlorinated, typically using phosphorous oxychloride (General method 6). The resulting chloride 15 may be reacted with 2,4- dimethoxybenzylamine using General Method 7, for example using basic conditions such as K2CO3, CS2CO3, or pyridine in a solvent such as NMP or MeCN, either thermally and/or under microwave conditions. RgT refers to various substituents as required by the example compounds. Typically, at the end of the synthetic sequence, the 2,4-dimethoxybenzyl protecting group is removed using TFA at 50 °C, either undiluted or diluted in DCM (General Scheme 8, General Method 8). RgT, RjA and RjB refer to various substituents as required by the examples.

Alternatively, when starting materials are available with the amine already installed, a carbamate protecting group can be used. For example, as outlined in General Scheme 9, the amine is reacted with methyl chloroformate under basic conditions with organic bases such as TEA or DIPEA in a solvent such as DCM to afford the methyl carbamate 21 (General Method 9). RjC refers to various substituents as required by the examples.

Typically, at the end of the synthetic sequence, the methyl carbamate protecting group is deprotected using basic conditions, such as KOH or LiOH in solvents such as 1,4-dioxane, MeCN, THF and optionally 10% water, at elevated temperature, typically 50 °C (General Scheme 10, General Method 10). RjC and RjD refer to various substituents as required by the examples.

Another protecting group that may be used where example compounds described herein contain a 6,6 ring system is boc.

Where, for example, example compounds described herein contain a 5,6 ring system, SEM, boc and sulfonyl protecting groups may typically be used. Protecting groups may subsequently be deprotected using standard literature procedures, for example those described by T. W. Greene and P. G. M. Wuts in "Protective groups in organic chemistry" John Wiley and Sons, 4 th Edition, 2006.

An example of the installation of a SEM protecting group is shown in General Scheme 11 whereby the indole 24 is treated with a base such as NaH in a solvent such as DMF, followed by addition of SEM-CI (General Method 2).

Typically, at the end of the synthetic sequence the SEM protecting group is deprotected using acidic conditions such as TFA in DCM (General Scheme 12, General Method 11). RjD refers to various substituents as required by the examples.

In examples where RgA contains an aromatic or heteroaromatic bicycle they can be synthesised using procedures described in the literature and, for example, as depicted in General Scheme 13.

Amino pyridines such as 3a can be reacted with chloromethyl aldehydes and ketones 28 by refluxing in a solvent such as ethanol (General Method 12). Hydroxylation of the aryl bromide 3b to the corresponding aryl alcohol 3c is typically completed in a one-pot tandem process via Miyaura borylation (Palladium(O)- Catalyzed Cross-Coupling Reaction of Alkoxydiboron with Haloarenes: A Direct Procedure for Arylboronic Esters. T. Ishiyama, M. Murata, N. Miyaura, J. Org. Chem., 1995, 60, 7508-7510) and subsequent hydroxylation using hydrogen peroxide (General Method 13).

Other examples herein may be based on an azabicyclohexyl central core group, as shown in General Schemes 14 - 17. These General Schemes may also be applied to other central core groups such as bicyclopentyl, oxabicyclohexyl and oxabicycloheptyl. The starting materials may be readily available from commercial sources or may be known in the literature. Alternatively, some prior manipulation may be required, for example, as shown in General Scheme 14, a carboxylic acid such as le can be esterified using methods generally known in the art, for example via formation of the acid chloride in an appropriate alcohol solvent such as methanol (General Method 14).

When azabicyclohexyl rings are used they may be protected with a suitable nitrogen protecting group such as Boc or Cbz (General Scheme 15). Alternatively, the nitrogen can be reacted using methods generally known in the art and detailed herein below, to provide amide, urea, thiourea, alkyl, heteroaryl and sulfonamide analogues as shown in General Schemes 15 and 17.

As outlined in previous schemes and as repeated in General Scheme 16, RgA can be installed by transforming the alcohol Id to a leaving group, such as a mesylate or halide 7b and reacting with aryl alcohols (General Method 2). Alternatively, the alcohol Id can be directly reacted with phenols under Mitsunobu conditions (General Method 15) using standard conditions such as DIAD and triphenylphosphine in THF. The alcohol Id can also be coupled with aryl halides under SNAr in the presence of NaH (General Method 7a), or under Buchwald conditions (General Method 4), typically using RockPhos Pd G3 as the catalyst and elevating the temperature to 100 °C. The ester 8b is then reduced to alcohol 5c under conditions well known in the art such as lithium borohydride (LiBH4) or lithium aluminium hydride (LiAIH4) or lithium triethylborohydride (LiEt3BH) (General Method 3b) depending on other functionality contained in the compound. Oxidation to aldehyde 5d is typically achieved using a reagent such as Dess Martin periodinane (General Method 16). Finally, reductive amination (General Method 17) with amine 6a completes the route. Alternatively (not shown in the Schemes) the alcohol 5c can be transformed to an amine using methods generally known in the art, such as via conversion to a leaving group (General Method 1) then via an azide or Gabriel synthesis to provide the primary amine and finally completing the route via a Buchwald coupling (General Method 4) as shown previously in General Scheme

1.

When azabicyclohexyl rings are used, in which the nitrogen may be protected by a protecting group (i.e. 7b where V= NRi), a late stage functionalisation is possible, as shown in General Scheme 17. The nitrogen protecting group is removed using the appropriate conditions (for example boc deprotection via General Method 5, or Cbz deprotection via General Method 3e, 2,4-dimethoxybenzyl deprotection via General Method 8, or SEM deprotection via General Method 11). Manipulation of 7c using chemistry well known in the art can provide a variety of compound classes. For example, reductive alkylation with aldehydes (General Method 17) can give alkylated compounds such as 29a. Cross-coupling reactions with aryl halides, such as Buchwald reactions (General Method 4) or SNAr alkylation (General Method 7) can install aromatic groups to give 29f. Reaction of 7c with carboxylic acids or acid chlorides can be used to provide amides 29b (General Method 18) or reaction with sulphonyl chlorides (General Method 19) can provide sulphonamides 29c. Reaction with CDI and alcohols can provide carbamates 29d (General Method 20). Ureas such as 29e can be formed by reaction of 7c with isocyanates, or by reacting with CDI or triphosgene and amines (General Method 21).

The routes described are also applicable to azabicyclohexyl isomers and the order of steps may vary depending on starting material availability or as chemical functionality requires.

Synthesis of Intermediates

Intermediate 1 Methyl (6-bromoisoquinolin-l-yl)carbamate

Following General Method 9, 6-bromoisoquinolin-l-amine (1.50 g, 6.72 mmol) was protected. The crude was suspended in water (100 mL) and stirred for 30 min before being collected by filtration and dried in the vacuum oven overnight to give the product (1.12 g, 44% yield).

[M+H] + = 281.1

1 H NMR (500 MHz, DMSO-d6) 3.70 (3H, s), 7.58 - 7.72 (1H, m), 7.79 (1H, d, J = 9.0, 2.0 Hz), 8.04 (1H, d, J = 9.1 Hz), 8.25 - 8.30 (1H, m), 8.33 (1H, d, J = 5.8 Hz), 10.18 (1H, s)

Intermediate 2

Methyl (6-bromo-4-chloroisoquinolin-l-yl)carbamate

Methyl N-(6-bromo-l-isoquinolyl)carbamate (100 mg, 0.36 mmol) was dissolved in chloroform (5 mL), NCS (52 mg, 0.39 mmol) was added and the reaction stirred at reflux for 18 h. To the reaction was added sat. NaHCO3 (aq.) (30 mL) and it was washed with DCM (30 mL), dried (Na2SO4) and concentrated. The crude product was purified by flash chromatography (Silica, 0-80% EtOAc in Pet. Ether) to give the product (74 mg, 59% yield).

[M+H] + = 316.8/318.7

1 H NMR (CDCI 3 , 400 MHz) 6 3.84 (3H, s), 7.36 (1H, s), 7.75 (1H, dd, J = 9.0, 1.9 Hz), 7.93 (1H, d, J = 9.0 Hz), 8.37 (2H, d, J = 4.9 Hz)

Intermediate 3

Methyl (5-bromoisoquinolin-l-yl)carbamate Following General Method 9, 5-bromoisoquinolin-l-amine (1.12 g, 5.02 mmol) was protected. The product was dried under high vacuum to yield (838 mg, 56% yield).

[M+H] + = 281.1

Intermediate 4

5-Bromo-N-(2,4-dimethoxybenzyl)isoquinolin-l-amine

Following a variation of General Method 7d, to a solution of 5-bromo-l-chloroisoquinoline (0.5 g, 2.06 mmol) in pyridine (3 mL), was added 2,4-dimethoxybenzylamine (0.69 g, 4.12 mmol). The reaction was heated at 150 °C in a CEM Microwave for 60 min. The mixture was diluted with DCM (20 mL) and water (20 mL). The aqueous layer was re-extracted with DCM (3 x 10 mL) and the combined organics were washed with brine (20 mL). The organic layer was dried (Na2SO4), filtered and concentrated. Purification by flash chromatography (Silica, 20-50% EtOAc in Pet. Ether) afforded the product (276 mg, 50% yield). [M+H] + = 373.0/375.0

1 H NMR (DMSO-d6, 400 MHz) 6 3.71 (3H, d, J = 2.6 Hz), 3.82 (3H, d, J = 2.8 Hz), 4.62 (2H, d, J = 5.4 Hz), 6.41 (1H, dd, J = 8.5, 2.5 Hz), 6.56 (1H, d, J = 2.6 Hz), 6.94 - 7.14 (2H, m), 7.42 (1H, t, J = 8.0 Hz), 7.96 (3H, ddd, J = 16.4, 7.1, 3.2 Hz), 8.38 (1H, d, J = 8.2 Hz)

Intermediate 5

Nl-(2,4-Dimethoxybenzyl)isoquinoline-l,5-diamine A mixture of 5-bromo-N-(2,4-dimethoxybenzyl)isoquinolin-l-amine (746 mg, 2.0 mmol), 2,2,2- trifluoroacetamide (339 mg, 3.0 mmol), Cul2 (38 mg, 0.2 mmol), K2CO3 (553 mg, 4.0 mmol) and N,N'- dimethylethylenediamine (35 mg, 0.4 mmol) were combined in a reaction vial. Anhydrous 1,4-dioxane (7 mL) was added, and the suspension purged with N2, before being capped and then heated to 75 °C for 24 h. The reaction was recharged with 2,2,2-trifluoroacetamide (339 mg, 3.0 mmol), Cul2 (38 mg, 0.2 mmol), K2CO3 (553 mg, 4.0 mmol) and N,N'-dimethylethylenediamine (35 mg, 0.4 mmol). The mixture was degassed with N2 and heated at 70 °C for 18 h. Water was added (3 mL) and the reaction heated at 80 °C for 6 h. The mixture was partitioned between EtOAc (30 mL) and water (10 mL). The aqueous layer was extracted with further EtOAc (2 x 30 mL) and the combined organics were washed with brine (50 mL), dried (Na2SO4 , filtered and concentrated. Purification by flash chromatography (Silica, 0-100% EtOAc/DCM) afforded the product (492 mg, 72% yield).

[M+H] + 310.1

1 H NMR (CDCI 3 , 400 MHz) 6 3.79 (3H, s), 3.85 (3H, s), 4.10 (2H, br s), 4.72 (2H, d, J = 5.3 Hz), 5.58 - 5.67 (1H, m), 6.44 (1H, dd, J = 8.2, 2.4 Hz), 6.49 (1H,d, J = 2.4 Hz), 6.78 - 6.86 (2H, m), 7.12 (1H, dt, J = 8.4, 1.1 Hz), 7.21 (1H, dd, J = 8.4, 7.4 Hz), 7.30 (1H, d, J = 8.2 Hz), 8.01 (1H, d, J = 6.1 Hz)

Intermediate 6

4-Bromo~l-((2-(tr!methylsilyl)ethoxy)methyl)-lH-pyrrolo[2 ,3-b]pyridine

Following General Method 2a, 4-bromo-lH-pyrrolo[2,3-b]pyridine (500 mg, 2.54 mmol) was reacted with SEM-CI (494 μL, 2.79 mmol) for 2 h. The reaction mixture was quenched by the careful addition of water (10 ml) followed by repeat extraction with EtOAc (3 x 20 ml). The combined organic layers were then washed with sat. NaHCO3 (aq) (30 ml), water (30 ml) and brine (30 ml), before being dried (MgSO4), filtered and concentrated. Purification by flash chromatography (Silica, 0-100% EtOAc in iso-hexane) afforded the product (500 mg, 57% yield).

[M+H] + - 32.7.2

1 H NMR (500 MHz, DMSO-d6) 6 -0.11 (s, 9H), 0.75 - 0.86 (m, 2H), 3.43 - 3.56 (m, 2H), 5.63 (s, 2H), 6.52 (d, J

= 3.6 Hz, 1H), 7.43 (d, J = 5.1 Hz, 1H), 7.79 (d, J = 3.6 Hz, 1H), 8.16 (d, J = 5.1 Hz, 1H) Intermediate 7

5-Bromo-3-chloro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH- pyrrolo[2,3-b]pyridine

Following General Method 2a, 5-bromo-3-chloro-lH-pyrrolo[2,3-b]pyridine (480 mg, 2.07 mmol) was reacted with SEM-CI (0.4 mL, 2.28 mmol) for 2 h. The reaction was quenched with water (2 mL) and diluted with EtOAc (40 mL). The organic layer was washed with water (20 mL), IM HCI (aq) (20 mL), 1:1 water/brine (20 mL) and brine (20 mL), dried (MgSO4), filtered and concentrated. Purification by flash chromatography (Silica, 0-100% EtOAc in iso-hexane) afforded the product (485 mg, 60% yield).

[M+H] + = 363.0

1 H NMR (500 MHz, DMSO-d6) 6 -0.10 (9H, s), 0.81 (2H, t, J = 7.9 Hz), 3.51 (2H, t, J = 7.9 Hz), 5.60 (2H, s), 7.98 -8.01 (1H, m), 8.20 - 8.24 (1H, m), 8.44 - 8.47 (1H, m)

Intermediate 8

5-Bromo-N-(2,4-dimethoxybenzyl)-4-fluoroisoquinolin-l-ami ne

5-Bromo-l-chloro-4-fluoroisoquinoline

A solution of 5-bromo-2H-isoquinolin-l-one (9.0 g, 40.2 mmol) and Selectfluor® (17.1 g, 48.2 mmol) in MeCN (120 mL) and MeOH (120 mL) were heated at 50 °C for 3 h. The reaction mixture was concentrated and reacted using General Method 6, in 1,2-dichloroethane (200 mL) using benzyltriethylammonium chloride (915mg, 4.0mmol) and phosphorus oxychloride (45mL) at 90 °C for 24 h. The reaction mixture was concentrated, and the residue partitioned between DCM (500 mL) and water (500 mL). The organic layer was washed with water (300 mL) and brine (300 mL), dried (MgSO4) and evaporated. The crude was purified by flash chromatography (Silica, 0-30% EtOAc in Pet. Ether) to give the product (5.70 g, 55% yield). [M+H] + = 261.9

1 H NMR (500 MHz, CDCI 3 ) 6 8.39 - 8.33 (m, 1H), 8.23 (d, J= 4.0 Hz, 1H), 8.12 - 8.06 (m, 1H), 7.57 (t, J= 8.0 Hz, 1H)

5-Bromo-N-(2,4-dimethoxybenzyl)-4-fluoroisoquinolin-l-ami ne

Following General Method 7d, 5-bromo-l-chloro-4-fluoroisoquinoline (5.70 g, 21.9 mmol) was reacted with 2,4-dimethoxybenzylamine (4.93 mL, 32.8 mmol) in l-methyl-2-pyrrolidinone (80 mL) at 100 °C for 48 h. The crude product was purified by flash chromatography (Silica, 0-30% EtOAc in Pet. Ether) to give the product (1.05 g, 12% yield).

1 H NMR (500 MHz, DMSO) 6 8.43 (dd, J= 8.1, 2.3 Hz, 1H), 8.06 (dd, J= 7.6, 0.9 Hz, 1H), 7.89 (d, J= 5.1 Hz, 1H), 7.81 (t, J= 5.6 Hz, 1H), 7.49 (t, J= 8.0 Hz, 1H), 7.05 (d, J= 8.3 Hz, 1H), 6.56 (d, J= 2.4 Hz, 1H), 6.41 (dd, J= 8.4, 2.4 Hz, 1H), 4.57 (d, J= 5.5 Hz, 2H), 3.82 (s, 3H), 3.72 (s, 3H)

19 F NMR (471 MHz, DMSO) 6 -149.9(s)

[M-H] = 389.2

Intermediate 9 l-(Benzenesulfonyl)-4-bromo-2-chloropyrrolo[2,3-b]pyridine l-(Benzenesulfonyl)-4-bromopyrrolo[2,3-b]pyridine

To a solution of 4-bromo-lH-pyrrolo[2,3-b]pyridine (5.00 g, 25.4 mmol) in DCM (130 mL) was added benzenesulfonyl chloride (4.86 mL, 38.1 mmol), 4-dimethylaminopyridine (310 mg, 2.54 mmol) and TEA (10.6 mL, 76.1 mmol). The reaction mixture was stirred at rt for 2 h. Upon completion the reaction mixture was concentrated. The product was suspended in DCM (50 mL) and concentrated onto silica. The material was purified via flash chromatography (Silica, 0-50% EtOAc in Pet. Ether) to afford the product (8.39 g, 98% yield).

[M+H] + = 338.9 l-(Benzenesulfonyl)-4-bromo-2-chloropyrrolo[2,3-b]pyridine l-(Benzenesulfonyl)-4-bromopyrrolo[2,3-b]pyridine (1.00 g, 2.97 mmol) in anhydrous THF (16 mL) was cooled to -41 °C and lithium diisopropylamide (2M in THF) (356 mL, 7.12 mmol) was added slowly. The resulting precipitate was stirred for 30 min at -41 °C before benzenesulfonyl chloride (757 μL, 5.93 mmol) was added. The reaction mixture was stirred for 2.5 h at -41 °C. The reaction mixture was quenched with water (10 mL) and diluted with EtOAc (20 mL). The layers were separated, and the aqueous layer was back extracted with EtOAc (2 x 20 mL). The organic layers were combined, washed with brine (10 mL), dried (MgSO4), filtered and concentrated. Purification by flash chromatography (Silica, 0-60% EtOAc in Pet. Ether) afforded the product (895 mg, 49% yield).

[M+H] + = 372.9 l-(Benzenesulfonyl)-4-bromo-2-chloropyrrolo[2,3-b]pyridine

To a solution of l-(Benzenesulfonyl)-4-bromo-2-chloropyrrolo[2,3-b]pyridine (895 mg, 1.44 mmol) in 1,4- dioxane (7 mL) was added NaOtBu (324 mg, 2.89 mmol). The reaction mixture was stirred at 80 °C for 2 h, diluted with EtOAc (10 mL) and washed with brine (10 mL). Layers were separated and the organic layer was dried (MgSO4), filtered and concentrated. Purification by flash chromatography (Silica, 0-25% EtOAc in Pet. Ether) afforded the product (295 mg, 88% yield).

[M+H] + = 232.9

1 H NMR (CDCl3. 400 MHz) 6 6.47 (1H, s), 7.32 (1H, d, J = 5.3 Hz), 8.11 (1H, d, J = 5.3 Hz,) NH proton not observed

Intermediate 10

3-Methylimidazo[l,2-a]pyridin-6-ol

According to a modification of General Method 13, 6-bromo-3-rnethylirnidazo[l,2-a]pyridine (525 mg, 2.49 mmol) was reacted with B2Pin2 (1.42 g, 5.6 mmol)at 100 °C for 5 h. The mixture was cooled to rt before AcOH (285 μL, 4.97 mmol) and water (1 mL) were added. The mixture was stirred for 15 min before a solution of H2O2 in water (30% w/w, 508 μL, 4.97 mmol) was added dropwise at 0 °C. The mixture was stirred for 90 min at rt, quenched with solid Na2S2O3 (5 mg), filtered through Celite® and concentrated. The product was passed directly through SCX and washed with MeOH. The product was eluted with a solution of IM NH3 in MeOH and concentrated to afford the product (160 mg, 33% yield). [M+H] + = 149.1

1 H NMR (DMSO, 400 MHz) 6 2.36 (3H, s), 6.92 (1H, dd, J = 9.6, 2.3 Hz), 7.25 (1H, s), 7.40 (1H, d, J = 9.5 Hz), 7.59 (1H, d, J = 1.9 Hz), 9.45 (1H, s)

Intermediate 11

7-hydroxy-2,3-dihydroindolizin-5(1H)-one

Ethyl 7-hydroxy-5-oxo-2,3-dihydro-lH-indolizine-8-carboxylate (125 mg, 0.56 mmol) was dissolved in 12M HCI (aq) (1.87 mL, 22.4 mmol) and stirred at reflux for 18 h. The solution was cooled to rt, then azeotroped with Et2O ( 3 x 5 mL) and concentrated to provide the product (123 mg, 87% yield) which was used for the next step without further purification.

[M+H] + = 152.1

Intermediate 12

Methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l-carboxylate;hy drochloride

The product was prepared from 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l-carboxylic acid (CAS 1522098-73-2, Tetrahedron Letters 55 (2014) 466-468) following the procedure described in ChemistrySelect 2019, 4, 4933 - 493.

Intermediate 13

Methyl 5-(hydroxymethyl)-2-oxabicyclo[3.1.1]heptane-l-carboxylate

Following General Method 14, 5-(hydroxymethyl)-2-oxabicyclo[3.1.1]heptane-l-carboxylic acid sodium salt (600 mg, 2.8 mmol) was reacted for 22 h. The reaction mixture was diluted with DCM (40 mL) and washed with water (20 mL). The aq layer was extracted with DCM (30 mL). The combined organic fractions were dried (Na2SO4) and concentrated to afford the product (409 mg, 71% yield).

[M+H] + = 187.1

1 H NMR (CDCI 3 , 400 MHz) 6 1.94 - 2.05 (4H, m), 2.33 (2H, ddt, J = 7.2, 2.3, 1.0 Hz), 3.54 (2H, s), 3.77 (3H, s), 4.19 - 4.27 (2H, m) Methyl 5-(methylsulfonyloxymethyl)-2-oxabicyclo[3.1.1]heptane-l-car boxylate

According to General Method la, methyl 5-(hydroxymethyl)-2-oxabicyclo[3.1.1]heptane-l-carboxylate (404 mg, 1.95 mmol) was reacted with MsCI (227 μL, 2.93 mmol for 22 h. Aqueous work up afforded the product (372 mg, 58% yield).

1 H NMR (CDCI 3 , 400 MHz) 6 2.05 - 2.13 (4H, m), 2.35 - 2.44 (2H, m), 3.05 (3H, s), 3.78 (3H, s), 4.12 (2H, s), 4.19 - 4.26 (2H, m)

Methyl 5-[(1,3-dioxoisoindol-2-yl)methyl]-2-oxabicyclo[3.1.1]heptan e-l-carboxylate

A suspension of methyl 5-(methylsulfonyloxymethyl)-2-oxabicyclo[3.1.1]heptane-l-car boxylate (100 mg, 0.303 mmol) and potassium phthalimide (62.0 mg, 0.333 mmol) in DMF (4 mL) was stirred at 100 °C for 3 h. The mixture was cooled to rt, taken up in water (20 mL) and washed with EtOAc (2x30 mL). The combined organic layers were washed with brine (10 mL), dried (Na2SO4) and concentrated to afford the product (98 mg, 82% yield).

[M+H] + = 316.0

1 H NMR (CDCI3, 400 MHz) 6 2.01 - 2.14 (4H, m), 2.30 - 2.39 (2H, m), 3.73 (5H, s), 4.15 - 4.21 (2H, m), 7.71

- 7.79 (2H, m), 7.83 - 7.90 (2H, m)

[5-(Aminomethyl)-2-oxabicyclo[3.1.1]heptan-l-yl]methanol

To a stirred solution of methyl 5-[(1,3-dioxoisoindol-2-yl)methyl]-2-oxabicyclo[3.1.1]heptan e-l- carboxylate (98 mg, 0.25 mmol) in IPA (4 mL) and water (1 mL), was added NaBF (56 mg, 1.49 mmol). After stirring for 24 h, AcOH (256 μL, 4.48 mmol) was added slowly and when the foaming subsided, the reaction was heated to 80 °C for 22 h. The reaction mixture was concentrated, the residue taken up in MeOH (1 mL) and the mixture was passed directly through an SCX column and washed with MeOH. The product was eluted with a solution of IM NH3 in MeOH and concentrated to afford the product (39 mg, 100% yield).

[M+H] + = 158.1

1 H NMR (CDCI3, 400 MHz) 6 1.60 - 1.67 (2H, m), 1.82 (2H, dd, J = 7.2, 2.7 Hz), 1.92 - 1.98 (2H, m), 2.64 (2H, s), 3.51 (2H, s), 4.07 - 4.16 (2H, m)

Intermediate 14 tert-Butyl N-[[5-(hydroxymethyl)-2-oxabicyclo[3.1.1]heptan-l-yl]methyl] carbamate

To a solution of [l-(aminomethyl)-2-oxabicyclo[3.1.1]heptan-5-yl]methanol hydrochloride (500 mg, 2.58 mmol) and TEA (1260 μL, 9.04 mmol) in THF (10 mL) at 0 °C was added BOC2O (676 mg, 3.1 mmol) and the mixture was stirred for 10 min at 0 °C. After that time the reaction was stirred for 18 h at rt. The reaction was quenched with water (30 mL), washed with EtOAc (2 x 50 mL), dried (MgSO4) and concentrated to afford the product (760 mg, 92% yield).

[M+Na] + = 280.2

1 H NMR (CDCI3) 6 1.43 (9H, s), 1.74 - 1.79 (4H, m), 1.94 (2H, t, J = 6.9 Hz), 2.53 (1H, d, J = 4.4 Hz), 3.17 (2H, d, J = 5.8 Hz), 3.46 (2H, d, J = 5.5 Hz), 4.07 (2H, t, J = 6.9 Hz), 4.94 (1H, s)

Intermediate 15

[l-[[(2-Methylpropan-2-yl)oxycarbonylamino]methyl]-2-oxab icyclo[3.1.1]heptan-5-yl]methyl methanesulfonate

According to a modification of General Method la, tert-butyl N-[[5-(hydroxymethyl)-2- oxabicyclo[3.1.1]heptan-l-yl]methyl]carbamate (760 mg, 2.36 mmol) was reacted with MsCI (219 μL, 2.84 mmol) at 0 °C for 90 min. Aqueous work up afforded the product (950 mg, 96% yield) which was used without further purification.

[M+Na] + = 358.3

1 H NMR (CDCI3) 6 1.44 (9H, s), 1.81 - 1.89 (4H, m), 2.00 (2H, t, J = 6.8 Hz), 3.02 (3H, s), 3.18 (2H, d, J = 6.0

Hz), 4.06 - 4.10 (4H, m), 4.90 (1H, s)

Intermediate 16

2-(tert-Butyl) 1-methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylat e

To a solution of methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l-carboxylate;hy drochloride (40.5 g, 176 mmol) and TEA (75 mL, 538 mmol) in anhydrous DCM (200 mL) and anhydrous MeOH (200 mL) at rt was added BOC2O (42.1 g, 193 mmol). After stirring for 16 h. The reaction was quenched with brine (200 mL). The layers were separated, and the aqueous layer was extracted with DCM (2 x 100 mL). The combined organic phases were dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (Silica, 0-20% MeOH in DCM) provided the product (37.1 g, 74% yield).

[M+H] + = 272.1

1 H NMR (CDCI3, 400 MHz) 6 1.41 (9H, s), 1.75 (2H, dd, J = 4.7, 1.9 Hz), 1.95 - 2.06 (2H, m), 2.17 (1H, s),

3.37 (2H, s), 3.76 (3H, s), 3.79 (2H, s)

Intermediate 17

2-O-Benzyl 1-O-methyl 4-(methylsulfonyloxymethyl)-2-azabicyclo[2.1.1]hexane-l,2-di carboxylate

2-O-Benzyl 1-O-methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylat e

Methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l-carboxylate hydrochloride (600 mg, 2.89 mmol) was dissolved in DCM (15 mL) and cooled to 0 °C. TEA (805 μL, 5.78 mmol) was added followed by benzyl chloroformate (407 μL, 2.89 mmol). The reaction was stirred at rt overnight before retreating with benzyl chloroformate (200 μL, 1.42 mmol) at 0 °C and stirring at rt for an additional 1 h. DCM (30 mL) and water (10 mL) were added. The organic layer was dried (MgSO4), filtered and concentrated. The crude product was purified by flash chromatography (Silica, 20-100% EtOAc in Pet. Ether) to afford the product (465 mg, 53% yield).

[M+H] + = 306.2

1 H NMR (DMSO-d6, 400 MHz) 6 1.56 - 1.66 (2H, m), 1.89 - 1.97 (2H, m), 3.32 (2H, s), 3.50 - 3.65 (5H, m), 4.74 - 4.78 (1H, m), 5.04 (2H, s), 7.30 - 7.42 (5H, m)

2-O-Benzyl 1-O-methyl 4-(methylsulfonyloxymethyl)-2-azabicyclo[2.1.1]hexane-l,2-di carboxylate

Following General Method la, 2-O-benzyl 1-O-methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l,2- dicarboxylate (1206 mg, 3.95 mmol) was reacted with MsCI (367 μL, 4.74 mmol) for 90 min, after which time aqueous work up afforded the product (1480 mg, 98% yield).

[M+H] + = 384.1.

1 H NMR (CDCI 3 , 400 MHz) 6 1.91 (2H, dd, J = 4.8, 1.9 Hz), 2.12 (2H, dd, J = 5.0, 1.9 Hz), 3.04 (3H, s), 3.51 (2H, s), 3.67 (3H, s), 4.40 (2H, s), 5.11 (2H, s), 7.27 - 7.44 (5H, m).

Intermediate 18

2-(tert-Butyl) 1-methyl 4-(((tert-butyldiphenylsilyl)oxy)methyl)-2-azabicyclo[2.1.1] hexane-l,2- dicarboxylate

To a solution of 2-(tert-butyl) 1-methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylat e (30 g, 111 mmol) and imidazole (8.28 g, 122 mmol) in anhydrous DCM (150 mL) at 0 °C was added TBDPS- Cl (31.6 mL, 122 mmol) and the reaction stirred for 1 h at 0 °C, then at rt and for 16 h. After quenching with brine (200 mL) and EtOAc (100 mL), the layers were separated, and the aqueous layer was extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with IM HCI (aq) (200 mL), brine (200 mL), IM NaOH (aq) (200 mL), dried (Na2SO4, filtered, and concentrated to afford the product (59.5 g, 95% yield).

1 H NMR (CDCI 3 , 300 MHz) 6 1.06 (9H, s), 1.44 (9H, s), 1.75 (2H, d, J = 4.6 Hz), 2.01 (2H, d, J = 4.7 Hz), 3.37 (2H, s), 3.78 - 3.79(3H, m), 3.80 (2H, s), 7.35 - 7.46 (6H, m), 7.58 - 7.67 (4H, m)

Intermediate 19 tert-Butyl 4-(((1,6-dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)- l-(hydroxymethyl)-2- azabicyclo[2.1.1]hexane-2-carboxylate

2-(tert-Butyl) 1-methyl 4-(((1,6-dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)- 2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate According to General Method 15, 2-(tert-butyl) 1-methyl 4-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane- 1,2-dicarboxylate (10.0 g, 36.8 mmol) was reacted with 4-hydroxy-l,6-dimethyl-pyridin-2-one (6.00 g, 41.0 mmol). The reaction mixture was filtered over Celite®. The solid was washed with EtOAc (200 mL) and the filtrate was concentrated. Purification by flash chromatography (silica, 0-7% MeOH in EtOAc) afforded the product (6.20 g, 30% yield, 70% pure).

[M+H] + = 393.2

1 H NMR (DMSO-d6, 400 MHz) 6 1.36 (9H, s), 1.67 (2H, dd, J = 4.5, 1.9 Hz), 2.01 - 2.09 (2H, m), 3.32 (3H, s), 3.35 (2H, s), 3.66 (3H, s), 3.69 (3H, s), 4.16 (2H, s), 5.69 (1H, d, J = 2.8 Hz), 5.89 (1H, dd, J = 2.8, 0.9 Hz) tert-Butyl 4-(((1,6-dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)- l-(hydroxymethyl)-2- azabicyclo[2.1.1]hexane-2-carboxylate

According to a variation of General Method 3b, 2-(tert-butyl) 1-methyl 4-(((1,6-dimethyl-2-oxo-l,2- dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-l, 2-dicarboxylate (6.20 g, 11.1 mmol) was reduced in the presence of LiBH4 (2M in THF, 11 mL, 22.0 mmol) for 72 h. The mixture was quenched with H2O (30 mL) and concentrated. The aqueous phase was extracted with DCM (3 x 60 mL). The combined organic layers were dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (Silica, 0-12% MeOH in EtOAc) afforded the product (2.70 g, 67% yield). [M+H] + = 366.1

1 H NMR (CDCI3, 400 MHz) 6 1.48 (9H, s), 1.66 (1H, s), 1.70 - 1.79 (4H, m), 2.30 (3H, d, J = 0.7 Hz), 3.37 (2H, s), 3.45 (3H, s), 3.94 (2H, d, J = 6.4 Hz), 4.04 (2H, s), 5.79 (2H, d, J = 0.8 Hz)

Intermediate 20

(4-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)-2-oxabicyclo[2.1.1]hexan-l- yl)methanol

According General Method 4, a suspension of [4-(aminomethyl)-2-oxabicyclo[2.1.1]hexan-l-yl]methanol (500 mg, 3.49 mmol), 5-bromo-N-(2,4-dimethoxybenzyl)isoquinolin-l-amine (1434 mg, 3.84 mmol) and NaO l Bu (503 mg, 5.24 mmol) in 1,4-dioxane (5 mL) was reacted in the presence of Brettphos Pd G4 (241 mg, 0.26 mmol) in a sealed vial at 40 °C for 18 h and then at 60 °C for 2 h. Flash chromatography (Silica, 20-100% EtOAc in Pet. Ether) afforded the product (861 mg, 57% yield).

[M+H] + = 436.2 Intermediate 21 (4-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)m ethyl)-2-oxabicyclo[2.1.1]hexan-l- yl)methyl methanesulfonate According to General Method la, a solution of (4-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5- yl)amino)methyl)-2-oxabicyclo[2.1.1]hexan-l-yl)methanol (800 mg, 1.84 mmol) was reacted with methane sulfonyl chloride (252 mg, 2.2 mmol) to afford the product (914 mg, 97% yield) which was used without purification. [M+H] + = 514.2

Intermediate 22 (1-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amino)m ethyl)-2-oxabicyclo[2.1.1]hexan-4- yl)methanol

According to General Method 4, a suspension of [l-(aminomethyl)-2-oxabicyclo[2.1.1]hexan-4- yl]methanol hydrochloride (250 mg, 1.39 mmol), 5-bromo-N-(2,4-dimethoxybenzyl)isoquinolin-l-amine (571 mg, 1.53 mmol) and NaC t Bu (334 mg, 3.48 mmol) in 1,4-dioxane (5 mL) was reacted in the presence of Brettphos Pd G4 (96 mg, 0.1 mmol) in a sealed vial at 40 °C for 18 h. Flash chromatography (Silica, 20- 100% EtOAc in Pet. Ether) afforded the product (429 mg, 71% yield).

[M+H] + = 436.2

Intermediate 23

(1-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)-2-oxabicyclo[2.1.1]hexan-4- yl)methyl methanesulfonate According to General Method la, a solution of (1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5- yl)amino)methyl)-2-oxabicyclo[2.1.1]hexan-4-yl)methanol (429 mg, 0.99 mmol) was reacted with methane sulfonyl chloride (135 mg, 1.18 mmol) to afford the product (440 mg, 87% yield) which was used without purification by chromatography.

[M+H] + = 514.2

Intermediate 24 (1-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amino)m ethyl)-2-oxabicyclo[3.1.1]heptan-5- yl)methanol

According to General Method 4, a suspension of [l-(aminomethyl)-2-oxabicyclo[3.1.1]heptan-5- yl]methanol hydrochloride (300 mg, 1.55 mmol), 5-bromo-N-(2,4-dimethoxybenzyl)isoquinolin-l-amine (578 mg, 1.55 mmol) and NaCfBu (372 mg, 3.87 mmol) in 1,4-dioxane (6 mL) was reacted in the presence of Brettphos Pd G4 (107 mg, 0.12 mmol) in a sealed vial at 40 °C for 3 days and at 60 °C for 18 h. Flash chromatography (Silica, 0-100% EtOAc in Pet. Ether followed by 0-12% MeOH in EtOAc) afforded the product (190 mg, 27% yield).

[M+H] + = 450.3

Intermediate 25 (1-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amino)m ethyl)-2-oxabicyclo[3.1.1]heptan-5- yl)methyl methanesulfonate

According to General Method la, a solution of (1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5- yl)amino)methyl)-2-oxabicyclo[3.1.1]heptan-5-yl)methanol (160 mg, 0.36 mmol) was reacted with MsCI (36 μL, 0.46 mmol) to afford the product (190 mg, 91% yield) which was used without purification.

[M+H] + = 528.2

1 H NMR (CDCI 3 ) 6 1.45 (9H, s), 1.92 (4H, s), 2.07 (2H, t, J = 6.9 Hz), 3.23 (2H, d, J = 5.9 Hz), 3.74 (2H, s), 4.09 - 4.14 (2H, m), 5.03 (1H, t, J = 5.9 Hz), 6.96 (1H, dd, J = 9.7, 2.3 Hz), 7.48 - 7.51 (2H, m), 7.56 (1H, d, J = 0.8 Hz), 7.66 (1H, d, J = 1.8 Hz)

Intermediate 26

4-((4-(((1*((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)a mino)methyl)-2-azabicyclo[2.1.1]hexan-l- yl)methoxy)-l-methylpyridin-2(1H)-one

2-O-Benzyl 1-O-methyl 4-(azidomethyl)-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylate

To 2-O-benzyl 1-O-methyl 4-(methylsulfonyloxymethyl)-2-azabicyclo[2.1.1]hexane-l,2-di carboxylate (458 mg, 1.19 mmol) in DMF (5 mL) was added NaN 3 (155 mg, 2.39 mmol) at rt. The mixture was stirred at rt for 4 days. The mixture was diluted with EtOAc (50 mL) and washed with water (4 x 10 mL) and brine (10 mL), dried (MgSO4) and concentrated to afford the product (330 mg, 75% yield). [M+H] + = 331.1

1 H NMR (CD 3 CN) 6 1.73 (2H, dd, J = 4.7, 1.9 Hz), 2.01 (2H, dd, J = 4.6, 1.4 Hz), 3.38 (2H, s), 3.56 (5H, s), 5.04 (2H, s), 7.28 - 7.37 (5H, m)

2-O-Benzyl 1-O-methyl 4-(azidomethyl)-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylate

2-O-Benzyl 1-O-methyl 4-(azidomethyl)-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylate (330 mg, 0.9 mmol) was dissolved in THF (6 mL). Triphenylphosphine, polymer bound (3mmol/ lg of resin) (453 mg, 1.35 mmol) was added and the reaction stirred at rt for 2 h. Water (162 μL, 8.99 mmol) was added and the reaction heated at reflux for 3 h. The mixture was cooled to rt and filtered, washing the resin with 10% MeOH in DCM (25 mL). The filtrate was concentrated to afford the product (240 mg, 79% yield).

[M+H] + = 305.1

1 H NMR (CDCk) 6 1.29 (2H, br s), 1.70 (2H, dd, J = 4.6, 1.7 Hz), 1.90 (2H, d, J = 4.6 Hz), 2.85 (2H, s), 3.34

(2H, s), 3.53 (3H, br s), 5.02 (2H, s), 7.19 - 7.28 (5H, m)

2-Benzyl 1-methyl 4-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate

According to a variation of General Method 4, 5-bromo-N-(2,4-dimethoxybenzyl)isoquinolin-l-amine (2239 mg, 6.00 mmol) was reacted with 2-O-benzyl 1-O-methyl 4-(aminomethyl)-2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate (1927 mg, 5.70 mmol), CS2CO3 (5610 mg, 17.0 mmol) and Brettphos Pd G4 (801 mg, 0.852 mmol) in 1,4-dioxane (25 mL) at 65 °C for 6 h. The reaction mixture was cooled, filtered (Celite®) washing with EtOAc (200 mL), and the filtrate was concentrated. Purification by flash chromatography (Silica, 0-70% EtOAc in hexanes) afforded the product (2580 mg, 76%).

[M+H] + = 597.3

1 H NMR (CDCI3, 500 MHz) 6 1.91 (2H, dd, J = 4.7, 1.9 Hz), 2.11 (2H, d, J = 4.8 Hz), 3.51 - 3.70 (7H, m), 3.80

(3H, s), 3.86 (3H, s), 4.26 (1H, s), 4.72 (2H, d, J = 5.3 Hz), 5.12 (2H, s), 5.63 (1H, t, J = 5.3 Hz), 6.45 (1H, dd, J = 8.3, 2.4 Hz), 6.50 (1H, d, J = 2.4 Hz), 6.66 (1H, d, J = 7.7 Hz), 6.75 (1H, d, J = 6.1 Hz), 7.08 (1H, d, J = 8.3 Hz), 7.28 - 7.39 (7H, m), 8.02 (1H, d, J = 6.1 Hz)

Benzyl 4-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-l-(hydroxymethyl)-2- azabicyclo[2.1.1]hexane-2-carboxylate Following a variation of General Method 3b, 2-benzyl 1-methyl 4-(((1-((2,4- dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-2-azabi cyclo[2.1.1]hexane-l,2-dicarboxylate (1713 mg, 2.87 mmol) was reduced in the presence of lithium triethylborohydride (IM in THF, 7.2 mL, 7.20 mmol) at 0 °C for 2.5 h. The reaction mixture was diluted with water (10 mL), brine (20 mL) and EtOAc (90 mL). The phases were separated, and the aqueous phase was extracted with EtOAc (2 x 50 mL). The combined organic phases were dried (MgSO4) and concentrated. Purification by flash chromatography (Silica, 20-68% EtOAc in hexanes) afforded the product (1096 mg, 67% yield).

[M+H] + = 569.8

1 H NMR (CDCI3, 400 MHz) 6 1.78 (4H, s), 3.51 (2H, s), 3.54 (2H, s), 3.80 (3H, s), 3.86 (3H, s), 3.98 (2H, d, J = 7.4 Hz), 4.24 (1H, s), 4.54 (1H, s), 4.72 (2H, d, J = 5.3 Hz), 5.14 (2H, s), 5.63 (1H, t, J = 5.4 Hz), 6.45 (1H, dd, J = 8.2, 2.4 Hz), 6.50 (1H, d, J = 2.4 Hz), 6.67 (1H, dd, J = 7.8, 0.8 Hz), 6.76 (1H, dd, J = 6.1, 0.9 Hz), 7.05 - 7.11 (1H, m), 7.37 (7H, d, J = 4.6 Hz), 8.02 (1H, d, J = 6.1 Hz)

Benzyl 4-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-l-(((1-methyl-2-oxo-l,2- dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-2- carboxylate

According to General Method 15, benzyl 4-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5- yl)amino)methyl)-l-(hydroxymethyl)-2-azabicyclo[2.1.1]hexane -2-carboxylate (1046 mg, 1.84 mmol), was reacted with 4-hydroxy-l-methyl-pyridin-2-one (254 mg, 1.93 mmol). After aqueous work up, purification by flash column chromatography (Silica, 0-20% MeOH in EtOAc) afforded the product (319 mg, 26% yield). [M+H] + = 676.4

1 H NMR (CDCI 3 , 400 MHz) 6 1.73 (2H, dd, J = 4.5, 1.9 Hz), 2.01 - 2.04 (2H, m), 3.46 (3H, s), 3.55 (2H, s), 3.57 (3H, s), 3.80 (3H, s), 3.86 (3H, s), 4.26 (1H, s), 4.47 - 4.56 (2H, m), 4.73 (2H, d, J = 5.3 Hz), 5.10 (2H, s), 5.62 (1H, d, J = 5.5 Hz), 5.82 - 5.94 (2H, m), 6.45 (1H, dd, J = 8.2, 2.4 Hz), 6.50 (1H, d, J = 2.4 Hz), 6.68 (1H, d, J = 7.7 Hz), 6.77 (1H, dd, J = 6.2, 0.9 Hz), 7.08 (2H, dd, J = 8.0, 4.4 Hz), 7.28 - 7.39 (6H, m), 8.03 (1H, d, J = 6.1 Hz) 4-((4-(((1*((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)-2-azabicyclo[2.1.1]hexan-l- yl)methoxy)-l-methylpyridin-2(1H)-one

According to a variation of General Method 3e, a solution of benzyl 4-(((1-((2,4- dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-l-(((1- methyl-2-oxo-l,2-dihydropyridin-4- yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (319 mg, 0.472 mmol) in MeOH (3 mL) was added to a mixture of Pd(OH)2/C (20 wt%, 82.0 mg, 0.0117 mmol) in MeOH (1.8 mL). The mixture was subjected to hydrogenation at 1 atmosphere at rt for 4 h. The mixture was filtered over Celite® washing with MeOH (2 x 25 mL), and the filtrate was concentrated to afford the product (234 mg, 91% yield).

[M+H] + = 542.4

1 H NMR (DMSO-d6, 500 MHz) 6 1.34 (2H, dd, J = 4.0, 1.7 Hz), 1.62 (2H, dd, J = 4.2, 1.6 Hz), 2.83 (2H, s), 3.17 (1H, d, J = 2.6 Hz), 3.31 (3H+H2O, s), 3.49 (2H, d, J = 5.8 Hz), 3.71 (3H, s), 3.82 (3H, s), 4.03 (2H, s), 4.58 (2H, d, J = 5.6 Hz), 5.76 (1H, d, J = 2.8 Hz), 5.89 (1H, dd, J = 7.5, 2.8 Hz), 5.95 (1H, t, J = 6.0 Hz), 6.39 (1H, dd, J = 8.4, 2.4 Hz), 6.55 (1H, d, J = 2.4 Hz), 6.66 (1H, d, J = 7.7 Hz), 7.02 (1H, d, J = 8.4 Hz), 7.12 (1H, d, J = 6.2 Hz), 7.23 (1H, t, J = 8.0 Hz), 7.36 (1H, t, J = 5.8 Hz), 7.42 (1H, d, J = 8.4 Hz), 7.52 (1H, d, J = 7.6 Hz), 7.72 (1H, d, J = 6.0 Hz)

Intermediate 27

4-((1-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)a mino)methyl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l-methylpyridin-2(1H)-one 2-O-Benzyl 1-O-methyl 4-[(1-methyl-2-oxopyridin-4-yl)oxymethyl]-2-azabicyclo[2.1.1 ]hexane-l,2- dicarboxylate

According to General Method 2b, 2-O-benzyl 1-0-methyl 4-(methylsulfonyloxymethyl)-2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate (703 mg, 1.56 mmol) was reacted with 4-hydroxy-l-methyl-2- pyridone (195 mg, 1.56 mmol) and K2CO3 (646 mg, 4.68 mmol) in DMF (10 mL) at 90 °C for 17 h. After aqueous work up the residue was purified by flash chromatography (Silica, 0-9% MeOH in DCM) to afford the product (620 mg, 96% yield).

[M+H] + = 413.4

1 H NMR (DMSO-d6, 400 MHz) 6 1.74 (2H, dd, J = 4.6, 1.9 Hz), 2.10 (2H, dd, J = 4.7, 1.8 Hz), 3.32 (3H, s),

3.45 (2H, s), 3.57 (3H, s), 4.19 (2H, s), 5.06 (2H, s), 5.79 (1H, d, J = 2.8 Hz), 5.93 (1H, dd, J = 7.6, 2.8 Hz),

7.36 (5H, m), 7.56 (1H, d, J = 7.5 Hz)

Benzyl l-(hydroxymethyl)-4-[(1-methyl-2-oxopyridin-4-yl)oxymethyl]- 2-azabicyclo[2.1.1]hexane-2- carboxylate

According to a modification of General Method 3b, 2-O-Benzyl 1-O-methyl 4-[(1-methyl-2-oxopyridin-4- yl)oxymethyl]-2-azabicyclo[2.1.1]hexane-l,2-dicarboxylate (582 mg, 1.41 mmol) was reduced in the presence of lithium borohydride (IJBH4) (2M solution in THF) (776 μL, 1.55 mmol) over 72 h. Water (10 mL) and DCM (30 mL) were added. The organic layer was isolated and the aqueous layer re-extracted with DCM (3 x 10 mL). The combined organic layers were dried (MgSO4), filtered and concentrated to afford the product (464 mg, 86% yield).

[M+H] + = 385.3 1 H NMR (DMSO-d6, 400 MHz) 6 1.46 (2H, dd, J = 4.4, 1.8 Hz), 1.83 - 1.95 (2H, m), 3.33 (3H, s), 3.41 (2H, s), 3.90 (2H, d, J = 6.1 Hz), 4.17 (2H, s), 4.70 (1H, t, J = 6.2 Hz), 5.06 (2H, s), 5.80 (1H, d, J = 2.8 Hz), 5.93 (1H, dd, J = 7.5, 2.8 Hz), 7.27 - 7.36 (1H, m), 7.37 (4H, d, J = 3.7 Hz), 7.56 (1H, d, J = 7.6 Hz)

Benzyl-l-formyl-4-[(1-methyl-2-oxopyridin-4-yl)oxymethyl] -2-azabicyclo[2.1.1]hexane-2-carboxylate

According to General Method 16 benzyl l-(hydroxymethyl)-4-[(1-methyl-2-oxo-4-pyridyl)oxymethyl]-2- azabicyclo[2.1.1]hexane-2-carboxylate (1.20 g, 3.12 mmol) was oxidised. Aqueous work up afforded the product (2.00 g, 99% yield).

[M+H] + = 384.4

1 H NMR (DMSO-d6, 400 MHz) 6 1.73 (2H, dd, J = 4.4, 1.8 Hz), 2.05 (2H, d, J = 5.7 Hz), 3.32 (3H, s), 3.50 (2H, s), 4.20 (2H, s), 5.12 (2H, s), 5.80 (1H, d, J = 2.7 Hz), 5.94 (1H, dd, J = 7.5, 2.8 Hz), 7.27 - 7.44 (5H, m), 7.56 (1H, d, J = 7.6 Hz), 9.77 (1H, s)

Benzyl l-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-4-(((1-methyl-2-oxo-l,2- dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-2- carboxylate

Following General Method 17, benzyl l-formyl-4-[(1-methyl-2-oxo-4-pyridyl)oxymethyl]-2- azabicyclo[2.1.1]hexane-2-carboxylate (2.00 g, 60% purity, 3.14 mmol) was reacted with Nl-(2,4- dimethoxybenzyl)isoquinoline-l,5-diamine (0.97 g, 3.14 mmol) for 24h. After aqueous work up, the crude product was purified by flash chromatography (Silica, 0-30% MeOH in EtOAc) to afford the product (1.60 g, 67% yield).

[M+H] + = 676.8

1 H NMR (DMSO-d6, 400 MHz) 6 1.55 (2H, d, J = 3.1 Hz), 1.91 (2H, s), 3.31 (3H, s), 3.42 (2H, d, J = 6.9 Hz), 3.71 (3H, s), 3.83 (3H, s), 3.95 (2H, d, J = 6.1 Hz), 4.13 (2H, s), 4.59 (2H, d, J = 5.6 Hz), 5.12 (2H, s), 5.76 (1H, d, J = 2.7 Hz), 5.88 (1H, dd, J = 7.5, 2.8 Hz), 6.01 (1H, s), 6.39 (1H, dd, J = 8.4, 2.4 Hz), 6.55 (1H, d, J = 2.4 Hz), 6.73 (1H, d, J = 7.7 Hz), 6.88 (1H, d, J = 6.1 Hz), 7.02 (1H, d, J = 8.4 Hz), 7.24 (1H, t, J = 8.0 Hz), 7.29 - 7.42 (6H, m), 7.46 (1H, d, J = 8.4 Hz), 7.51 (1H, d, J = 7.6 Hz), 7.71 (1H, d, J = 6.1 Hz)

4-((1 _ (((1 _ ((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amino)met hyl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l-methylpyridin-2(1H)-one

Following a variation of General Method 3e, a solution of benzyl l-(((1-((2,4- dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-4-(((1- methyl-2-oxo-l,2-dihydropyridin-4- yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (1350 mg, 2.00 mmol) in MeOH (10 mL) was added to a mixture of Pd(OH)2/C (20 wt%) (338 mg, 0.481 mmol) in MeOH (10 mL). The mixture was subjected to hydrogenation at 1 atmosphere at rt for 66 h. The mixture was filtered over Celite® washing with MeOH (3 x 20 mL) and the filtrate was concentrated to afford the product (1100 mg, 95% yield). [M+H] + = 542.4

1 H NMR (DMSO-d6, 400 MHz) 6 1.41 (2H, s), 1.65 (2H, s), 2.87 (2H, s), 3.35 (3H, s), 3.44 (2H, d, J = 5.1 Hz), 3.71 (3H, s), 3.82 (3H, s), 4.14 (2H, s), 4.59 (2H, d, J = 5.4 Hz), 5.77 (1H, d, J = 2.7 Hz), 5.83 - 5.94 (2H, m), 6.39 (1H, dd, J = 8.4, 2.3 Hz), 6.55 (1H, d, J = 2.3 Hz), 6.68 (1H, d, J = 7.9 Hz), 7.02 (1H, d, J = 8.3 Hz), 7.08 (1H, d, J = 6.1 Hz), 7.25 (1H, s), 7.38 (1H, s), 7.45 (1H, d, J = 8.4 Hz), 7.52 (1H, d, J = 7.6 Hz), 7.73 (1H, d, J = 6.1 Hz) Intermediate 28

4-((1-(((1*Aminoisoquinolin-5-yl)amino)methyl)-2-azabicyc lo[2.1.1]hexan-4-yl)methoxy)-l- methylpyridin-2(1H)-one

According to General Method 8, 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)- 2-azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H) -one (553 mg, 0.935 mmol) was deprotected in DCM (7.0 mL) at rt for 18h. The reaction mixture was diluted with Et2O (25 mL), stirred for 15 min and the resulting precipitate filtered-off and triturated with Et2O (3 x 25 mL). The solid was diluted with MeOH (30 mL) and Amberlite IRN78 (10 mL) was added. The mixture was stirred for 2 h and filtered. The resin was washed with MeOH (100 mL). The filtrate was concentrated to afford the product (281 mg, 77% yield).

[M+H] + = 392.3

1 H NMR (DMSO-d6, 400 MHz) 6 1.33 - 1.43 (2H, m), 1.57 - 1.67 (2H, m), 2.54 (1H, s), 2.83 (2H, s), 3.31 (3H, s), 3.40 (2H, d, J =5.4 Hz), 4.14 (2H, s), 5.74 - 5.81 (2H, m), 5.89 (1H, dd, J = 7.5, 2.8 Hz), 6.48 (2H, s), 6.64 (1H, dd, J = 7.7, 0.9 Hz), 7.10 (1H, dd, J= 6.3, 0.9 Hz), 7.20 (1H, t, J = 8.0 Hz), 7.32 (1H, d, J = 8.3 Hz), 7.52 (1H, d, J = 7.6 Hz), 7.72 (1H, d, J = 6.1 Hz)

Intermediate 29

4-((1-(((1-((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)a mino)methyl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l,6-dimethylpyridin-2(1H)-one tert-Butyl 4-(((1,6-dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)- l-formyl-2- azabicyclo[2.1.1]hexane-2-carboxylate

Following General Method 16, tert-butyl 4-[(1,2-dimethyl-6-oxo-4-pyridyl)oxymethyl]-l- (hydroxymethyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (6500 mg, 17.8 mmol) was oxidised. Aqueous work up afforded the product (6.28 g, 78% yield, 80% pure).

1 H NMR (CDCI 3 , 400 MHz) 6 1.46 (9H, s), 1.78 (2H, dd, J = 4.6, 1.9 Hz), 2.11 (2H, dd, J = 4.6, 1.8 Hz), 2.30 (3H, s), 3.45 (3H, s), 3.47 (2H, s), 4.06 (2H, s), 5.76 (1H, dt, J = 2.8, 0.9 Hz), 5.83 (1H, d, J = 2.8 Hz), 9.83 (1H, s) tert-Butyl l-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-4-(((1,6-dimethyl-2- oxo-1, 2-dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane- 2-carboxyl ate

According to General Method 17, tert-butyl 4-[(1,2-dimethyl-6-oxo-4-pyridyl)oxymethyl]-l-formyl-2- azabicyclo[2.1.1]hexane-2-carboxylate (6.28 g, 13.9 mmol) was reacted with Nl-[(2,4- dimethoxyphenyl)methyl]isoquinoline-l,5-diamine (4.99 g, 15.3 mmol) for 72 h. The reaction was quenched with a mixture of sat. NaHCO3 (aq) and brine (1:1 v/v, 40 mL). The mixture was stirred for 30 min at rt and the phases were separated. The aqueous phase was extracted with DCM (3 x 40 mL), and the combined organic layers were dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (Silica, 0-9% MeOH in EtOAc) afforded the product (2.90 g, 63% yield).

[M+H] + = 655.8

1 H NMR (DMSO-d6, 400 MHz) 6 1.44 (3H, s), 1.48 (10H, s), 1.53 (2H, d, J = 4.9 Hz), 1.91 (3H, s), 2.03 (1H, s), 2.29 (3H, s), 2.33 (6H, t, J = 0.7 Hz), 3.34 (3H, s), 3.36 (8H, d, J = 2.7 Hz), 3.73 (4H, s), 3.75 (3H, s), 3.87 (3H, s), 3.95 (2H, d, J = 5.9 Hz), 4.13 (2H, s), 4.63 (2H, d, J = 5.4 Hz), 5.67 - 5.78 (3H, m), 5.84 - 5.96 (3H, m), 6.07 (1H, s), 6.43 (1H, dd, J = 8.4, 2.4 Hz), 6.59 (1H, d, J = 2.4 Hz), 6.77 (1H, d, J = 7.9 Hz), 6.99 (1H, d, J = 6.2 Hz), 7.07 (1H, d, J = 8.3 Hz), 7.30 (1H, t, J = 8.0 Hz), 7.45 (1H, d, J = 5.9 Hz), 7.51 (1H, d, J = 8.4 Hz), 7.77 (1H, d, J = 6.1 Hz)

4-((1 _ (((1 _ ((2,4-Dimethoxybenzyl)amino)isoquinolin-5-yl)amino)met hyl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l,6-dimethylpyridin-2(1H)-one

According to a variation of General Method 5a, HCI (4M in 1,4-dioxane, 26.7 mL, 107 mmol) was added to a mixture of tert-butyl l-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-4-(((1,6- dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)-2-azabicy clo[2.1.1]hexane-2-carboxylate (7.00 g, 10.7 mmol) in anhydrous THF (55 mL) at 0 °C. The mixture was warmed to rt, stirred for 18 h and filtered washing with Et2O (5 x 25 mL) to afford the product as an HCI salt. The resulting solid was taken up in a mixture of DCM and MeOH (9:1 v/v, 200 mL) and sat. NaHCO3 (aq) (125 mL, pH 9). The phases were separated, and the aqueous phase was extracted with a mixture of DCM and MeOH (9:1, 2 x 150 mL). The combined organic phases were dried (Na2SO4 , filtered, and concentrated. Purification by flash chromatography (Silica, 0-20% (5% Et3N in MeOH)) in DCM) afforded the product (3.30 g, 53% yield). [M+H] + = 556.8 1 H NMR (DMSO-d6, 400 MHz) 6 1.41 (2H, s), 1.66 (2H, d, J = 3.8 Hz), 2.30 (3H, s), 2.87 (2H, s), 3.20 (1H, d, J = 5.0 Hz), 3.34 (3H, s), 3.38 (2H, s), 3.45 (2H, d, J = 5.3 Hz), 3.75 (3H, s), 3.86 (3H, s), 4.15 (2H, s), 4.62 (2H, d, J = 5.4 Hz), 5.72 (1H, d, J = 2.8 Hz), 5.89 (1H, dd, J = 2.8, 0.9 Hz), 5.93 (1H, t, J = 5.6 Hz), 6.42 (1H, dd, J = 8.4, 2.4 Hz), 6.59 (1H, d, J = 2.4 Hz), 6.70 (1H, d, J = 7.8 Hz), 7.05 (1H, d, J = 8.3 Hz), 7.12 (1H, d, J = 6.1 Hz), 7.28 (1H, t, J = 8.0 Hz), 7.45 (1H, d, J = 5.9 Hz), 7.49 (1H, d, J = 8.5 Hz), 7.76 (1H, d, J = 6.1 Hz)

Intermediate 30

4-((1-(((1*Aminoisoquinolin-5-yl)amino)methyl)-2-azabicyc lo[2.1.1]hexan-4-yl)methoxy)-l,6- dimethylpyridin-2(1H)-one

4-((1-(((1*Aminoisoquinolin-5-yl)amino)methyl)-2-azabicyc lo[2.1.1]hexan-4-yl)methoxy)-l,6- dimethylpyridin-2(1H)-one

According to General Method 8, 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)- 2-azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2 (1H)-one (1.51 g, 12.58 mmol) was deprotected in DCM (25 mL) at rt for 18 h. The mixture was diluted with Et2O (75 mL), stirred for 15 min and the precipitate was isolated by filtration and triturated with Et2O (3 x 25 mL). The solid was taken up in MeOH (25 mL) and Amberlite IRN78 (20 mL) was added. The mixture was stirred for 2 h and filtered. The resin was washed with MeOH (100 mL) and the filtrate concentrated to afford the product (889 mg, 85% yield).

[M+H] + = 406.2

1 H NMR (DMSO-d6, 400 MHz) 6 1.32 - 1.43 (2H, m), 1.58 - 1.64 (2H, m), 2.26 (3H, s), 2.55 (1H, t, J = 6.2 Hz), 2.82 (2H, d, J = 5.3 Hz), 3.30 (3H, s), 3.40 (2H, d, J = 5.5 Hz), 4.11 (2H, s), 5.68 (1H, d, J = 2.8 Hz), 5.80 (1H, t, J = 5.6 Hz), 5.85 (1H, dd, J = 2.8, 0.9 Hz), 6.50 (2H, s), 6.64 (1H, d, J = 7.7 Hz), 7.11 (1H, d, J = 6.1 Hz), 7.20 (1H, t, J = 8.0 Hz), 7.33 (1H, d, J = 8.3 Hz), 7.73 (1H, d, J = 6.1 Hz)

Intermediate 31

N5-((4-(((tert-Butyldiphenylsilyl)oxy)methyl)-2-azabicycl o[2.1.1]hexan-l-yl)methyl)-Nl-(2,4- dimethoxybenzyl)isoquinoline-l,5-diamine tert-Butyl 4-(((tert-butyldiphenylsilyl)oxy)methyl)-l-(hydroxymethyl)-2 -azabicyclo[2.1.1]hexane-2- carboxylate

According to a variation of General Method 3b, 2-(tert-butyl) 1-methyl 4-(((tert- butyldiphenylsilyl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-l,2 -dicarboxylate (90%, 59.5 g, 105 mmol) was reduced using LiBH4 (8.00 g, 330 mmol). The mixture was cooled to 0 °C and quenched with sat. NH4CI (aq) (350 mL), H2O (500 mL), and EtOAc (500 mL). The layers were separated, and the aqueous layer was extracted with EtOAc (2 x 500 mL). The combined organic layers were dried (Na2SO4 , filtered, and concentrated to afford the product (51.1 g, 91% yield).

[M-tBu+H] + = 426.4

1 H NMR (CDCI3, 300 MHz) 6 1.06 (9H, s), 1.47 (9H, s), 3.25 (2H, s), 3.80 (2H, s), 3.91 (2H, s), 7.32 - 7.47 (6H, m), 7.57 - 7.68(4H, m) tert-Butyl 4-(((tert-butyldiphenylsilyl)oxy)methyl)-l-(((1-((2,4-dimeth oxybenzyl)amino)isoquinolin-5- yl)amino)methyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate

Following General Method 16, tert-butyl 4-(((tert-butyldiphenylsilyl)oxy)methyl)-l-(hydroxymethyl)-2 - azabicyclo[2.1.1]hexane-2-carboxylate (7.00 g, 13.1 mmol, 90% purity) was oxidised for 16 h. Aqueous work up afforded the intermediate tert-butyl 4-(((tert-butyldiphenylsilyl)oxy)methyl)-l-formyl-2- azabicyclo[2.1.1]hexane-2-carboxylate which was reacted directly with Nl-(2,4- dimethoxybenzyl)isoquinoline-l,5-diamine (4.50 mg, 14.5 mmol) following General Method 17 for 16 h. Aqueous work up and purification by flash chromatography (Silica, 0-52% EtOAc in hexanes) afforded the product (4.05 g, 40% yield).

[M+H] + = 774.8

1 H NMR (DMSO-d6, 300 MHz) 6 0.93 (9H, s), 1.41 (11H, s), 1.78 (2H, s), 3.22 (2H, s), 3.70 (3H, s), 3.78 (2H, s), 3.82 (3H, s), 3.88 (2H, d, J = 5.5 Hz), 4.59 (2H, d, J = 5.6 Hz), 5.96 (1H, s), 6.36 (1H, d, J = 8.4 Hz), 6.55 (1H, d, J = 2.3 Hz), 6.71 (1H, d, J = 7.8 Hz), 6.95 (1H, d, J = 6.4 Hz), 7.02 (1H, d, J = 8.4 Hz), 7.26 (1H, t, J = 8.0 Hz), 7.34 - 7.49 (8H, m), 7.53 (4H, d, J = 7.6 Hz), 7.72 (1H, d, J = 6.1 Hz)

N5-((4-(((tert-Butyldiphenylsilyl)oxy)methyl)-2-azabicycl o[2.1.1]hexan-l-yl)methyl)-Nl-(2,4- dimethoxybenzyl)isoquinoline-l,5-diamine Following General Method 5a, tert-butyl 4-(((tert-butyldiphenylsilyl)oxy)methyl)-l-(((1-((2,4- dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-2-azabi cyclo[2.1.1]hexane-2-carboxylate (4.05 g, 4.72 mmol) was deprotected in THF (30 mL) at rt for 72 h. After concentration and trituration with Et2O, the residue was diluted with NaOH (aq) (IN, 100 mL) and 1:8 MeOH:DCM (100 mL). The layers were separated and extracted with 1:8 MeOH/DCM (3 x 75 mL). The combined organic layers were washed with brine (200 mL), dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (Silica, 0- 10% (10% NH4OH in MeOH) in DCM) afforded the product (1.3 g, 41% yield).

[M+H] + = 673.5

1 H NMR (DMSO-d6, 300 MHz) 6 0.97 (9H, s), 1.29 - 1.37 (2H, m), 1.49 - 1.57 (2H, m), 2.77 (2H, s), 3.39 (2H, d,J = 5.3 Hz), 3.71 (3H, s), 3.83 (5H, s), 4.59 (2H, d, J = 5.4 Hz), 5.72 - 5.77 (1H, m), 6.34 - 6.42 (1H, m), 6.55(1H, d, J = 2.0 Hz), 6.67 (1H, d, J = 7.8 Hz), 7.03 (1H, d, J = 8.3 Hz), 7.07 (1H, d, J = 6.0 Hz), 7.25 (1H, t, J =8.1 Hz), 7.35 - 7.49 (8H, m), 7.54 - 7.61 (4H, m), 7.73 (1H, d, J = 6.0 Hz)

Intermediate 32

4-(((1,6-Dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methy l)-2-(5-methoxybenzo[d]oxazol-2-yl)-2- azabicyclo[2.1.1]hexane-l-carbaldehyde

4-((1-(Hydroxymethyl)-2-azabicyclo[2.1.1]hexan-4-yl)metho xy)-l,6-dimethylpyridin-2(1H)-one Following general Method 5a, tert-butyl 4-[(1,2-dimethyl-6-oxopyridin-4-yl)oxymethyl]-l- (hydroxymethyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (2.10 g, 5.76 mmol) was deprotected at rt for 18 h. Concentration afforded the product (1.70 g, 98% yield).

[M+H] + = 265.1

4-((1-(Hydroxymethyl)-2-(5-methoxybenzo[d]oxazol-2-yl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6- dimethylpyridin-2(1H)-one

According to General Method 2b, 4-[[l-(hydroxymethyl)-2-azabicyclo[2.1.1]hexan-4-yl]methoxy] -l,6- dimethylpyridin-2-one hydrochloride (1.50 mg, 4.99 mmol) was reacted with 2-chloro-5- methoxybenzo[d]oxazole (1.10 mg, 5.98 mmol) in DMSO (3 mL) using K2CO3 (2.76 mg, 19.95 mmol), at rt for 3 days. The solvent was removed (Biotage V10) and the residue was partitioned between DCM (50 mL) and water (50 mL). The organic phase was dried (MgSO4) and concentrated to afford the product (2.00 g, 97% yield).

[M+H] + = 412.2

4-(((1,6-Dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methy l)-2-(5-methoxybenzo[d]oxazol-2-yl)-2- azabicyclo[2.1.1]hexane-l-carbaldehyde Following General Method 16, 4-[[l-(hydroxymethyl)-2-(5-methoxy-l,3-benzoxazol-2-yl)-2- azabicyclo[2.1.1]hexan-4-yl]methoxy]-l,6-dimethylpyridin-2-o ne (200 mg, 0.49 mmol) was oxidised. After quenching with 1:1 sat. Na2S2O3 (aq) (10 mL) and sat. NaHCO3 (aq) (10 mL), aqueous work up afforded the product (195 mg, 98% yield).

[M+H] + = 410.1

Intermediate 33

Nl-[(2,4-Dimethoxyphenyl)methyl]-N5-methyl-isoquinoline-l ,5-diamine

Copper(ll) acetate (304 mg, 1.59 mmol) was added to a solution of Nl-[(2,4- dimethoxyphenyl)methyl]isoquinoline-l,5-diamine (249 mg, 0.645 mmol) and pyridine (180 μL, 2.21 mmol) in 1,4-dioxane (8 mL) at rt. The mixture was stirred for 10 min at rt and methylboronic acid (98.8 mg, 1.62 mmol) was added. The mixture was heated at reflux for 18 h, cooled to rt, filtered over Celite® and washed with EtOAc (3 x 20 mL). The filtrate was concentrated, and the residue was purified by flash chromatography (silica, 0-88% EtOAc in hexanes) to afford the product (127 mg, 61%).

[M+H] + = 324.2

1 H NMR (DMSO-d6, 400 MHz) 6 2.81 (3H, d, J = 4.8 Hz), 3.71 (3H, s), 3.82 (3H, s), 4.59 (2H, d, J = 5.6 Hz), 6.12 (1H, q, J = 4.9 Hz), 6.39 (1H, dd, J = 8.3, 2.4 Hz), 6.52 - 6.58 (2H, m), 7.02 (1H, d, J = 2.7 Hz), 7.04 (1H, d, J = 0.8 Hz), 7.28 (1H, t, J = 8.0 Hz), 7.36 (1H, t, J = 5.9 Hz), 7.44 (1H, d, J = 8.4 Hz), 7.72 (1H, d, J = 6.0 Hz)

Specific Examples of the Present Invention

Example number 1

N5-((4-(5-Phenyloxazol-2-yl)-2-oxabicyclo[2.1.1]hexan-l-y l)methyl)isoquinoline-l,5-diamine tert-Butyl ((4-(hydroxymethyl)-2-oxabicyclo[2.1.1]hexan-l-yl)methyl)car bamate

To a solution of [l-(aminomethyl)-2-oxabicyclo[2.1.1]hexan-4-yl]methanol hydrochloride (1.26 g, 7.01 mmol) and TEA (2.50 mL, 17.94 mmol) in THF (35 mL) at 0 °C was added BOC2O (2.00 g, 9.16 mmol) and the mixture was stirred for 10 min at 0 °C. The reaction was then stirred for 18 h at rt. The reaction was quenched with water (30 mL) and washed with EtOAc (2 x 50 mL), dried (MgSO4) and concentrated. Purification by flash chromatography (Silica, 0-20% MeOH in DCM) afforded the product (1.29g 75% yield).

[M+Na] + = 266.2

1 H NMR (DMSO-d6, 400 MHz) 6 1.26 - 1.34 (2H, m), 1.37 (9H, s), 1.52 - 1.66 (2H, m), 3.18 (2H, d, J = 6.1 Hz), 3.51 (2H, s), 3.61 (2H, d, J = 5.4 Hz), 4.62 (1H, t, J = 5.5 Hz), 6.84 (1H, t, J = 6.1 Hz) l-(((tert-Butoxycarbonyl)amino)methyl)-2-oxabicyclo[2.1.1]he xane-4-carboxylic acid

To a mixture of tert-butyl N-[[4-(hydroxymethyl)-2-oxabicyclo[2.1.1]hexan-l-yl]methyl]c arbamate (500 mg, 2.06 mmol) and NaICU (1758 mg, 8.22 mmol) in CCI4 (4mL), MeCN (4 mL) and H2O (6 mL) was added RUCI3.H2O (15 mg, 0.07 mmol). The mixture was stirred at rt for 2 h and diluted with EtOAc (25 mL) and sat. NH4CI (aq) (25 mL). The aqueous phase was extracted with EtOAc (3 x 20 mL) and the combined organic phases were washed brine (50 mL), dried (Na2SO4), filtered and concentrated. The residue was diluted in EtOAc (10 mL) and filtered on a Florisil pad. The filtrate was concentrated to afford the product (390 mg, 74% yield).

1 H NMR (DMSO-d6, 300 MHz) 6 1.38 (9H, s), 1.61 (2H, d, J = 4.2 Hz), 1.92 (2H, d, J = 4.0 Hz), 3.20 (2H, d, J = 6.0 Hz), 3.74(2H, s), 6.92 (1H, t, J = 5.9 Hz), 12.66 (1H, s)

Tert-butyl ((4-((2-oxo-2-phenylethyl)carbamoyl)-2-oxabicyclo[2.1.1]hexa n-l-yl)methyl)carbamate According to General Method 18b, l-[(tert-butoxycarbonylamino)methyl]-2-oxabicyclo[2.1.1]hexa ne-4- carboxylic acid (130 mg, 0.51 mmol) was coupled to 2-amino-l-phenyl-ethanone hydrochloride (87 mg, 0.51 mmol). After aqueous work up, purification by flash chromatography (Silica, 0-90% EtOAc in hexanes) afforded the product (137 mg, 72% yield).

[M-H]’ 373.4

1 H NMR (CDCI3, 300 MHz) 6 1.45 (9H, s), 1.95 (2H, d, J = 4.4 Hz), 2.05 (2H, d, J = 4.5 Hz), 3.44 - 3.54 (2H, m), 3.97 (2H, s),4.72 - 4.86 (3H, m), 6.70 (1H, s), 7.52 (2H, t, J = 7.6 Hz), 7.65 (1H, t, J = 7.3 Hz), 7.99 (2H, d, J = 7.6 Hz)

(4-(5-Phenyloxazol-2-yl)-2-oxabicyclo[2.1.1]hexan-l-yl)me thanamine

A mixture of tert-butyl N-[[4-(phenacylcarbamoyl)-2-oxabicyclo[2.1.1]hexan-l-yl]meth yl]carbarriate (120 mg, 0.32 mmol) in IM H2SO4 (aq) (1 mL) was stirred at rt for 1 h. The mixture was basified with a solution of IM NH4OH (aq) and extracted with EtOAc (3 x 10 mL). The combined organic layers were dried (Na2SO4), filtered and concentrated to afford the product (52mg, 38% yield).

[M+H] + 257.3

N5-((4-(5-Phenyloxazol-2-yl)-2-oxabicyclo[2.1.1]hexan-l-y l)methyl)isoquinoline-l,5-diamine

Following a variation on General Method 4, [4-(5-phenyloxazol-2-yl)-2-oxabicyclo[2.1.1]hexan-l- yl]methanamine (50 mg, 0.12 mmol, 60% pure) was coupled to 5-bromo-N-(2,4- dimethoxybenzyl)isoquinolin-l-amine (53mg, 0.14 mmol) in the presence of NaO l Bu (30 mg, 0.31 mmol) and Pd-175 (15 mg, 0.02 mmol) in 1,4-dioxane at 100 °C for 16 h. After cooling to rt, the reaction mixture was filtered over Celite® washing with EtOAc (3 x 25 mL) and the filtrate was concentrated to afford the intermediate Nl-[(2,4-dimethoxyphenyl)methyl]-N5-[[4-(5-phenyloxazol-2-yl )-2- oxabicyclo[2.1.1]hexan-l-yl]methyl]isoquinoline-l,5-diamine.

[M+H] + = 549.3

Deprotection was performed according to a variation of General Method 8, in DCM (2 mL) at rt for 3 h. After concentration and aqueous work up, purification by flash chromatography (C18, 10-60% MeCN in 10 mM NH4HCO3 (aq)), followed by automated prep HPLC (mass directed, 42-52% MeCN in 10 mM NH4HCO3 (aq) over 9 min) afforded the product (11 mg, 24% yield).

[M+H] + = 399.3

1 H NMR (DMSO-d6, 300 MHz) 6 1.94 (2H, d, J = 4.2 Hz), 2.32 (2H, d, J = 4.4 Hz), 3.63 (2H, d, J = 5.4 Hz), 4.06 (2H, s), 6.00(1H, t, J = 5.8 Hz), 6.51 (2H, s), 6.74 (1H, d, J = 7.7 Hz), 7.14 (1H, d, J = 6.1 Hz), 7.23 (1H, t, J = 7.9 Hz), 7.32 - 7.40 (2H, m), 7.41 -7.50 (2H, m), 7.63 (1H, s), 7.66 - 7.77 (3H, m)

Example number 15

4-((1-(((1-Aminoisoquinolin-5-yl)amino)methyl)-2-(2-(chro man-6-yl)acetyl)-2-azabicyclo[2.1.1]hexan-

4-yl)methoxy)-l-methylpyridin-2(1H)-one

According to General Method 18a, 2-chroman-6-ylacetic acid (28 mg, 0.14 mmol) was coupled to 4-((1- (((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)meth yl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l-methylpyridin-2(1H)-one (61 mg, 0.11 mmol). Concentratration afforded 4-((2-(2- (chroman-6-yl)acetyl)-l-(((1-((2,4-dimethoxybenzyl)amino)iso quinolin-5-yl)amino)methyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H)-o ne which was deprotected according to General Method 8 in DCM (1 mL) at rt for 18 h. Purification by flash chromatography (C18, 20-47% MeCN in 10 mM (NH4)HCO3 (aq)) provided the product (41 mg, 65% yield).

[M+H] + = 566.6

1 H NMR (DMSO-d6, 500 MHz) 6 1.47 - 1.53 (2H, m), 1.87 - 1.94 (4H, m), 2.69 (2H, t, J = 6.4 Hz), 3.31 (3H, s), 3.52 (2H, s), 3.56 (2H, s), 3.98 (2H, d, J = 6.6 Hz), 4.07 - 4.13 (2H, m), 4.14 (2H, s), 5.78 (1H, d, J = 2.7 Hz), 5.88 (1H, dd, J = 7.5, 2.8 Hz), 6.16 (1H, t, J = 6.6 Hz), 6.51 (2H, d, J = 5.3 Hz), 6.66 (1H, d, J = 8.1 Hz), 6.79 (2H, t, J = 7.5 Hz), 6.97 (2H, d, J = 8.9 Hz), 7.21 (1H, t, J = 8.0 Hz), 7.33 (1H, d, J = 8.3 Hz), 7.54 (1H, d, J = 7.5 Hz), 7.70 (1H, d, J = 6.1 Hz)

Example numbers 24 and 25

4-((1-(((1-Aminoisoquinolin-5-yl)amino)methyl)-2-(2-trans -(-4-methoxycyclohexyl)acetyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2(1 H)-one

And

4-((1-(((1-aminoisoquinolin-5-yl)amino)methyl)-2-(2-cis-( 4-methoxycyclohexyl)acetyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2(1 H)-one

According to General Method 18a, 2-(4-methoxycyclohexyl)acetic acid (55 mg, 0.30 mmol) was coupled with 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l-methylpyridin-2(1H)-one (154 mg, 0.26 mmol). Concentration afforded 4-((1-(((1-((2,4- dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-2-(2-(4 -methoxycyclohexyl)acetyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H)-o ne which was deprotected according to General Method 8 in DCM (4 mL) at rt for 18 h. Purification by flash chromatography (C18, 20-42% MeCN in 10 mM (NH4)HCO3 (aq)) followed by automated prep. HPLC (mass directed, 30-40% MeCN in 10 mM (NH4)HCO3 (aq) over 9 min) provided the product 4-((1-(((1-aminoisoquinolin-5-yl)amino)methyl)-2- (2-trans-(-4-methoxycyclohexyl)acetyl)-2-azabicyclo[2.1.1]he xan-4-yl)methoxy)-l,6-dimethylpyridin- 2(1H)-one (46 mg, 31% yield) as the first eluting isomer.

[M+H] + = 560.6

1 H NMR (DMSO-d6, 400 MHz) 6 0.93 - 1.17 (4H, m), 1.45 - 1.50 (2H, m), 1.71 - 1.81 (3H, m), 1.84 - 1.90 (2H, m), 1.93 - 2.01 (2H, m), 2.15 (2H, d, J = 6.5 Hz), 2.26 (3H, s, 2.99 - 3.08 (1H, m), 3.22 (3H, s), 3.31 (3H, s), 3.46 (2H, s), 3.97 (2H, d, J = 6.5 Hz), 4.10 (2H, s), 5.68 (1H, d, J = 2.8 Hz), 5.84 (1H, dd, J = 2.8, 0.9 Hz), 6.20 (1H, t, J = 6.8 Hz), 6.52 (2H, s), 6.76 - 6.84 (2H, m), 7.20 (1H, t, J = 8.0 Hz), 7.32 (1H, d, J = 8.3 Hz), 7.70 (1H, d, J = 6.1 Hz) and 4-((1-(((1-aminoisoquinolin-5-yl)amino)methyl)-2-(2-cis-(4-m ethoxycyclohexyl)acetyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2(1 H)-one (18 mg, 12% yield) as the second eluting iomer.

[M+H] + = 560.7

1 H NMR (DMSO-d6, 500 MHz) 6 1.22 - 1.32 (2H, m), 1.37 - 1.51 (6H, m), 1.73 - 1.82 (2H, m), 1.83 - 1.92 (3H, m), 2.15 (2H, d, J = 7.0 Hz), 2.26 (3H, s), 3.19 (3H, s), 3.31 (3H, s), 3.34 - 3.38 (1H, m), 3.48 (2H, s), 3.98 (2H, d, J = 6.6 Hz), 4.10 (2H, s), 5.68 (1H, d, J = 2.8 Hz), 5.84 (1H, dd, J = 2.8, 0.9 Hz), 6.23 (1H, t, J = 6.7 Hz), 6.60 (2H, s), 6.78 - 6.86 (2H, m), 7.22 (1H, t, J = 8.0 Hz), 7.33 (1H, d, J = 8.3 Hz), 7.70 (1H, d, J = 6.0 Hz)

Example number 29

N-(1-(1-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((1,6 -dimethyl-2-oxo-l,2-dihydropyridin-4- yl)oxy)methyl)-2-azabicyclo[2.1.1]hexan-2-yl)-l-oxopropan-2- yl)acetamide According to General Method 4, 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)- 2-azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2 (1H)-one (60 mg, 0.11 mmol) was coupled to 5-bromo-2,4-dimethyl-oxazole (24 mg, 0.13 mmol) using NaOtBu (27 mg, 0.27 mmol) and Pd-PEPPSI- IPent (9 mg, 0.01 mmol) in 1,4-dioxane (1 mL) at reflux for 24 h. 5-Bromo-2,4-dimethyl-oxazole (23.5 mg, 0.130 mmol) and Pd-PEPPSI-IPent (9 mg, 0.0108 mmol) were added and the mixture was refluxed for 48 h, cooled to rt, diluted with MeOH (500 μL), filtered washing with MeOH (5 mL) and concentrated to afford N-(1-(1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)am ino)methyl)-4-(((1,6-dimethyl-2- oxo-1, 2-dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexan-2 -yl)-l-oxopropan-2-yl)acetamide which was deprotected according to General Method 8 in DCM (2 mL) at rt for 18 h. Flash chromatography (C18, 20-60% MeCN in 10 mM (NH4)HCO3 (aq)) followed by an automated prep HPLC (UV directed 30-40% MeCN in 10 mM (NH4)HCO3 (aq) over 8 min) provided the product (24 mg, 45% yield).

[M+H] + = 518.8

1 H NMR (DMSO-d6, 400 MHz) 6 1.27 (3H, d, J = 6.9 Hz), 1.53 (1H, d, J = 3.4 Hz), 1.89 (3H, s), 1.92 (2H, t, J = 5.9 Hz), 2.30 (3H, s), 3.34 (4H, s), 3.57 (1H, d, J = 7.4 Hz), 3.72 (1H, d, J = 7.5 Hz), 3.94 - 4.11 (2H, m), 4.17 (2H, s), 4.47 (1H, t, J = 7.1 Hz), 5.73 (1H, d, J = 2.8 Hz), 5.89 (1H, dd, J = 2.9, 0.9 Hz), 6.16 (1H, t, J = 6.5 Hz), 6.56 (2H, s), 6.82 (1H, d, J = 7.7 Hz), 6.87 (1H, d, J = 6.2 Hz), 7.25 (1H, t, J = 8.0 Hz), 7.37 (1H, d, J = 8.0 Hz), 7.74 (1H, d, J = 8.0 Hz) 8.24 (1H, d, J = 8.0 Hz)

Example number 30 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((1-methyl-2-o xo-l,2-dihydropyridin-4-yl)oxy)methyl)-

N-(1-phenylcyclopropyl)-2-azabicyclo[2.1.1]hexane-2-carbo xamide According to General Method 21a, 1-phenylcyclopropanamine (20 μL, 0.16 mmol) was reacted with CDI (25.8 mg, 0.151 mmol) and 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H)-o ne (60 mg, 0.10 mmol). The mixture was concentrated to afford l-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-4-(((1- methyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)-N-(1-phenyl cyclopropyl)-2-azabicyclo[2.1.1]hexane-2- carboxamide which was deprotected according to General Method 8 in DCM (1 mL) at rt for 18 h. Flash chromatography (C18, 10-40% MeCN in 10 mM (NH4)HCO3 (aq)) afforded the product (33 mg, 61% yield).

[M+H] + = 552.4

1 H NMR (DMSO-d6, 500 MHz) 6 1.13 - 1.22 (4H, m), 1.45 - 1.50 (2H, m), 1.78 - 1.83 (2H, m), 3.31 (3H, s), 3.36 (2H, s), 3.90 (2H, d, J = 6.4 Hz), 4.15 (2H, s), 5.78 (1H, d, J = 2.8 Hz), 5.89 (1H, dd, J = 7.5, 2.7 Hz), 6.19 (1H, t, J = 6.5 Hz), 6.55 (2H, s), 6.75 (1H, d, J = 7.7 Hz), 6.79 (1H, d, J = 6.2 Hz), 7.13 - 7.32 (8H, m), 7.53 (1H, d, J = 7.6 Hz), 7.64 (1H, d, J = 6.1 Hz)

Example number 59 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((5-oxo-l,2,3, 5-tetrahydroindolizin-7-yl)oxy)methyl)-N- (1-phenylcyclopropyl)-2-azabicyclo[2.1.1]hexane-2-carboxamid e

2-(tert-Butyl) 1-methyl 4-(((5-oxo-l,2,3,5-tetrahydroindolizin-7-yl)oxy)methyl)-2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate

According to General Method 2b, 2-O-tert-butyl 1-O-methyl 4-(methylsulfonyloxymethyl)-2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate (300 mg, 0.86 mmol) was reacted with 7-hydroxy-2,3- dihydro-lH-indolizin-5-one (143 mg, 0.94 mmol) in DMF (3 mL) using K2CO3 (356 mg, 2.58 mmol) at 60 °C for 20 h. Aqueous work up afforded the product (356 mg, 95% yield).

[M+H] + = 405.3 tert-Butyl l-(hydroxymethyl)-4-(((5-oxo-l,2,3,5-tetrahydroindolizin-7-y l)oxy)methyl)-2- azabicyclo[2.1.1]hexane-2-carboxylate

2-(tert-Butyl) 1-methyl 4-(((5-oxo-l,2,3,5-tetrahydroindolizin-7-yl)oxy)methyl)-2- azabicyclo[2.1.1]hexane-l,2-dicarboxylate (356 mg, 0.82 mmol) was reduced using LiBI- (2M in THF, 700 μL, 1.4 mmol) in anhydrous THF (5 mL) at rt for 4 days. Water (10 mL) and DCM (20 mL) were added. The organic layer was separated and the aqueous layer washed with DCM (2 x 10 mL). The combined organic layers were dried (MgSO4), filtered and concentrated to afford the product (320 mg, 88% yield). [M+H] + = 377.2 tert-Butyl l-formyl-4-(((5-oxo-l,2,3,5-tetrahydroindolizin-7-yl)oxy)met hyl)-2-azabicyclo[2.1.1]hexane-

2-carboxylate

According to General Method 16, tert-butyl l-(hydroxymethyl)-4-[(5-oxo-2,3-dihydro-lH-indolizin-7- yl)oxymethyl]-2-azabicyclo[2.1.1]hexane-2-carboxylate (376 mg, 0.85 mmol) was oxidised over 48 h. Aqueous work up afforded the product (271 mg, 85% yield).

[M+H] + = 375.2 tert-Butyl l-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-4-(((5-oxo-l,2,3,5- tetrahydroindolizin-7-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexa ne-2-carboxylate

According to General Method 17, tert-butyl l-formyl-4-[(5-oxo-2,3-dihydro-lH-indolizin-7- yl)oxymethyl]-2-azabicyclo[2.1.1]hexane-2-carboxylate (271 mg, 0.72 mmol) was reacted with l-N-[(2,4- dimethoxyphenyl)methyl]isoquinoline-l,5-diamine (335 mg, 1.08 mmol) for 20 h. Aqueous work up and purification by flash chromatography (Silica, 0-100% EtOAc in Pet. Ether, followed by 0-20% MeOH in EtOAc) afforded the product (403 mg, 84% yield).

[M+H] + = 668.4 7-((1-(((1-Aminoisoquinolin-5-yl)amino)methyl)-2-azabicyclo[ 2.1.1]hexan-4-yl)methoxy)-2,3- dihydroindolizin-5(1H)-one tert-Butyl l-(((1-((2, 4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-4-((( 5-oxo-l, 2,3,5- tetrahydroindolizin-7-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexa ne-2-carboxylate (100 mg, 0.15 mmol) was deprotected according to General Method 8 at rt for 60 min to afford the product (55 mg, 70% yield). [M+H] + = 418.2

N-(1-Phenylcyclopropyl)-lH-imidazole-l-carboxamide

To a solution of 1-phenylcyclopropan-l-amine (500 mg, 3.75 mmol) and DBU (95 μL, 0.64 mmol) in dry DCM (10 mL) was added CDI (621 mg, 3.83 mmol) and stirred at rt for 1 h. The mixture was quenched with water (50 mL), extracted with DCM (2 x 50 mL), washed with brine (50 mL), dried (MgSO4) and concentrated to afford the product (570 mg, 53% yield).

[M+H]+ = 228.1 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((5-oxo-l,2,3, 5-tetrahydroindolizin-7-yl)oxy)methyl)-N- (1-phenylcyclopropyl)-2-azabicyclo[2.1.1]hexane-2-carboxamid e

To a solution of N-(1-phenylcyclopropyl)imidazole-l-carboxamide (45 mg, 0.2 mmol) and 7-[[l-[[(1- aminoisoquinolin-5-yl)amino]methyl]-2-azabicyclo[2.1.1]hexan -4-yl]methoxy]-2,3-dihydro-lH-indolizin- 5-one (70 mg, 0.13 mmol) in dry DMF (1.5 mL) was added DBU (30 μL, 0.2 mmol) after 10 min. The reaction was stirred at rt for 18 h, quenched with water (10 mL) and concentrated. The residue was partitioned between EtOAc (10 mL) and water (10 mL). The aqueous layer was re-extracted with EtOAc (3 x 20 mL) and the combined organics were dried (MgSO4) and concentrated. Purification by automated HPLC (mass directed 2-60% MeCN in [2.5% NH3 in water]) afforded the product (24 mg, 32% yield). [M+H] + = 577.3

1 H NMR (DMSO-d6, 400 MHz) 6 1.12 - 1.24 (4H, m), 1.41 - 1.53 (2H, m), 1.76 - 1.84 (2H, m), 1.97 - 2.10 (2H, m), 2.95 (2H, t, J = 7.7 Hz), 3.36 (2H, s), 3.79 - 3.86 (2H, m), 3.90 (2H, d, J = 6.4 Hz), 4.13 (2H, s), 5.61 (1H, d, J = 2.5 Hz), 5.80 - 5.89 (1H, m), 6.18 (1H, t, J = 6.5 Hz), 6.51 (2H, s), 6.73 (1H, d, J = 7.7 Hz), 6.78 (1H, d, J = 6.0 Hz), 7.13 - 7.22 (4H, m), 7.23 - 7.32 (4H, m), 7.64 (1H, d, J = 6.1 Hz) Example number 65 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-N-(2,3-dihydro-lH -inden-5-yl)-4-(((1,6-dimethyl-2-oxo- l,2-dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexan e-2-carboxamide

According to General Method 21c, 5-indanyl isocyanate (30 mg, 0.18 mmol) was reacted with 4-((1-(((1- ((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-2 -azabicyclo[2.1.1]hexan-4-yl)methoxy)- l,6-dimethylpyridin-2(1H)-one (77 mg, 0.13 mmol) for 2 h. Concentration afforded the intermediate N- (2,3-dihydro-lH-inden-5-yl)-l-(((1-((2,4-dimethoxybenzyl)ami no)isoquinolin-5-yl)amino)methyl)-4-(((1,6- dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)-2-azabicy clo[2.1.1]hexane-2-carboxamide which was deprotected according to General Method 8, using TFA (250 μL, 3.33 mmol) in DCM (2 mL) at rt for 18 h. Following work up (iv), purification by flash chromatography (C18, 20-52% MeCN in 10 mM (NH4)HCO3 (aq)) afforded the product (23 mg, 32%).

[M+H] + = 565.7

1 H NMR (DMSO-d6, 400 MHz) 6 1.52 - 1.58 (2H, m), 1.84 (2H, d, J = 4.6 Hz), 1.99 (2H, p, J = 7.4 Hz), 2.26 (3H, s), 2.80 (4H, dt,J = 11.3, 7.4 Hz), 3.30 (3H, s), 3.50 (2H, s), 3.99 (2H, d, J = 6.2 Hz), 4.14 (2H, s), 5.69 (1H, d, J = 2.8 Hz), 5.84 (1H, dd, J = 2.8, 0.9Hz), 6.20 (1H, t, J = 6.4 Hz), 6.50 (2H, s), 6.74 (1H, d, J = 7.7 Hz), 6.98 (1H, d, J = 6.2 Hz), 7.07 (1H, d, J = 8.1 Hz), 7.17 - 7.26(2H, m), 7.33 (1H, d, J = 8.3 Hz), 7.41 (1H, s), 7.71 (1H, d, J = 6.0 Hz), 8.40 (1H, s)

Example number 66 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((1-methyl-2-o xo-l,2-dihydropyridin-4-yl)oxy)methyl)-

N-(2-phenylpropan-2-yl)-2-azabicyclo[2.1.1]hexane-2-carbo xamide

According to General Method 21b, 2-phenylpropan-2-amine (24 mg, 0.25 mmol) was reacted for 18h and a further 3h following the addition of 4-((1-(((1-aminoisoquinolin-5-yl)amino)methyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H)-o ne (53 mg, 0.13 mmol). Concentration and purification by flash chromatography (C18, 20-47% MeCN in 10 mM (NH4)HCO3 (aq)) followed by automated prep HPLC (UV directed 31-41% MeCN in 10 mM (NH4)HCO3 (aq)) over 8 min) afforded the product (70 mg, 94% yield).

[M+H] + = 553.7

1 H NMR (DMSO-d6, 500 MHz) 6 1.44 - 1.53 (2H, m), 1.56 (6H, s), 1.76 - 1.81 (2H, m), 3.31 (3H, s), 3.46 (2H, s), 3.81 (2H, d, J =6.3 Hz), 4.16 (2H, s), 5.79 (1H, d, J = 2.8 Hz), 5.91 (1H, dd, J = 7.5, 2.7 Hz), 6.04 (1H, t, J = 6.4 Hz), 6.42 (1H, s), 6.47 (2H, s), 6.63- 6.72 (2H, m), 7.13 - 7.22 (2H, m), 7.25 - 7.32 (3H, m), 7.35 - 7.41 (2H, m), 7.54 (1H, d, J = 7.6 Hz), 7.64 (1H, d, J = 6.1 Hz)

Example number 71

4-(((1,6-Dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methy l)-l-(((2-hydroxypyridin-3- yl)amino)methyl)-N-(1-phenylcyclopropyl)-2-azabicyclo[2.1.1] hexane-2-carboxamide tert-Butyl 4-(((1,6-dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)- l-(((2-methoxypyridin-3- yl)amino)methyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate

According to General Method 17, tert-butyl 4-[(1,2-dimethyl-6-oxopyridin-4-yl)oxymethyl]-l-formyl-2- azabicyclo[2.1.1]hexane-2-carboxylate (100 mg, 0.28 mmol) was reacted with 3-amino-2- methoxypyridine (38 mg, 0.3 mmol) at rt for 18 h. Aqueous work up and purification by HPLC (mass directed 2-60% MeCN in [2.5% NH4OH in water]) afforded the product (53 mg, 41% yield).

[M+H] + = 471.3

4-[[l-[[(2-Hydroxypyridin-3-yl)amino]methyl]-2-azabicyclo [2.1.1]hexan-4-yl]methoxy]-l,6- dimethylpyridin-2-one hydrochloride

According to General Method 5a, tert-butyl 4-(((1,6-dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)- l-(((2-methoxypyridin-3-yl)amino)methyl)-2-azabicyclo[2.1.1] hexane-2-carboxylate (54 mg, 0.11 mmol) was deprotected at rt for 3 days. The solution was concentrated to afford the product (44 mg, 98% yield).

[M+H] + = 357.2

4-(((1,6-Dimethyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methy l)-l-(((2-hydroxypyridin-3- yl)amino)methyl)-N-(1-phenylcyclopropyl)-2-azabicyclo[2.1.1] hexane-2-carboxamide

To a solution of 4-[[l-[[(2-hydroxypyridin-3-yl)amino]methyl]-2-azabicyclo[2. 1.1]hexan-4-yl]methoxy]- l,6-dimethylpyridin-2-one hydrochloride (43 mg, 0.11 mmol) in MeCN (2 mL) and DMF (1 mL) was added DBU (33 μL, 0.22 mmol). After 10 min N-(1-phenylcyclopropyl)imidazole-l-carboxamide (64 mg, 0.22 mmol) was added and the reaction stirred at rt for 36 h. The reaction mixture was quenched with water (10 mL) and extracted with EtOAc (3 x 20 mL) before being washed with brine (2 x 10 mL), dried (MgSO4) and concentrated. Purification by HPLC (mass directed 2-60% MeCN in [2.5% NH4OH in water]) afforded the product (21 mg, 37% yield). [M+H] + = 516.3

1 H NMR (DMSO-d6, 400 MHz) 6 1.05 - 1.23 (4H, m), 1.39 - 1.53 (2H, m), 1.75 (2H, d, J = 4.3 Hz), 2.28 (3H, s), 3.32 (3H, s), 3.35 (2H, s), 3.73 (2H, d, J = 6.5 Hz), 4.15 (2H, s), 5.50 (1H, t, J = 6.7 Hz), 5.72 (1H, d, J = 2.8 Hz), 5.89 (1H, d, J = 2.5 Hz), 6.02 (1H, t, J = 6.8 Hz), 6.24 (1H, dd, J = 7.2, 1.6 Hz), 6.49 - 6.60 (1H, m), 7.06 - 7.18 (3H, m), 7.19 - 7.30 (3H, m), 11.32 (1H, s)

Example number 90 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-N-(2,3-dihydroben zofuran-3-yl)-4-(((1,6-dimethyl-2-oxo- l,2-dihydropyridin-4-yl)oxy)methyl)-N-methyl-2-azabicyclo[2. 1.1]hexane-2-carboxamide

tert-Butyl N-(2,3-dihydrobenzofuran-3-yl)carbamate

BOC2O (766 μL, 3.33 mmol) was added to a mixture of 2,3-dihydrobenzofuran-3-amine hydrochloride

(500 mg, 2.91 mmol) and TEA (1.3 mL, 9.33 mmol) in DCM (10 mL) at rt and stirred for 18 h. The mixture was quenched with brine (20 mL) and DCM (20 mL). The layers were separated, and the organic layer was washed with brine (20 mL), dried (Na2SO4), filtered, and concentrated to afford the product (648 mg, 85% yield).

1 H NMR (CDCI3, 400 MHz) 6 1.46 (9H, s), 4.33 (1H, dd, J = 10.0, 4.1 Hz), 4.65 (1H, dd, J = 10.0, 7.8 Hz),

4.88 (1H, s), 5.35 (1H, s), 6.81 - 6.85 (1H, m), 6.91 (1H, td, J = 7.5, 1.0 Hz), 7.22 (1H, dddd, J = 8.0, 7.4, 1.4,

0.6 Hz), 7.33 (1H, d, J = 7.4 Hz) tert-Butyl (2,3-dihydrobenzofuran-3-yl)(methyl)carbamate

According to General Method 2a, tert-butyl N-(2,3-dihydrobenzofuran-3-yl)carbamate (648 mg, 2.48 mmol) was reacted with Mel (231 μL, 3.72 mmol) at rt for 18 h. The mixture was quenched with brine (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried (Na 2 SO4), filtered, and concentrated to afford the product.

N-Methyl-2,3-dihydrobenzofuran-3-amine hydrochloride

According to a variation on General Method 5a, tert-butyl (2,3-dihydrobenzofuran-3- yl)(methyl)carbamate was deprotected in MeOH (5mL) for 96 h at rt. The mixture was concentrated. THF (3 mL) was added, and the mixture was sonicated for 1 min, filtered, and washed with THF (2 x 5 mL), hexanes (2 x 5 mL), and Et 2 O (2 x 5 mL) to afford the product (501 mg, 44% yield).

1 H NMR (D 2 O, 400 MHz) 6 2.72 (3H, s), 4.72 (1H, dd, J = 12.0, 7.3 Hz), 4.84 (1H, d, J = 12.0 Hz), 5.09 (1H, d, J = 7.2 Hz), 7.06 (1H, d, J = 8.1 Hz), 7.14 (1H, t, J = 7.6 Hz), 7.49 (1H, t, J = 7.9 Hz), 7.61 (1H, d, J = 7.6 Hz)

(2,3-Dihydrobenzofuran-3-yl)(methyl)carbamic chloride

According to a modification of General Method 21b Part 1, N-methyl-2,3-dihydrobenzofuran-3-amine HCI (100 mg, 0.27 mmol, 40% pure) was reacted in anhydrous DCM (ImL) at 0 °C for 30 min then at rt for 18 h. After quenching with IM HCI (aq) (15 mL), the aqueous phase was extracted with DCM (3 x 25 mL). The combined organic layers were washed with IM HCI (aq) (25 mL), sat. NaHCO3 (aq) (25 mL), dried (Na 2 SO4), filtered, and concentrated. The product was used directly in the next step.

1 H NMR (CDCI 3 , 500 MHz) 6 0.32 (4H, s), 1.25 (1H, d, J = 6.5 Hz), 1.36 - 1.39 (3H, m), 3.25 - 3.28 (4H, m) l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-N-(2,3-dihydroben zofuran-3-yl)-4-(((1,6-dimethyl-2-oxo- l,2-dihydropyridin-4-yl)oxy)methyl)-N-methyl-2-azabicyclo[2. 1.1]hexane-2-carboxamide

According to a modification of General Method 21b, part 2, 4-[[l-[[(1-amino-5- isoquinolyl)amino]methyl]-2-azabicyclo[2.1.1]hexan-4-yl]meth oxy]-l,6-dimethylpyridin-2-one (50 mg, 0.12 mmol, 95% purity) was reacted with N-(2,3-dihydrobenzofuran-3-yl)-N-methyl-carbamoyl chloride

(61 mg, 0.14 mmol) in THF (2.00 mL) at rt for 72 h. The mixture was concentrated. Purification by flash chromatography (C18, 25-56% MeCN in 10 mM (NH4)HCO3 (aq)) followed by automated prep HPLC (UV directed 38-48% MeCN in 10 mM (NH4)HCO3 (aq) over 8 min) afforded the product (19 mg, 28% yield). [M+H] + = 581.4

1 H NMR (DMSO-d6, 400 MHz) 6 1.59 (2H, t, J = 4.2 Hz), 1.77 (2H, t, J = 6.2 Hz), 2.25 (3H, s), 2.52 - 2.55 (3H, m), 3.28 - 3.30 (4H, m), 3.38 (1H, d, J = 7.3 Hz), 3.83 (1H, dd, J = 14.5, 6.2 Hz), 3.92 (1H, dd, J = 14.5, 6.0 Hz), 4.10 (2H, s), 4.45 (1H, dd, J = 10.3, 4.2 Hz), 4.65 (1H, dd, J = 10.2, 9.2 Hz), 5.66 (1H, d, J = 2.8 Hz), 5.83 (1H, d, J = 2.6 Hz), 5.97 (1H, dd, J = 9.1, 4.2 Hz), 6.17 (1H, t, J = 6.2 Hz), 6.48 (2H, s), 6.75 (1H, d, J = 7.8 Hz), 6.87 (1H, d, J = 8.0 Hz), 6.92 (1H, ddd, J = 8.3, 7.0, 0.9 Hz), 7.10 (1H, d, J = 6.2 Hz), 7.20 (1H, t, J = 8.0 Hz), 7.22 - 7.29 (2H, m), 7.30 (1H, d, J = 8.6 Hz), 7.71 (1H, d, J = 6.1 Hz)

Example number 63

1-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((1-methyl- 2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)-

2-azabicyclo[2.1.1]hexane-2-carboxamide

According to General Method 21b, cumylamine (30 mg, 0.22 mmol) was reacted in DCM (1.5 mL) at rt for 90 min and then with 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H)-o ne ( 81 mg, 0.142mmol) for 36 h.

Aqueous work up afforded l-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)me thyl)-4-(((1- methyl-2-oxo-l,2-dihydropyridin-4-yl)oxy)methyl)-N-(2-phenyl propan-2-yl)-2-azabicyclo[2.1.1]hexane-2- carboxamide which was deprotected according to General Method 8 in DCM (2 mL) and TFA (250 μL, 3.33 mmol) at rt for 18 h. Purification by flash chromatography (C18, 20-32% MeCN in 10 mM (NH4)HCO3 (aq)) followed by automated prep HPLC (UV directed 14-24% MeCN in 10 mM (NH4)HCO3 (aq) over 8 min) afforded the product (12.8 mg, 13% yield).

[M+H] + = 435.6

1 H NMR (DMSO-d6, 400 MHz) 6 1.45 - 1.52 (2H, m), 1.77 - 1.85 (2H, m), 3.30 (2H, s), 3.30 (3H, s), 3.89 (2H, d, J = 6.2 Hz), 4.13 (2H, s), 5.77 (1H, d, J = 2.7 Hz), 5.89 (1H, dd, J = 7.5, 2.7 Hz), 5.92 (2H, s), 6.50 - 6.59 (3H, m), 6.77 (1H, d, J = 7.7 Hz), 6.88 (1H, d, J = 6.2 Hz), 7.20 (1H, t, J = 8.0 Hz), 7.31 (1H, d, J = 8.3

Hz), 7.53 (1H, d, J = 7.5 Hz), 7.70 (1H, d, J = 6.1 Hz)

Example number 91 l-(((1-Aminoisoquinolin-5-yl)amino)methyl)-4-(((1-methyl-2-o xo-l,2-dihydropyridin-4-yl)oxy)methyl)-

N-phenyl-2-azabicyclo[2.1.1]hexane-2-carbothioamide

A solution of 4-[[l-[[(1-amino-5-isoquinolyl)amino]methyl]-2-azabicyclo[2. 1.1]hexan-4-yl]methoxy]-l- methylpyridin-2-one (18 mg, 0.05 mmol) and TEA (20 μL, 0.14 mmol) in DMSO (750 μL) was added to isothiocyanatobenzene (8 mg, 0.06 mmol), and the mixture was stirred for 16 h. The mixture was quenched with MeOH (250 μL), filtered and purified by HPLC (mass directed, MeCN in 10 mM (NH4)HCO3 (aq)) to provide the product (16 mg, 64% yield).

[M+H] + = 527.1

1 H NMR (DMSO-d6, 300 MHz) 6 1.90 (2H, s), 2.04 (2H, s), 3.32 (3H, s), 3.74 (2H, s), 4.07 (2H, s), 4.23 (2H, s), 5.83 (1H, d, J = 2.6 Hz), 5.93 (1H, dd, J = 7.5, 2.7 Hz), 6.20 (1H, br s), 6.59 (2H, s), 6.83 - 6.97 (2H, m), 7.05 - 7.20 (5H, m), 7.24 (1H, t, J = 8.0 Hz), 7.49 (1H, d, J = 8.6 Hz), 7.56 (1H, d, J = 7.6 Hz), 7.79 (1H, d, J = 6.0 Hz), 9.93 (1H, s)

Example number 95

4-((1-(((1 _ Aminoisoquinolin-5-yl)amino)methyl)-2-(6-cyclopropylpy ridazin-3-yl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2(1 H)-one According to General Method 4, 4-((1-(((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amin o)methyl)- 2-azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-methylpyridin-2(1H) -one (60 mg, 0.11 mmol) was reacted with 3-chloro-6-cyclopropyl-pyridazine (20 mg, 0.129 mmol), using NaC Bu (16 mg, 0.16 mmol), RuPhosPd-G3 (13mg, 0.0148 mmol) and RuPhos (7 mg, 0.0147 mmol) in THF (1 mL) at reflux for 16 h. The mixture was cooled to rt, diluted with MeOH (500 μL), filtered, washing with MeOH (5 mL) and the filtrate was concentrated to afford the intermediate 4-((2-(6-cyclopropylpyridazin-3-yl)-l-(((1-((2,4- dimethoxybenzyl)amino)isoquinolin-5-yl)amino)methyl)-2-azabi cyclo[2.1.1]hexan-4-yl)methoxy)-l,6- dimethylpyridin-2(1H)-one which was deprotected according to General Method 8, in DCM (2 mL) at rt for 4 h. Aqueous work up and purification by flash chromatography (C18, 20-50% MeCN in 10 mM (NH4)HCO3 (aq)) afforded the product (32 mg, 57% yield).

[M+H] + = 524.4

1 H NMR (DMSO-d6, 300 MHz) 6 0.88 - 1.02 (4H, m), 1.66 (2H, d, J = 3.3 Hz), 1.91 (2H, d, J = 4.2 Hz), 2.05 - 2.16 (1H, m), 2.25(3H, s), 3.30 (3H, s), 3.42 (2H, s), 4.14 (2H, s), 4.18 (2H, d, J = 6.1 Hz), 5.68 (1H, d, J = 2.6 Hz), 5.83 (1H, d, J = 2.6 Hz), 6.43 -6.55 (3H, m), 6.76 (1H, d, J = 7.6 Hz), 6.96 (1H, d, J = 9.2 Hz), 7.01 (1H, d, J = 6.1 Hz), 7.17 - 7.29 (2H, m), 7.32 (1H, d, J = 8.4Hz), 7.69 (1H, d, J = 6.1 Hz)

Example number 97

4-((1-(((1-Aminoisoquinolin-5-yl)amino)methyl)-2-(4,5-dih ydrooxazol-2-yl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l,6-dimethylpyridin-2(1H)-one

According to General Method 21c, 4-((1-(((1-aminoisoquinolin-5-yl)amino)methyl)-2- azabicyclo[2.1.1]hexan-4-yl)methoxy)-l,6-dimethylpyridin-2(1 H)-one (100 mg, 0.25 mmol) was reacted with l-chloro-2-isocyanato-ethane (31 μL, 0.35 mmol) in anhydrous THF (5 mL) at 80 °C for 42 h. Concentration and purification by flash chromatography (C18, 20-51% MeCN in 10 mM NH4HCO3 (aq)) followed by automated prep HPLC (UV directed 30-40% MeCN in (10 mM NH4HCO3 (aq)) over 8 min) afforded the product (4 mg, 4% yield).

[M+H] + = 475.4

1 H NMR (DMSO-d6, 400 MHz) 6 1.55 (2H, s), 1.86 (2H, d, J = 4.7 Hz), 2.29 (3H, s), 3.32 (3H, s), 3.37 (2H, s), 3.79 (2H, t, J =8.7 Hz), 3.95 (2H, d, J = 5.8 Hz), 4.14 (2H, s), 4.27 (2H, t, J = 8.7 Hz), 5.70 (1H, d, J = 2.8 Hz), 5.87 (1H, d, J = 2.8 Hz), 6.52 (2H,d, J = 4.2 Hz), 6.69 - 6.79 (2H, m), 7.00 (1H, d, J = 6.2 Hz), 7.23 (1H, t, J = 8.0 Hz), 7.34 (1H, d, J = 8.3 Hz), 7.75 (1H, d, J = 6.1Hz)

Example number 102

4-((4-(((1-Aminoisoquinolin-5-yl)amino)methyl)-2-(3-cyclo hexylpropanoyl)-2-azabicyclo[2.1.1]hexan-l- yl)methoxy)-l-methylpyridin-2(1H)-one

According to General Method 18a, 3-cyclohexylpropanoic acid (25 mg, 0.16 mmol) was coupled to 4-((1- (((1-((2,4-dimethoxybenzyl)amino)isoquinolin-5-yl)amino)meth yl)-2-azabicyclo[2.1.1]hexan-4- yl)methoxy)-l-methylpyridin-2(1H)-one (80 mg, 0.15 mmol) over 18 h. The mixture was concentrated to afford 4-((2-(3-cyclohexylpropanoyl)-l-(((1-((2,4-dimethoxybenzyl)a mino)isoquinolin-5- yl)amino)methyl)-2-azabicyclo[2.1.1]hexan-4-yl)methoxy)-l-me thylpyridin-2(1H)-one which was deprotected according to General Method 8 in DCM (1 mL) and TFA (1379 μL, 13.5 mmol) at rt for 24 h. Purification by flash chromatography (C18, 5-60% MeCN in 10 mM (NH4)HCO3 (aq)) provided the product (38 mg, 48% yield).

[M+H] + = 530.3

1 H NMR (400 MHz, DMSO-d6) 6 0.86 (2H t, J = 11.7 Hz), 1.06 - 1.26 (5H, m), 1.37 (2H, q, J = 7.3 Hz), 1.57 (3H, s ), 1.65 (6H, s), 1.86 - 1.95 (2H, m), 2.12 - 2.23 (2H, m), 3.51 -3.53 (4H, m), 4.59 (2H, s), 5.65 (1H, s), 5.84 - 5.90 (1H, m,), 5.96 (1H, s), 6.48 (2H, d, J = 4.4 Hz), 6.68 (1H, d, J = 7.7 Hz), 7.15 - 7.18 (1H, d, J = 6.0 Hz), 7.20 (1H, t, J = 8.0 Hz), 7.33 (1H, d, J = 8.3 Hz), 7.52 (1H, d, J = 7.5 Hz), 7.72 (1H, d, J = 6.0 Hz) Example Number 104 l-(((1-aminoisoquinolin-5-yl)(methyl)amino)methyl)-N-(chroma n-6-yl)-4-(((1,6-dimethyl-2-oxo-l,2- dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-2- carboxamide tert-Butyl 4-[[tert-butyl(diphenyl)silyl]oxymethyl]-l-[[[l-[(2,4-dimeth oxyphenyl)methylamino]-5- isoquinolyl]-methyl-amino]methyl]-2-azabicyclo[2.1.1]hexane- 2-carboxylate

Dess-Martin periodinane (524 mg, 1.17 mmol) was added to a solution of tert-butyl 4-[[tert- butyl(diphenyl)silyl]oxymethyl]-l-(hydroxymethyl)-2-azabicyc lo[2.1.1]hexane-2-carboxylate (400 mg, 0.748 mmol) in DCM (7 mL) at 0 °C and stirred for 15 min. The mixture was warmed to rt and stirred for 3 h. Sat. NaHCO3 (aq) (5mL) and sat. Na2S2O3 (aq) (5 mL) were added. The layers were separated, and the aqueous layer was extracted with DCM (3x15 mL). The combined organic layers were washed with sat. Na2S2O3 (aq)(15 mL), sat. NaHCO3 (aq)(15 mL), and brine (15 mL), dried (Na2SO4) and concentrated to afford the aldehyde intermediate which was used without further purification in the next step.

[M+H] + = 478.9

AcOH (60 μL, 1.0 mmol) was added to a mixture of the aldehyde intermediate and Nl-[(2,4- dimethoxyphenyl)methyl]-N5-methyl-isoquinoline-l,5-diamine (274 mg, 1.17 mmol) in DCE (7 mL) at rt and stirred for 5 min. NaBH(OAc)3 (435 mg, 1.64 mmol) was added and the mixture stirred at rt for 16 h. The mixture was cooled in an ice/water bath and sat. NaHCO3 (aq) (5 mL) and brine (5 mL) were added. The layers were separated and the aqueous layer extracted with DCM (3 x 15 mL). The combined organic layers were dried (Na2SO4), filtered and concentrated, and the residue purified by flash chromatography (Silica, 0-48% EtOAc in hexanes) to afford the product (184 mg, 30% yield over two steps).

[M+H] + = 787.5

1 H NMR (DMSO-d6, 500 MHz) 6 0.92 (9H, s), 1.25 (2H, dd, J = 4.4, 1.8 Hz), 1.38 (9H, s), 1.73 - 1.80 (2H, m), 2.78 (3H, s), 3.13 (2H, s), 3.70 (3H, s), 3.73 (2H, s), 3.82 (3H, d, J = 1.3 Hz), 3.84 (2H, s), 4.60 (2H, d, J = 5.6 Hz), 6.36 (1H, dd, J = 8.4, 2.4 Hz), 6.55 (1H, d, J = 2.3 Hz), 7.06 (1H, d, J = 8.3 Hz), 7.10 (1H, dd, J = 6.0, 0.8 Hz), 7.34 - 7.41 (6H, m), 7.41 - 7.47 (2H, m), 7.49 - 7.54 (4H, m), 7.58 (1H, t, J = 5.9 Hz), 7.76 (1H, d, J = 6.0 Hz), 7.99 (1H, d, J = 8.1 Hz) tert-Butyl l-[[[l-[(2,4-dimethoxyphenyl)methylamino]-5-isoquinolyl]-met hyl-amino]methyl]-4-

(hydroxymethyl)-2-azabicyclo[2.1.1]hexane-2-carboxylate

TBAF (IM in THF, 500 μL, 0.500 mmol) was added to a mixture of tert-butyl 4-[[tert- butyl(diphenyl)silyl]oxymethyl]-l-[[[l-[(2,4-dimethoxyphenyl )methylamino]-5-isoquinolyl]-methyl- amino]methyl]-2-azabicyclo[2.1.1]hexane-2-carboxylate (184 mg, 0.222 mmol) in THF (1.5 mL). The mixture was stirred at rt for 2 h then concentrated. The residue was diluted with DCM (20 mL) and washed with a mixture of sat. NaHCO3 (aq) and brine (1:1 v/v, 10 mL). The layers were separated, and the aqueous layer was extracted with DCM (2x 20 mL). The combined organic layers were dried (Na2SO4), filtered, and concentrated. The residue was purified by flash chromatography (Silica, 100% EtOAc) to afford the product (91 mg, 74%).

[M+H] + = 549.8 tert-Butyl l-[[[l-[(2,4-dimethoxyphenyl)methylamino]-5-isoquinolyl]-met hyl-amino]methyl]-4-[(1,2- dimethyl-6-oxo-4-pyridyl)oxymethyl]-2-azabicyclo[2.1.1]hexan e-2-carboxylate

DIAD (50 μL, 0.25 mmol) was added to a mixture of tert-butyl l-[[[l-[(2,4-dimethoxyphenyl)methylamino]- 5-isoquinolyl]-methyl-amino]methyl]-4-(hydroxymethyl)-2-azab icyclo[2.1.1]hexane-2-carboxylate (91 mg, 0.17 mmol), triphenylphospine (68 mg, 0.26 mmol) and 4-hydroxy-l,6-dimethyl-pyridin-2-one (28 mg, 0.19 mmol) in THF (2 mL) at rt. The mixture was stirred at rt for 18 h and filtered (Celite®). The solid was washed with EtOAc and the filtrate was concentrated. The residue was purified by flash chromatography (silica, 0-100% hexanes in EtOAc followed by 0-18% MeOH in EtOAc) to afford the product (75 mg, 68% yield).

[M+H] + = 670.8

4-[[4-[[[l-[(2,4-Dimethoxyphenyl)methylamino]-5-isoquinol yl]-methyl-amino]methyl]-2- azabicyclo[2.1.1]hexan-l-yl]methoxy]-l,6-dimethyl-pyridin-2- one

HCI (IM in EtOAc, 2 mL, 2 mmol) was added to a mixture of tert-butyl 4-[[[l-[(2,4- dimethoxyphenyl)methylamino]-5-isoquinolyl]-methyl-amino]met hyl]-l-[(1,2-dimethyl-6-oxo-4- pyridyl)oxymethyl]-2-azabicyclo[2.1.1]hexane-2-carboxylate (75 mg, 0.11 mmol) at rt The mixture was heated to 30 °C and stirred for 18 h and concentrated. The solid was diluted with a mixture of DCM and MeOH (9:1 v/v, 20 mL) and with NaOH (aq) (2M, 5 mL). The phases were separated, and the aqueous phase was extracted with a mixture of DCM and MeOH (9:1, 2 x 20 mL). The combined organic layers were dried (Na2SO4 , filtered, and concentrated to afford the product (63 mg, 99% yield).

[M+H] + = 570.3 l-(((1-aminoisoquinolin-5-yl)(methyl)amino)methyl)-N-(chroma n-6-yl)-4-(((1,6-dimethyl-2-oxo-l,2- dihydropyridin-4-yl)oxy)methyl)-2-azabicyclo[2.1.1]hexane-2- carboxamide

Chroman-6-amine (24 mg, 0.15 mmol) in a mixture of THF (500 μL) and DCM (500 μL) was added to a mixture of triphosgene (16.4 mg, 0.0542 mmol) and pyridine (20 μL, 0.245 mmol) in THF (500 μL) at rt. The mixture was stirred at rt for 4 h and added to a mixture of 4-[[l-[[[l-[(2,4- dimethoxyphenyl)methylamino]-5-isoquinolyl]-methyl-amino]met hyl]-2-azabicyclo[2.1.1]hexan-4- yl]methoxy]-l,6-dimethyl-pyridin-2-one (63 mg, 0.11 mmol) and TEA (30 μL, 0.22 mmol) in DCM (1 mL) at rt. The mixture was stirred for 1 h and concentrated to afford the intermediate N-chroman-6-yl-l-[[[l- [(2,4-dimethoxyphenyl)methylamino]-5-isoquinolyl]-methyl-ami no]methyl]-4-[(1,2-dimethyl-6-oxo-4- pyridyl)oxymethyl]-2-azabicyclo[2.1.1]hexane-2-carboxamide. [M+H] + = 745.7

TFA (250 μL, 3.33 mmol) was added to a mixture of intermediate N-chroman-6-yl-l-[[[l-[(2,4- dimethoxyphenyl)methylamino]-5-isoquinolyl]-methyl-amino]met hyl]-4-[(1,2-dimethyl-6-oxo-4- pyridyl)oxymethyl]-2-azabicyclo[2.1.1]hexane-2-carboxamide in DCM (2 mL) at rt. The mixture was stirred at rt for 18 h and diluted with NaOH (aq) (2M, 10 mL) and a mixture of DCM and MeOH (9:1 v/v, 20 mL). The phases were separated, and the aqueous phase was extracted with a mixture of DCM and MeOH (9:1 v/v, 2 x 20 mL). The combined organic phases were concentrated, and the residue was purified by automated prep HPLC (UV directed 34-44% MeCN in 10 mM NH4HCO3 (aq) over 9 min) and lyophilized to afford the product (26 mg, 40% yield over two steps).

[M+H] + = 595.5

1 H NMR (DMSO-d6, 500 MHz) 6 1.43 (2H, dd, J = 4.6, 1.7 Hz), 1.75 - 1.81 (2H, m), 1.83 - 1.89 (2H, m), 2.27 (3H, s), 2.61 (2H, t, J = 6.5 Hz), 2.81 (3H, s), 3.37 (2H, s), 3.91 (2H, s), 4.02 - 4.06 (2H, m), 4.10 (2H, s), 5.67 (1H, d, J = 2.8 Hz), 5.82 (1H, dd, J = 2.8, 0.9 Hz), 6.52 - 6.58 (1H, m), 6.68 (2H, s), 7.01 - 7.08 (2H, m), 7.11 - 7.16 (1H, m), 7.37 (1H, t, J = 7.9 Hz), 7.42 (1H, dd, J = 7.6, 1.1Hz), 7.78 (1H, d, J = 6.0 Hz), 7.87 (1H, d, J =

8.2 Hz), 8.56 (1H, s); 3H under the water peak.

EXAMPLES

Table 6: Compound Names

Table 7: 1 H NMR data of examples (solvent d6 DMSO unless otherwise indicated)

Biological Methods Determination of FXIIa inhibition

In vitro inhibition of Factor XI la was determined using an IC 5 o assay based on standard literature methods (see e.g Baeriswyl et al., ACS Chem. Biol., 2015, 10 (8) 1861; Bouckaert et al., European Journal of Medicinal Chemistry 110 (2016) 181). Human Factor Xlla (Enzyme Research Laboratories) was incubated at 25 °C with the fluorogenic substrate H-DPro-Phe-Arg-AFC (Peptide Protein Science) and various concentrations of the test compound. Protease activity was measured by monitoring the accumulation of liberated fluorescence from the substrate over 5 min at 25 °C. The linear rate of fluorescence increase per minute was expressed as percentage (%) activity. The Km for the cleavage of the substrate by FXIIa was determined by standard transformation of the Michaelis-Menten equation. The compound inhibitor assays were performed at substrate Km concentration. IC 5 o values were calculated as the concentration of inhibitor giving 50% inhibition ( IC 5 o) of the uninhibited enzyme activity (100%). Data acquired from this assay are shown in Table 16 below using the following scale:

For the test compounds that did not achieve 50% inhibition the following scale is used:

Table 8: Human FXIIa data, molecular weight and LCMS data

Determination of related protease inhibition

In vitro inhibition of related proteases was determined using an IC 5 o assay based on standard literature methods (see e.g. Shori et al., Biochem. Pharmacol., 1992,43, 1209; Bouckaert et al., European Journal of Medicinal Chemistry 110 (2016) 181). Human serine protease enzyme Plasma Kallikrein, KLK1, FXa, Plasmin, Thrombin and Trypsin were assayed for enzymatic activity using an appropriate fluorogenic substrate at Km concentration, FXIa at fixed substrate concentration of lOOpM and various concentrations of the test compound. Protease activity was measured by monitoring the accumulation of liberated fluorescence from the substrate over 5 min at 25 °C. The linear rate of fluorescence increase per minute was expressed as percentage (%) activity. IC 5 o values were calculated as the concentration of inhibitor giving 50% inhibition of the uninhibited enzyme activity (100%).

Data acquired from this assay are shown in Table 17 using the scale shown in Table 18. Table 9: Enzyme selectivity data

Table 10: Scale used to present enzyme selectivity data