Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NAPHTHYRIDINE COMPOUNDS AS INHIBITORS OF KRAS
Document Type and Number:
WIPO Patent Application WO/2023/034290
Kind Code:
A1
Abstract:
Disclosed are compounds of Formula (I), methods of using the compounds for inhibiting KRAS activity and pharmaceutical compositions comprising such compounds. The compounds are useful in treating, preventing or ameliorating diseases or disorders associated with KRAS activity such as cancer.

Inventors:
LI GENCHENG (US)
YIN HAOLIN (US)
QI CHAO (US)
WANG XIAOZHAO (US)
YAO WENQING (US)
HU BIN (US)
CARLSEN PETER (US)
Application Number:
PCT/US2022/042023
Publication Date:
March 09, 2023
Filing Date:
August 30, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INCYTE CORP (US)
International Classes:
C07D471/14; A61K31/4375; A61P35/00; C07D519/00
Domestic Patent References:
WO2011054846A12011-05-12
WO2006056399A22006-06-01
WO2004005281A12004-01-15
WO2003037347A12003-05-08
WO2003099771A22003-12-04
WO2004046120A22004-06-03
WO2000009495A12000-02-24
WO2005028444A12005-03-31
WO2004080980A12004-09-23
WO2004056786A22004-07-08
WO2003024967A22003-03-27
WO2001064655A12001-09-07
WO2000053595A12000-09-14
WO2001014402A12001-03-01
WO2009085185A12009-07-09
WO2012129344A12012-09-27
WO2011101409A12011-08-25
WO2003062236A12003-07-31
WO2010075074A12010-07-01
WO2012061156A12012-05-10
WO2003042402A22003-05-22
WO2008156712A12008-12-24
WO2010089411A22010-08-12
WO2010036959A22010-04-01
WO2011066342A22011-06-03
WO2011159877A22011-12-22
WO2011082400A22011-07-07
WO2011161699A22011-12-29
WO2002000196A22002-01-03
Foreign References:
US5521184A1996-05-28
USPP60578491P
US7488802B22009-02-10
US7943743B22011-05-17
US8008449B22011-08-30
US8168757B22012-05-01
US8217149B22012-07-10
US10308644B22019-06-04
US20170145025A12017-05-25
US20170174671A12017-06-22
US20170174679A12017-06-22
US20170320875A12017-11-09
US20170342060A12017-11-30
US20170362253A12017-12-21
US20180016260A12018-01-18
US20180057486A12018-03-01
US20180177784A12018-06-28
US20180177870A12018-06-28
US20180179179A12018-06-28
US20180179201A12018-06-28
US20180179202A12018-06-28
US20180273519A12018-09-27
US20190040082A12019-02-07
US20190062345A12019-02-28
US20190071439A12019-03-07
US20190127467A12019-05-02
US20190144439A12019-05-16
US20190202824A12019-07-04
US20190225601A12019-07-25
US20190300524A12019-10-03
US20190345170A12019-11-14
US20180179197A12018-06-28
US201916369654A2019-03-29
USPP62688164P
Other References:
OLIVIER MIRGUET ET AL: "Naphthyridines as Novel BET Family Bromodomain Inhibitors", CHEMMEDCHEM COMMUNICATIONS, vol. 9, no. 3, 2 September 2013 (2013-09-02), DE, pages 580 - 589, XP055577472, ISSN: 1860-7179, DOI: 10.1002/cmdc.201300259
JASON G. KETTLE ET AL: "Covalent inhibitors of the GTPase KRAS G12C : a review of the patent literature", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 30, no. 2, 8 January 2020 (2020-01-08), GB, pages 103 - 120, XP055722707, ISSN: 1354-3776, DOI: 10.1080/13543776.2020.1709443
MCDONALD, R. ET AL., CELL, vol. 170, no. 1, 2017, pages 577 - 592
COX, A.D. ET AL., NAT REV DRUG DISCOV, vol. 13, 2014, pages 828 - 51
COX, A.D. ET AL., NAT. REV. DRUG DISCOV., vol. 13, 2014, pages 828 - 51
A. KEREKES, J. MED. CHEM., vol. 54, 2011, pages 201 - 210
R. XU, J. LABEL COMPD. RADIOPHARM., vol. 58, 2015, pages 308 - 312
BERGE ET AL., J. PHARM. SCI., vol. 66, no. 1, 1977, pages 1 - 19
ORG. LETT., vol. 4, 2002, pages 979 - 981
SMITH ET AL.: "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2007, GOWER PUBLISHING COMPANY
ROBERTSON: "Protecting Group Chemistry", 2000, OXFORD UNIVERSITY PRESS
PETURSSION ET AL.: "Protecting Groups in Carbohydrate Chemistry", J. CHEM. EDUC., vol. 74, no. 11, 1997, pages 1297
WUTS ET AL.: "Protective Groups in Organic Synthesis", 2006, WILEY
KNICKELBEIN, K. ET AL., GENES & CANCER, 2015, pages 4 - 12
FERNANDEZ-MEDARDE, A. ET AL., GENES & CANCER, 2011, pages 344 - 358
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
"Pharmaceutical Preformulation and Formulation", 2009, THE PHARMACEUTICAL PRESS AND THE AMERICAN PHARMACEUTICAL ASSOCIATION
K. BLOM: "Two-Pump At Column Dilution Configuration for Preparative LC-MS", J. COMBI. CHEM., vol. 4, 2002, pages 295
K. BLOMR. SPARKSJ. DOUGHTYG. EVERLOFT. HAQUEA. COMBS: "Optimizing Preparative LC-MS Configurations and Methods for Parallel Synthesis Purification", J. COMBI. CHEM., vol. 5, 2003, pages 670
K. BLOMB. GLASSR. SPARKSA. COMBS: "Preparative LC-MS Purification: Improved Compound Specific Method Optimization", J. COMBI. CHEM., vol. 6, 2004, pages 874 - 883
K. BLOMB. GLASSR. SPARKSA. COMBS: "Preparative LCMS Purification: Improved Compound Specific Method Optimization", J. COMB. CHEM., vol. 6, 2004, pages 874 - 883
BLOM, B. GLASSR. SPARKSA. COMBS, J. COMB. CHEM., vol. 6, 2004, pages 874 - 883
Attorney, Agent or Firm:
TRINQUE, Brian, C. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein:

— represents a single bond or a double bond;

-AR1=BR2- is selected from -N=CR2-, and -CR1=N-;

X is selected from N and CR5;

Y is selected from N and CR6;

R1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1 , NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)2NRc1Rd1, and BRh1Ri1 ; wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9;

R2 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(=NRe2)Rb2, C(=NORa2)Rb2, C(=NRe2)NRc2Rd2, NRc2C(=NRe2)NRc2Rd2, NRc2C(=NRe2)Rb2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, S(O)2NRc2Rd2, and BRh2R'2; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; Cy1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R10; when R3N— CR4 is a single bond, then R4 is selected from =0 and =S; and

R3 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, and 5-10 membered heteroaryl-Ci.3 alkylene; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; when R3N— CR4 is a double bond, then R3 is absent; and

R4 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NR^R^, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3C(O)NRc3Rd3, C(=NRe3)Rb3, C(=NORa3)Rb3, C(=NRe3)NRc3Rd3, NRc3C(=NRe3)NRc3Rd3, NRc3C(=NRe3)Rb3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S^NR^R^, S(O)2Rb3, S(O)2NRc3Rd3, and BRh3R'3; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30;

R5 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa5, SRa5, C(O)Rb5, NRc5C(=NRe5)Rb5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S^NR^R^, S(O)2Rb5, S(O)2NRc5Rd5, and BRh5R'5; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9;

R6 is selected from H, Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, NR^R^, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, C(=NRe6)Rb6, C(=NORa6)Rb6, C(=NRe6)NRc6Rd6, NRc6C(=NRe6)NRc6Rd6, NRc6C(=NRe6)Rb6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S^NR^R^, S(O)2Rb6, S(O)2NRc6Rd6, and BRh6R'6; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R60;

R7 is selected from H, Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7C(O)NRc7Rd7, C(=NRe7)Rb7, C(=NORa7)Rb7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, NRc7C(=NRe7)Rb7, NRc7S(O)Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, S(O)2NRc7Rd7, and BRh7R'7; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R70;

Cy2 is selected from C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-14 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4- 14 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered

180 heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R20; each R10 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa1°, SRa1°, C(O)Rb1°, C(O)NRc10Rd1°, C(O)ORa1°, OC(O)Rb1°, OC(O)NRc10Rd1°, NRc10Rd1°, NRc10C(O)Rb1°, NRc10C(O)ORa1°, NRc10C(O)NRc10Rd1°, C(=NRe10)Rb1°, C(=NORa10)Rb1°, C(=NRe10)NRc10Rd1°, NRc10C(=NRe10)NRc10Rd1°, NRc10S(O)Rb1°, NRc10S(O)2Rb1°, NRc10S(O)2NRc10Rd1°, S(O)Rb1°, S(O)NRc10Rd1°, S(O)2Rb1°, S(O)2NRc10Rd10, and BRh10Ri1°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R11 ; each R11 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa11 , SRa11 , C(O)Rb11 , C(O)NRc11 Rd11 , C(O)ORa11 , OC(O)Rb11 , OC(O)NRc11 Rd11 , NRc11 Rd11 , NRc11C(O)Rb11 , NRc11C(O)ORa11 , NRc11C(O)NRc11 Rd11 , NRc11S(O)Rb11 , NRc11S(O)2Rb11 , NRc11S(O)2NRc11 Rd11 , S(O)Rb11 , S(O)NRc11 Rd11 , S(O)2Rb11 , S(O)2NRc11 Rd11 , and BRh11 R'11 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R12; each R12 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, ORa12, SRa12, C(O)Rb12, C(O)NRc12Rd12, C(O)ORa12, OC(O)Rb12, OC(O)NRc12Rd12, NRc12Rd12, NRc12C(O)Rb12, NRc12C(O)ORa12, NRc12C(O)NRc12Rd12, NRc12S(O)Rb12, NRc12S(O)2Rb12, NRc12S(O)2NRc12Rd12, S(O)Rb12, S(O)NRc12Rd12, S(O)2Rb12, S(O)2NRc12Rd12, and BRh12R'12; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each R20 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w

181 aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa2°, SRa2°,

C(O)Rb2°, C(O)NRc20Rd2°, C(O)ORa2°, OC(O)Rb2°, OC(O)NRc20Rd2°, NRc20Rd2°,

NRc20C(O)Rb2°, NRc20C(O)ORa2°, NRc20C(O)NRc20Rd2°, C(=NRe20)Rb2°, C(=NORa20)Rb2°,

C(=NRe20)NRc20Rd2°, NRc20C(=NRe20)NRc20Rd2°, NRc20S(O)Rb2°, NRc20S(O)2Rb2°, NRc20S(O)2NRc20Rd2°, S(O)Rb2°, S(O)NRc20Rd2°, S(O)2Rb2°, S(O)2NRc20Rd2°, and BRh20Ri2°; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21 ; each R21 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa21 , SRa21 , C(O)Rb21 , C(O)NRc21 Rd21 , C(O)ORa21 , OC(O)Rb21 , OC(O)NRc21 Rd21 , NRc21 Rd21 , NRc21C(O)Rb21 , NRc21C(O)ORa21 , NRc21C(O)NRc21 Rd21 , NRc21S(O)Rb21 , NRc21S(O)2Rb21 , NRc21S(O)2NRc21 Rd21 , S(O)Rb21 , S(O)NRc21 Rd21 , S(O)2Rb21 , S(O)2NRc21 Rd21 , and BRh21 R'21 ; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R22; each R22 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, ORa22, SRa22, C(O)Rb22, C(O)NRc22Rd22, C(O)ORa22, OC(O)Rb22, OC(O)NRc22Rd22, NRc22Rd22, NRc22C(O)Rb22, NRc22C(O)ORa22, NRc22C(O)NRc22Rd22, NRc22S(O)Rb22, NRc22S(O)2Rb22, NRc22S(O)2NRc22Rd22, S(O)Rb22, S(O)NRc22Rd22, S(O)2Rb22, S(O)2NRc22Rd22, and BRh22R'22; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each R23 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa23, SRa23, C(O)Rb23, C(O)NRc23Rd23, C(O)ORa23, OC(O)Rb23, OC(O)NRc23Rd23, NRc23Rd23, NRc23C(O)Rb23, NRc23C(O)ORa23, NRc23C(O)NRc23Rd23, NRc23S(O)Rb23, NRc23S(O)2Rb23, NRc23S(O)2NRc23Rd23, S(O)Rb23, S(O)NRc23Rd23, S(O)2Rb23, S(O)2NRc23Rd23, and BRh23R'23;

182 wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R24; each R24 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, ORa24, SRa24, C(O)Rb24, C(O)NRc24Rd24, C(O)ORa24, OC(O)Rb24, OC(O)NRc24Rd24, NRc24Rd24, NRc24C(O)Rb24, NRc24C(O)ORa24, NRc24C(O)NRc24Rd24, NRc24S(O)Rb24, NRc24S(O)2Rb24, NRc24S(O)2NRc24Rd24, S(O)Rb24, S(O)NRc24Rd24, S(O)2Rb24, S(O)2NRc24Rd24, and BRh24R'24; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each R30 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa3°, SRa3°, C(O)Rb3°, C(O)NRc30Rd3°, C(O)ORa3°, OC(O)Rb3°, OC(O)NRc30Rd3°, NR^R^0, NRc30C(O)Rb3°, NRc30C(O)ORa3°, NR^C^NR^R^0, NRc30S(O)Rb3°, NRc30S(O)2Rb3°, NRc30S(O)2NRc30Rd3°, S(O)Rb3°, S^NR^R^0, S(O)2Rb3°, S(O)2NRc30Rd30, and BRh30Ri3°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31 ; each R31 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa31 , SRa31 , wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R32;

183 each R32 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, S(O)2NRc32Rd32, and BRh32R'32; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each R60 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-io cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa6°, SRa6°, C(O)Rb6°, C(O)NRc60Rd6°, C(O)ORa6°, OC(O)Rb6°, OC(O)NRc60Rd6°, NR^R^0, NRc60C(O)Rb6°, NRc60C(O)ORa6°, NRc60C(O)NRc60Rd6°, NRc60S(O)Rb6°, NRc60S(O)2Rb6°, NRc60S(O)2NRc60Rd6°, S(O)Rb6°, S^NR^R^0, S(O)2Rb6°, S(O)2NRc60Rd60, and BRh60Ri6°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ciw alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R61 ; each R61 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa61 , SRa61 , C(O)Rb61 , C(O)NRc61 Rd61 , C(O)ORa61 , OC(O)Rb61 , OC(O)NRc61Rd61, NR^ R^1 , NRc61C(O)Rb61 , NRc61C(O)ORa61 , NRc61C(O)NRc61Rd61 , NRc61S(O)Rb61 , NRc61S(O)2Rb61 , NRc61S(O)2NRc61 Rd61 , S(O)Rb61 , S^NR^ R^1 , S(O)2Rb61 , S(O)2NRc61 Rd61 , and BRh61 R'61 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R62; each R62 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl,

184 S(O)2NRc62Rd62, and BRh62R'62; wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each R70 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa7°, SRa7°,

C(O)Rb7°, C(O)NRc70Rd7°, C(O)ORa7°, OC(O)Rb7°, OC(O)NRc70Rd7°, NRc70Rd7°,

NRc70C(O)Rb70, NRc70C(O)ORa7°, NRc70C(O)NRc70Rd7°, NRc70S(O)Rb7°, NRc70S(O)2Rb7°, NRc70S(O)2NRc70Rd7°, S(O)Rb7°, S(O)NRc70Rd7°, S(O)2Rb7°, S(O)2NRc70Rd70, and BRh70Ri7°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Ra1, Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R9; each Rh1 and Ri1 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh1 and Ri1 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and C1.6 haloalkyl; each Ra2, Rb2, Rc2 and Rd2 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; each Re2 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh2 and R'2 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh2 and R'2 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra3, Rb3, Rc3 and Rd3 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl, and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; each Re3 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh3 and R'3 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh3 and R'3 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra5, Rb5, R05, and Rd5 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R9; each Re5 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh5 and R'5 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh5 and R'5 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra6, Rb6, R06, and Rd6 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R60; or any Rc6 and Rd6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R60; each Re6 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh6 and R'6 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh6 and R'6 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra7, Rb7, Rc7 and Rd7 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R70; or any Rc7 and Rd7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R70; each Re7 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh7 and R'7 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh7 and R'7 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra1°, Rb1°, Rc1° and Rd1° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10

187 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R11; or any Rc1° and Rd1° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R11; each Re1° is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh1° and R'10 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh1° and R'10 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra11 , Rb11 , Rc11 and Rd11, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R12; or any Rc11 and Rd11 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R12; each Rh11 and R'11 is independently selected from OH, C1-6 alkoxy, and C1-6 haloalkoxy; or any Rh11 and R'11 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from C1-6 alkyl and C1-6 haloalkyl; each Ra12, Rb12, Rc12 and Rd12, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Rh12 and R'12 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh12 and R'12 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra2°, Rb2°, Rc2° and Rd2° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-

188 aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21; or any Rc20 and Rd20 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21; each Re20 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh20 and R'20 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh20 and R'20 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra21 , Rb21 , Rc21 and Rd21, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R22; or any Rc21 and Rd21 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R22; each Rh21 and R'21 is independently selected from OH, C1-6 alkoxy, and C1-6 haloalkoxy; or any Rh21 and R'21 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from C1-6 alkyl and C1-6 haloalkyl; each Ra22, Rb22, Rc22 and Rd22, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Rh22 and R'22 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh22 and R'22 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl;

189 each Ra23, Rb23, Rc23 and Rd23, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R24; or any Rc23 and Rd23 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R24; each Rh23 and R'23 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh23 and R'23 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra24, Rb24, Rc24 and Rd24, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Rh24 and R'24 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh24 and R'24 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra3°, Rb3°, Rc3° and Rd3° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31; or any Rc3° and Rd3° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31; each Rh3° and R'30 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh3° and R'30 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra31 , Rb31 , R031 and Rd31, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl,

190 phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R32; or any R031 and Rd31 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R32; each Rh31 and R'31 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh31 and R'31 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra32, Rb32, Rc32 and Rd32, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc32 and Rd32 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Rh32 and R'32 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh32 and R'32 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra6°, Rb6°, Rc6° and Rd6° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R61; or any Rc6° and Rd6° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R61; each Rh6° and R'60 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh6° and R'60 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra61 , Rb61 , R061 and Rd61, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl,

191 phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R62; or any R061 and Rd61 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R62; each Rh61 and R'61 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh61 and R'61 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra62, Rb62, Rc62 and Rd62, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc62 and Rd62 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Rh62 and R'62 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh62 and R'62 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra7°, Rb7°, Rc7° and Rd7° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc7° and Rd7° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; each Rh7° and R'70 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh7° and R'70 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; and each R9 is independently selected from D, OH, NO2, ON, halo, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, C3-6 cycloalkyl-Ci.2 alkylene, Ci-e alkoxy, Ci-e haloalkoxy, Ci-3 alkoxy-Ci-3 alkyl, Ci-3 alkoxy-Ci-3 alkoxy, HO-C1.3 alkoxy, HO-C1.3 alkyl,

192 cyano-Ci-3 alkyl, H2N-C1.3 alkyl, amino, Ci-e alkylamino, di(Ci-6 alkyl)amino, thio, Ci-e alkylthio, C1.6 alkylsulfinyl, Ci-e alkylsulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci-e alkylcarbonyl, Ci-e alkoxycarbonyl, Ci-e alkylcarbonylamino, Ci-e alkoxycarbonylamino, Ci-e alkylcarbonyloxy, aminocarbonyloxy, Ci-e alkylaminocarbonyloxy, di(Ci-6 alkyl)aminocarbonyloxy, Ci-e alkylsulfonylamino, aminosulfonyl, Ci-e alkylaminosulfonyl, di(Ci-6 alkyl)aminosulfonyl, aminosulfonylamino, Ci-e alkylaminosulfonylamino, di(Ci-6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci-e alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino; provided that,

(a) Cy1 is other than 3,5-dimethylisoxazol-4-yl; and

(b) R3 is other than CH2CF3.

2. The compound of claim 1 , or a pharmaceutically acceptable salt thereof, wherein

— represents a single bond or a double bond;

-AR1=BR2- is selected from -N=CR2-, and -CR1=N-;

X is selected from N and CR5;

Y is selected from N and CR6;

R1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, ORa1 , SRa1 , C(O)Rb1 , C(O)NRc1 Rd1 , C(O)ORa1 , OC(O)Rb1 , OC(O)NRc1 Rd1 , NRc1 Rd1 , NRc1C(O)Rb1 , NRc1C(O)ORa1 , NRc1C(O)NRc1Rd1 , NRc1S(O)Rb1, NRc1S(O)2Rb1 , NRc1S(O)2NRc1 Rd1 , S(O)Rb1 , S(O)NRc1 Rd1 , S(O)2Rb1 , S(O)2NRc1 Rd1 , and BRh1 Ri1 ; wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9;

R2 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(=NRe2)Rb2, C(=NORa2)Rb2, C(=NRe2)NRc2Rd2, NRc2C(=NRe2)NRc2Rd2, NRc2C(=NRe2)Rb2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, S(O)2NRc2Rd2, and BRh2R'2; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23;

193 Cy1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R10; when R3N— CR4 is a single bond, then R4 is selected from =0 and =S; and

R3 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, and 5-10 membered heteroaryl-Ci.3 alkylene; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; when R3N— CR4 is a double bond, then R3 is absent; and

R4 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NR^R^, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3C(O)NRc3Rd3, C(=NRe3)Rb3, C(=NORa3)Rb3, C(=NRe3)NRc3Rd3, NRc3C(=NRe3)NRc3Rd3, NRc3C(=NRe3)Rb3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S^NR^R^, S(O)2Rb3, S(O)2NRc3Rd3, and BRh3R'3; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30;

R5 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa5, SRa5, C(O)Rb5,

194 NRc5C(=NRe5)Rb5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S^NR^R^, S(O)2Rb5, S(O)2NRc5Rd5, and BRh5R'5; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9;

R6 is selected from H, Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, NR^R^, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, C(=NRe6)Rb6, C(=NORa6)Rb6, C(=NRe6)NRc6Rd6, NRc6C(=NRe6)NRc6Rd6, NRc6C(=NRe6)Rb6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S^NR^R^, S(O)2Rb6, S(O)2NRc6Rd6, and BRh6R'6; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R60;

R7 is selected from H, Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7C(O)NRc7Rd7, C(=NRe7)Rb7, C(=NORa7)Rb7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, NRc7C(=NRe7)Rb7, NRc7S(O)Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, S(O)2NRc7Rd7, and BRh7R'7; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R70;

Cy2 is selected from C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-14 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4- 14 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered

195 heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R20; each R10 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa1°, SRa1°, C(O)Rb1°, C(O)NRc10Rd1°, C(O)ORa1°, OC(O)Rb1°, OC(O)NRc10Rd1°, NRc10Rd1°, NRc10C(O)Rb1°, NRc10C(O)ORa1°, NRc10C(O)NRc10Rd1°, C(=NRe10)Rb1°, C(=NORa10)Rb1°, C(=NRe10)NRc10Rd1°, NRc10C(=NRe10)NRc10Rd1°, NRc10S(O)Rb1°, NRc10S(O)2Rb1°, NRc10S(O)2NRc10Rd1°, S(O)Rb1°, S(O)NRc10Rd1°, S(O)2Rb1°, S(O)2NRc10Rd10, and BRh10Ri1°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R11 ; each R11 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa11 , SRa11 , C(O)Rb11 , C(O)NRc11 Rd11 , C(O)ORa11 , OC(O)Rb11 , OC(O)NRc11 Rd11 , NRc11 Rd11 , NRc11C(O)Rb11 , NRc11C(O)ORa11 , NRc11C(O)NRc11 Rd11 , NRc11S(O)Rb11 , NRc11S(O)2Rb11 , NRc11S(O)2NRc11 Rd11 , S(O)Rb11 , S(O)NRc11 Rd11 , S(O)2Rb11 , S(O)2NRc11 Rd11 , and BRh11 R'11 ; each R20 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa2°, SRa2°, C(O)Rb2°, C(O)NRc20Rd2°, C(O)ORa2°, OC(O)Rb2°, OC(O)NRc20Rd2°, NRc20Rd2°, NRc20C(O)Rb2°, NRc20C(O)ORa2°, NRc20C(O)NRc20Rd2°, C(=NRe20)Rb2°, C(=NORa20)Rb2°, C(=NRe20)NRc20Rd2°, NRc20C(=NRe20)NRc20Rd20, NRc20S(O)Rb2°, NRc20S(O)2Rb2°, NRc20S(O)2NRc20Rd2°, S(O)Rb2°, S(O)NRc20Rd2°, S(O)2Rb2°, S(O)2NRc20Rd20, and BRh20Ri2°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21 ; each R21 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, ORa21 , SRa21 , C(O)Rb21 , C(O)NRc21 Rd21 , C(O)ORa21 , OC(O)Rb21 , OC(O)NRc21 Rd21 , NRc21 Rd21 , NRc21C(O)Rb21 , NRc21C(O)ORa21 , NRc21C(O)NRc21 Rd21 , NRc21S(O)Rb21 , NRc21S(O)2Rb21 , NRc21S(O)2NRc21 Rd21 , S(O)Rb21 , S(O)NRc21 Rd21 , S(O)2Rb21 , S(O)2NRc21 Rd21 , and BRh21 R'21 ; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R22; each R22 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, ORa22, SRa22, C(O)Rb22, C(O)NRc22Rd22, C(O)ORa22, OC(O)Rb22, OC(O)NRc22Rd22, NRc22Rd22, NRc22C(O)Rb22, NRc22C(O)ORa22, NRc22C(O)NRc22Rd22, NRc22S(O)Rb22, NRc22S(O)2Rb22, NRc22S(O)2NRc22Rd22, S(O)Rb22, S(O)NRc22Rd22, S(O)2Rb22, S(O)2NRc22Rd22, and BRh22R'22; each R23 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3- cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa23, SRa23, C(O)Rb23, C(O)NRc23Rd23, C(O)ORa23, OC(O)Rb23, OC(O)NRc23Rd23, NRc23Rd23, NRc23C(O)Rb23, NRc23C(O)ORa23, NRc23C(O)NRc23Rd23, NRc23S(O)Rb23, NRc23S(O)2Rb23, NRc23S(O)2NRc23Rd23, S(O)Rb23, S(O)NRc23Rd23, S(O)2Rb23, S(O)2NRc23Rd23, and BRh23R'23; each R30 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa3°, SRa3°, C(O)Rb3°, C(O)NRc30Rd3°, C(O)ORa3°, OC(O)Rb3°, OC(O)NRc30Rd3°, NR^R^0, NRc30C(O)Rb3°, NRc30C(O)ORa3°, NR^C^NR^R^0, NRc30S(O)Rb3°, NRc30S(O)2Rb3°, NRc30S(O)2NRc30Rd3°, S(O)Rb3°, S^NR^R^0, S(O)2Rb3°, S(O)2NRc30Rd3°, and BRh30Ri3°; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31 ; each R31 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, ORa31 , SRa31 , wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R32; each R32 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, each R60 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3- cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, ORa6°, SRa6°, C(O)Rb6°, C(O)NRc60Rd6°, C(O)ORa6°, OC(O)Rb6°, OC(O)NRc60Rd6°, NR^R^0, NRc60C(O)Rb6°, NRc60C(O)ORa6°, NR^C^NR^R^0, NRc60S(O)Rb6°, NRc60S(O)2Rb6°, NRc60S(O)2NRc60Rd6°, S(O)Rb6°, S^NR^R^0, S(O)2Rb6°, S(O)2NRc60Rd6°, and BRh60Ri6°; wherein said Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R61 ; each R61 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa61 , SRa61 , C(O)Rb61 , C(O)NRc61 Rd61 , C(O)ORa61 , OC(O)Rb61 , OC(O)NRc61Rd61, NR^ R^1 , NRc61C(O)Rb61 , NRc61C(O)ORa61 , NRc61C(O)NRc61Rd61 , NRc61S(O)Rb61 , NRc61S(O)2Rb61 , NRc61S(O)2NRc61 Rd61 , S(O)Rb61 , S^NR^ R^1 , S(O)2Rb61 , S(O)2NRc61 Rd61 , and BRh61 R'61 each R70 is independently selected from Ci-e alkyl, C2.6 alkenyl, C2.6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered

198 heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, NO2, ORa7°, SRa7°, C(O)Rb70, C(O)NRc70Rd7°, C(O)ORa7°, OC(O)Rb7°, OC(O)NRc70Rd7°, NRc70Rd7°, NRc70C(O)Rb7°, NRc70C(O)ORa7°, NRc70C(O)NRc70Rd7°, NRc70S(O)Rb7°, NRc70S(O)2Rb7°, NRc70S(O)2NRc70Rd7°, S(O)Rb7°, S(O)NRc70Rd7°, S(O)2Rb7°, S(O)2NRc70Rd70, and BRh70Ri7°; each Ra1, Rb1, Rc1, and Rd1 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R9; each Rh1 and Ri1 is independently selected from OH, C1-6 alkoxy, and C1-6 haloalkoxy; or any Rh1 and Ri1 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from C1-6 alkyl and C1-6 haloalkyl; each Ra2, Rb2, Rc2 and Rd2 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; each Re2 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh2 and R'2 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh2 and R'2 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and C1.6 haloalkyl; each Ra3, Rb3, Rc3 and Rd3 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-

199 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; each Re3 is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh3 and R'3 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh3 and R'3 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra5, Rb5, R05, and Rd5 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R9; or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R9; each Re5 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh5 and R'5 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh5 and R'5 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra6, Rb6, R06, and Rd6 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R60; or any Rc6 and Rd6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R60;

200 each Re6 is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh6 and R'6 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh6 and R'6 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra7, Rb7, Rc7 and Rd7 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R70; or any Rc7 and Rd7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R70; each Re7 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh7 and R'7 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh7 and R'7 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra1°, Rb1°, Rc1° and Rd1° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R11; or any Rc1° and Rd1° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R11; each Re1° is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl;

201 each Rh1° and R'10 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh1° and R'10 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra11 , Rb11 , Rc11 and Rd11, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl; or any Rc11 and Rd11 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Rh11 and R'11 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh11 and R'11 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra2°, Rb2°, Rc2° and Rd2° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21; or any Rc2° and Rd2° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21; each Re2° is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each Rh2° and R'20 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh2° and R'20 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra21 , Rb21 , Rc21 and Rd21, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R22;

202 or any Rc21 and Rd21 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R22; each Rh21 and R'21 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh21 and R'21 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra22, Rb22, Rc22 and Rd22, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; each Rh22 and R'22 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh22 and R'22 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra23, Rb23, Rc23 and Rd23, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any Rc23 and Rd23 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Rh23 and R'23 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh23 and R'23 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra3°, Rb3°, Rc3° and Rd3° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31; or any Rc3° and Rd3° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31; each Rh3° and R'30 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh3° and R'30 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally

203 substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra31 , Rb31 , R031 and Rd31, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R32; or any R031 and Rd31 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R32; each Rh31 and R'31 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh31 and R'31 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra32, Rb32, Rc32 and Rd32, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; or any Rc32 and Rd32 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Rh32 and R'32 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh32 and R'32 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra6°, Rb6°, Rc6° and Rd6° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R61; or any Rc6° and Rd6° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R61; each Rh6° and R'60 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh6° and R'60 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl;

204 each Ra61 , Rb61 , R061 and Rd61, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R061 and Rd61 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Rh61 and R'61 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh61 and R'61 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each Ra7°, Rb7°, Rc7° and Rd7° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; or any Rc7° and Rd7° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Rh7° and R'70 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any Rh7° and R'70 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; and each R9 is independently selected from D, OH, NO2, ON, halo, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, C3-6 cycloalkyl-Ci.2 alkylene, Ci-e alkoxy, Ci-e haloalkoxy, Ci-3 alkoxy-Ci-3 alkyl, Ci-3 alkoxy-Ci-3 alkoxy, HO-C1.3 alkoxy, HO-C1.3 alkyl, cyano-Ci-3 alkyl, H2N-C1.3 alkyl, amino, Ci-e alkylamino, di(Ci-6 alkyl)amino, thio, Ci-e alkylthio, Ci-6 alkylsulfinyl, Ci-e alkylsulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci-e alkylcarbonyl, Ci-e alkoxycarbonyl, Ci-e alkylcarbonylamino, Ci-e alkoxycarbonylamino, Ci-e alkylcarbonyloxy, aminocarbonyloxy, Ci-e alkylaminocarbonyloxy, di(Ci-6 alkyl)aminocarbonyloxy, Ci-e alkylsulfonylamino, aminosulfonyl, Ci-e alkylaminosulfonyl, di(Ci-6 alkyl)aminosulfonyl, aminosulfonylamino, Ci-e alkylaminosulfonylamino, di(Ci-6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci-e alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino.

3. The compound of claim 1 or 2, wherein the compound of Formula I is a compound of

Formula II:

205 or a pharmaceutically acceptable salt thereof, wherein:

-AR1=BR2- is selected from -N=CR2-, and -CR1=N-;

X is selected from N and CR5;

Y is selected from N and CR6;

R1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1 , NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1;

R2 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2; wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23;

Cy1 is selected from C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R10; R4 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa3, SRa3, C(O)Rb3, (O)ORa3, wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30;

R5 is selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa5, SRa5, C(O)Rb5,

R6 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa6, SRa6, C(O)Rb6, (O)ORa6, wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce- w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R60;

R7 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7C(O)NRc7Rd7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)2Rb7, and S(O)2NRc7Rd7;

Cy2 is selected from C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-

207 forming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4-

10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R20; each R10 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa1°, SRa1°, C(O)Rb1°, C(O)NRc10Rd1°, C(O)ORa1°, OC(O)Rb1°, OC(O)NRc10Rd1°, NRc10Rd1°, NRc10C(O)Rb1°, NRc10C(O)ORa1°, NRc10C(O)NRc10Rd1°, NRc10S(O)2Rb1°, NRc10S(O)2NRc10Rd1°, S(O)2Rb1°, and S(O)2NRc10Rd10; each R20 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa2°, SRa2°,

C(O)Rb2°, C(O)NRc20Rd2°, C(O)ORa2°, OC(O)Rb2°, OC(O)NRc20Rd2°, NRc20Rd2°,

NRc20C(O)R b20 , NRc20C(O)ORa2° , NRc20C(O)NRc2° Rd2° , NRc20S(O)2Rb2°, NRc20S(O)2NRc20Rd2°,

S(O)2Rb2°, S(O)2NRc20Rd2°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl,

4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3- cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21 ; each R21 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa21 , SRa21 , C(O)Rb21 , C(O)NRc21 Rd21 , C(O)ORa21 , OC(O)Rb21 , OC(O)NRc21 Rd21 , NRc21 Rd21 , NRc21C(O)Rb21 , NRc21C(O)ORa21 , NRc21C(O)NRc21 Rd21 , NRc21S(O)2Rb21 , NRc21S(O)2NRc21 Rd21 , S(O)2Rb21 , and S(O)2NRc21 Rd21 ; each R23 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa23, SRa23, C(O)Rb23, C(O)NRc23Rd23, C(O)ORa23, OC(O)Rb23, OC(O)NRc23Rd23, NRc23Rd23,

208 NRc23C(O)Rb23, NRc23C(O)ORa23, NRc23C(O)NRc23Rd23, NRc23S(O)2Rb23, NRc23S(O)2NRc23Rd23, S(O)2Rb23, and S(O)2NRc23Rd23; each R30 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa3°, SRa3°, S(O)2Rb3°, and S(O)2NRc30Rd3°; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31 ; each R31 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, ORa31 , SRa31 , each R60 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, ORa6°, SRa6°, each Ra1 , Rb1 , Rc1 , and Rd1 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-

10 membered heteroaryl; or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra2, Rb2, Rc2 and Rd2 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R23; each Ra3, Rb3, Rc3 and Rd3 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R30; each Ra5, Rb5, R05, and Rd5 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra6, Rb6, R06, and Rd6 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R60; or any Rc6 and Rd6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R60; each Ra7, Rb7, Rc7 and Rd7 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; or any Rc7 and Rd7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra1°, Rb1°, Rc1° and Rd1° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; or any Rc1° and Rd1° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra2°, Rb2°, Rc2° and Rd2° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W

210 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21; or any Rc20 and Rd20 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R21; each Ra21 , Rb21 , Rc21 and Rd21, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any Rc21 and Rd21 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra23, Rb23, Rc23 and Rd23, is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any Rc23 and Rd23 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra3°, Rb3°, Rc3° and Rd3° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31; or any Rc3° and Rd3° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R31; each Ra31 , Rb31 , R031 and Rd31, is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R031 and Rd31 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each Ra6°, Rb6°, Rc6° and Rd6° is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; and or any Rc6° and Rd6° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.

4. The compound of any one of claims 1-3, or a pharmaceutically acceptable salt thereof, wherein

-AR1=BR2- is selected from -N=CR2-, and -CR1=N-;

211 X is selected from N and CR5;

Y is selected from N and CR6;

R1 is selected from H, D, Ci-e alkyl, and Ci-e haloalkyl;

R2 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa2, and NRc2Rd2; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R23;

Cy1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4-10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2 or 3 substituents independently selected from R10;

R4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, D, CN, ORa3, C(O)NRc3Rd3and NRc3Rd3; wherein said Ci-e alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R30;

R5 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa5, and NR^R'15;

R6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, ORa6, and NRc6Rd6; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R60;

R7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, CN, ORa7, and NRc7Rd7;

Cy2 is selected from 4-7 membered heterocycloalkyl; wherein the 4-7 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 4-7 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R20; each R10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa1°, C(O)NRc10Rd1°, and NRc10Rd1°; each R20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa2°, C(O)Rb2°, C(O)NRc20Rd20and NRc20Rd2°; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R21;

212 each R21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa21, and NRc21Rd21; each R23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa23, and NRc23Rd23; each R30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, D, CN, ORa3°, C(O)NRc30Rd3°, and NRc30Rd3°; wherein said C1.6 alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R31; each R31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, ORa31, and NR^R^1; each R60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, D, CN, ORa6°, C(O)Rb6°, C(O)NRc60Rd6°, C(O)ORa6°, and NR^R'160; each Ra2, Rc2 and Rd2 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R23; each Ra3, Rb3, Rc3 and Rd3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl; wherein said C1.6 alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R30; or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R30; each Ra5, R05, and Rd5 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each Ra6, R06, and Rd6 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R60; each Ra7, Rc7 and Rd7 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each Ra1°, Rb1°, Rc1° and Rd1° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each Ra2°, Rb2°, Rc2° and Rd2° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R21; each Ra21 , Rc21 and Rd21 , is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl;

213 each Ra23, Rc23 and Rd23, is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each Ra3°, Rc3° and Rd3° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R31; or any Rc3° and Rd3° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R31; each Ra31 , R031 and Rd31 , is independently selected from H, Ci-e alkyl, Ci-e haloalkyl; each Ra6°, Rb6°, Rc6° and Rd6° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; and or any Rc6° and Rd6° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group.

5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein

-AR1=BR2- is selected from -N=CR2-, and -CR1=N-;

X is selected from N and CR5;

Y is selected from N and CR6;

R1 is H;

R2 is selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R23;

Cy1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R10;

R4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, CN, ORa3, and NRc3Rd3; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R30;

R5 is selected from H, Ci-e alkyl, and Ci-e haloalkyl;

R6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R60;

214 R7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, and CN;

Cy2 is selected from 4-6 membered heterocycloalkyl; wherein the 4-6 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 or 2 ring-forming heteroatoms independently selected from N, and O; and wherein the 4-6 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R20; each R10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and ORa1°; each R20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and C(O)Rb20; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R21; each R21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, ORa3°, C(O)NRc30Rd3°, and NRc30Rd3°; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R31; each R31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, ORa6°, C(O)Rb6°, C(O)NRc60Rd6°, C(O)ORa6°, and NRc60Rd6°; each Ra3, Rc3 and Rd3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R30; or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R30; each Ra1° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each Rb2° is independently selected from C2-6 alkenyl, and C2-6 alkynyl; wherein said C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R21; each Ra3°, Rc3° and Rd3° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R31;

215 or any Rc3° and Rd3° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R31; each Ra6°, Rb6°, Rc6° and Rd6° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; and or any Rc6° and Rd6° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group.

6. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt thereof, wherein

-AR1=BR2- is selected from -N=CR2-, and -CR1=N-;

X is N or CR5;

Y is N or CR6;

R1 is H;

R2 is Ci-6 alkyl optionally substituted with 1 , 2, or 3 substituents independently selected from R23;

Cy1 is selected from napthyl, benzothiazolyl, chromyl, phenyl, indolyl, indolinyl, tetrahydroquinolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally substituted with 1 , 2, or 3 substituents independently selected from R10;

R4 is selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, 6-10 membered heteroaryl, and ORa3; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl, and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R30;

R5 is H;

R6 is Ci-6 alkyl, 4-10 membered heterocycloalkyl, dihydroindolizinonyl, wherein Ci-e alkyl and 4-10 membered heterocycloalkyl are optionally substituted with 1 , 2, or 3 substituents independently selected from R60;

R7 is C1.3 alkyl or halo;

Cy2 is 4-14 membered heterocycloalkyl optionally substituted with 1 , 2, or 3 substituents independently selected from R20; each R10 is independently selected from Ci-e alkyl, halo, CN, and OH; each R20 is independently selected from Ci-e alkyl and C(O)Rb2°; wherein said Ci-e alkyl is optionally substituted with 1 , 2, or 3 substituents independently selected from R21; each R21 is independently selected from halo and CN; each R23 is CN;

216 each R30 is independently selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, C(O)NRc30Rd3°, and NRc30Rd3°; wherein said 4-10 membered heterocycloalkyl is optionally substituted with 1 , 2, or 3 substituents independently selected from R31; each R31 is independently selected from Ci-e alkyl and halo; each R60 is independently selected from 4-10 membered heterocycloalkyl, pyridinonyl, C(O)NRc60Rd6°, C(O)Ra6°, and C(O)ORa6°; each Ra3 is Ci-e alkyl optionally substituted with 1 , 2, or 3 substituents independently selected from R30; each Rb2° is C2-6 alkenyl optionally substituted with 1 , 2, or 3 substituents independently selected from R21; each Rc3° and Rd3° is independently H or Ci-e alkyl; and each Ra6°, Rc6° and Rd6° is independently H or Ci-e alkyl.

7. The compound of claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein — represents a double bond.

8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt thereof, wherein -AR1=BR2- is -N=CR2-.

9. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt thereof, wherein -AR1=BR2- is -CR1=N-

10. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein

X is CR5; and

Y is CR6.

11. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein

X is N; and

Y is CR6.

12. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein

X is N; and

Y is N.

217

13. The compound of any one of claims 1-7 and 9-12, or a pharmaceutically acceptable salt thereof, wherein R1 is H.

14. The compound of any one of claims 1-5, 7, 8, and 10-13, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from H, Ci-e alkyl, and Ci-e haloalkyl.

15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt thereof, wherein R2 is Ci-e alkyl optionally substituted with CN.

16. The compound of any one of claims 1-15, or a pharmaceutically acceptable salt thereof, wherein Cy1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R10.

17. The compound of any one of claims 1-15, or a pharmaceutically acceptable salt thereof, wherein Cy1 is selected from 5-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 5-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 5-10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the 5-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R10.

18. The compound of any one of claims 1-17, or a pharmaceutically acceptable salt thereof, wherein R3N— CR4 is a double bond, and R3 is absent.

19. The compound of any one of claims 1-5 and 7-18, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, and ORa3; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl,

218 and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R30.

20. The compound of any one of claims 1-10 and 13-19, or a pharmaceutically acceptable salt thereof, wherein R5 is H.

21. The compound of any one of claims 1-5 and 7-20, or a pharmaceutically acceptable salt thereof, wherein R6 is selected from Ci-e alkyl and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R60.

22. The compound of any one of claims 1-5 and 7-21 , or a pharmaceutically acceptable salt thereof, wherein R7 is selected from H, C1.3 alkyl, and halo.

23. The compound of any one of claims 1-3 and 7-22, or a pharmaceutically acceptable salt thereof, wherein Cy2 is 5-10 membered heterocycloalkyl optionally substituted with 1 or 2 substituents independently selected from R20.

24. The compound of any one of claims 1-3 and 4-23, or a pharmaceutically acceptable salt thereof, wherein Cy2 is 4-6 membered heterocycloalkyl optionally substituted with 1 or 2 substituents independently selected from R20.

25. The compound of any one of claims 1-24, or a pharmaceutically acceptable salt thereof, wherein each R10 is independently selected from Ci-e alkyl, halo, CN, and OH.

26. The compound of any one of claims 1-25, or a pharmaceutically acceptable salt thereof, wherein each R20 is independently selected from Ci-e alkyl and C(O)C2-6 alkenyl; wherein said Ci-e alkyl and C2-6 alkenyl are each optionally substituted with 1 or 2 substituents independently selected from CN and halo.

27. The compound of any one of claims 1-26, or a pharmaceutically acceptable salt thereof, wherein each R30 is independently selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, C(O)NRc30Rd3°, and NRc30Rd3°; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R31.

219

28. The compound of any one of claims 1-27, or a pharmaceutically acceptable salt thereof, wherein each R60 is independently selected from 4-10 membered heterocycloalkyl, C(O)Rb6°, C(O)NRc60Rd6°, and C(O)ORa6°.

29. The compound of any one of claims 1-28, or a pharmaceutically acceptable salt thereof, wherein each Ra3 is Ci-e alkyl optionally substituted with 1or 2 substituents independently selected from R30; and each R30 is 4-10 membered heterocycloalkyl optionally substituted with 1 or 2 substituents independently selected from Ci-e alkyl.

30. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt thereof, wherein each Rc3° and Rd3° is independently selected from H and Ci-e alkyl; wherein said Ci-6 alkyl is optionally substituted with 1 or 2 substituents independently selected from R31.

31. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt thereof, wherein Ra6°, Rc6° and Rd6° is Ci-e alkyl.

32. The compound of any one of claims 1-31 , wherein the compound of Formula I is selected from: methyl 3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3-hydroxynaphthalen- 1-yl)-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)propanoate; methyl 3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-4-(((2R,7aS)-2- fluorotetrahydro-1 H-pyrrolizin-7a(5H)-yl)methoxy)-7-(3-hydroxynaphthalen-1-yl)-1 H- pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)propanoate;

4-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-methyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-7-yl)naphthalen-2-ol;

4-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-methyl-4-((tetrahydro-1 H- pyrrolizin-7a(5H)-yl)methoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-7-yl)naphthalen-2-ol;

3-((1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-4- methyl-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)methyl)oxazolidin-2-one;

3-((1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(5-fluoro-1 H-indol-3-yl)-4-((S)- 1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)methyl)oxazolidin-2-one;

220 3-((1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(3,4-dihydroquinolin-1 (2H)-yl)-6-fluoro- 4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)methyl)oxazolidin-2-one;

1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(indolin-1-yl)-2-methyl-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridine;

3-(1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-2-yl)-N,N- dimethylpropanamide;

3-(1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(3-cyano-2,3-dihydro-1 H-inden-4-yl)-4- (3-(dimethylamino)azetidin-1-yl)-6-fluoro-1 H-imidazo[4,5-c][1 ,6]naphthyridin-2-yl)-N,N- dimethylpropanamide; methyl 3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-4-(3-(dimethylamino)azetidin-1- yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-2- yl)propanoate;

2-((2S,4S)-4-(4-(3-(ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-7-(5-fluoroquinolin-8- yl)-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)-1-((E)-4-fluorobut-2-enoyl)piperidin-2- yl)acetonitrile;

2-((2S,4S)-4-(7-(5,6-dimethyl-1 H-indazol-3-yl)-4-(3-(ethyl(methyl)amino)azetidin-1- yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)-1-(2-fluoroacryloyl)piperidin-2- yl)acetonitrile;

8-(1-((2S,4S)-2-(cyanomethyl)-1-(2-fluoroacryloyl)piperidin-4-yl)-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-7-yl)-1- naphthonitrile;

8-(1-((2S,4S)-2-(cyanomethyl)-1-((E)-4-fluorobut-2-enoyl)piperidin-4-yl)-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-7-yl)-1- naphthonitrile;

3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-4-(3-(dimethylamino)azetidin-1-yl)-7-(3- hydroxynaphthalen-1-yl)-6,8-dimethyl-1/7-imidazo[4,5-c][1 ,5]naphthyridin-2-yl)-N,N- dimethylpropanamide; and

3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-8-(2-cyanoethyl)-4-(3- (dimethylamino)azetidin-1-yl)-7-(3-hydroxynaphthalen-1-yl)-1/7-imidazo[4,5- c][1 ,5]naphthyridin-2-yl)-N,N-dimethylpropanamide; or a pharmaceutically acceptable salt thereof.

33. The compound of any one of claims 1-31 , wherein the compound of Formula I is selected from:

221 4-(1-((7/?,4/?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-methyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-7-yl)-6-fluoronaphthalen-2-ol;

1-((/?)-2-(7-(Benzo[d]thiazol-4-yl)-1-((7/?,4/?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidin-1-yl)ethan-1-one;

3-(1-((7F?,4F?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-7-(chroman-8-yl)-6-fluoro-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)-2,3- dihydroindolizin-5(1/7)-one;

1-(( ?)-1-(1-((7 ?,4 ?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-7-(chroman-8-yl)-6-fluoro-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2- y l)ethy I) py rid i n-2( 1 /-/)-one;

Methyl (F?)-2-(1-((7F?,4F?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2,3-dichlorophenyl)-4- (3-(dimethylamino)-3-methylazetidin-1-yl)-6-fluoro-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidine-1 -carboxylate;

Methyl (F?)-2-(1-((7F?,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2,3-dichlorophenyl)-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidine-1 -carboxylate; and

Methyl (R)-2-(1-((7R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2-chloro-3- methylphenyl)-6-fluoro-4-(6-(methylcarbamoyl)pyridin-2-yl)-1/7-pyrrolo[3,2- c][1 ,6]naphthyridin-2-yl)pyrrolidine-1 -carboxylate; or a pharmaceutically acceptable salt thereof.

34. The compound of any one of claims 1-31 , wherein the compound of Formula I is selected from: methyl (R)-2-(7-(benzo[d]thiazol-4-yl)-1-((1R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)- 6-fluoro-4-(((S)-1-(pyrrolidin-1-yl)propan-2-yl)oxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidine-1 -carboxylate;

(7aS)-5-(1-((1R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(chroman-8-yl)-6-fluoro-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)tetrahydro- 1/7,3/7-pyrrolo[1 ,2-c]oxazol-3-one; and

4-(1-((1R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(chroman-8-yl)-6-fluoro-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 /7-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)-/V,/\/, 1 -trimethyl- 1 /7-pyrazole-3-carboxamide; or a pharmaceutically acceptable salt thereof.

222

35. A pharmaceutical composition comprising a compound of any one of claims 1-34, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient.

36. A method of inhibiting KRAS activity, said method comprising contacting a compound of any one of claims 1-34, or a pharmaceutically acceptable salt thereof, or the composition of claim 35, with KRAS.

37. The method of claim 36, wherein the contacting comprises administering the compound to a patient.

38. A method of treating a disease or disorder associated with inhibition of KRAS interaction, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of claims 1-34, or a pharmaceutically acceptable salt thereof, or the composition of claim 35.

39. The method of claim 38, wherein the disease or disorder is an immunological or inflammatory disorder.

40. The method of claim 39, wherein the immunological or inflammatory disorder is Ras- associated lymphoproliferative disorder and juvenile myelomonocytic leukemia caused by somatic mutations of KRAS.

41. A method for treating a cancer in a patient, said method comprising administering to the patient a therapeutically effective amount of the compound of any one of claims 1-34, or a pharmaceutically acceptable salt thereof, or the composition of claim 35.

42. The method of claim 41, wherein the cancer is selected from carcinomas, hematological cancers, sarcomas, and glioblastoma.

43. The method of claim 42, wherein the hematological cancer is selected from myeloproliferative neoplasms, myelodysplastic syndrome, chronic and juvenile myelomonocytic leukemia, acute myeloid leukemia, acute lymphocytic leukemia, and multiple myeloma.

223

44. The method of claim 42, wherein the carcinoma is selected from pancreatic, colorectal, lung, bladder, gastric, esophageal, breast, head and neck, cervical, skin, and thyroid. 45. A method of treating a disease or disorder associated with inhibiting a KRAS protein harboring a G12V mutation, said method comprising administering to a patient in need thereof a therapeutically effective amount of the compound of any one of claims 1-34, or a pharmaceutically acceptable salt thereof, or the composition of claim 35.

224

Description:
NAPHTHYRIDINE COMPOUNDS AS INHIBITORS OF KRAS

RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application No. 63/239,152 filed on August 31, 2021 and U.S. Provisional Application No. 63/364,593 filed on May 12, 2022, the entire contents of which are hereby incorporated by reference in their entirety.

FIELD OF THE INVENTION

The disclosure provides compounds as well as their compositions and methods of use. The compounds modulate KRAS activity and are useful in the treatment of various diseases including cancer.

BACKGROUND OF THE INVENTION

Ras proteins are part of the family of small GTPases that are activated by growth factors and various extracellular stimuli. The Ras family regulates intracellular signaling pathways responsible for growth, migration, survival and differentiation of cells. Activation of RAS proteins at the cell membrane results in the binding of key effectors and initiation of a cascade of intracellular signaling pathways within the cell, including the RAF and PI3K kinase pathways. Somatic mutations in RAS may result in uncontrolled cell growth and malignant transformation while the activation of RAS proteins is tightly regulated in normal cells (Simanshu, D. et al. Cell 170.1 (2017): 17-33).

The Ras family is comprised of three members: KRAS, NRAS and HRAS. RAS mutant cancers account for about 25% of human cancers. KRAS is the most frequently mutated isoform accounting for 85% of all RAS mutations whereas NRAS and HRAS are found mutated in 12% and 3% of all Ras mutant cancers respectively (Simanshu, D. et al. Cell 170.1 (2017): 17-33). KRAS mutations are prevalent amongst the top three most deadly cancer types: pancreatic (97%), colorectal (44%), and lung (30%) (Cox, A.D. et al. Nat Rev Drug Discov (2014) 13:828-51). The majority of RAS mutations occur at amino acid residue 12, 13, and 61. The frequency of specific mutations varies between RAS gene isoforms and while G12 and Q61 mutations are predominant in KRAS and NRAS respectively, G12, G13 and Q61 mutations are most frequent in HRAS. Furthermore, the spectrum of mutations in a RAS isoform differs between cancer types. For example, KRAS G12D mutations predominate in pancreatic cancers (51%), followed by colorectal adenocarcinomas (45%) and lung cancers (17%) while KRAS G12 V mutations are associated with pancreatic cancers (30%), followed by colorectal adenocarcinomas (27%) and lung adenocarcinomas (23%) (Cox, A.D. et al. Nat Rev Drug Discov (2014) 13:828-51). In contrast, KRAS G12C mutations predominate in non-small cell lung cancer (NSCLC) comprising 11-16% of lung adenocarcinomas, and 2-5% of pancreatic and colorectal adenocarcinomas (Cox, A.D. et al. Nat. Rev. Drug Discov. (2014) 13:828-51). Genomic studies across hundreds of cancer cell lines have demonstrated that cancer cells harboring KRAS mutations are highly dependent on KRAS function for cell growth and survival (McDonald, R. et al. Cell 170 (2017): 577- 592). The role of mutant KRAS as an oncogenic driver is further supported by extensive in vivo experimental evidence showing mutant KRAS is required for early tumour onset and maintenance in animal models (Cox, A.D. et al. Nat Rev Drug Discov (2014) 13:828-51).

Taken together, these findings suggest that KRAS mutations play a critical role in human cancers; development of inhibitors targeting mutant KRAS may therefore be useful in the clinical treatment of diseases that are characterized by a KRAS mutation.

SUMMARY

The present disclosure provides, inter alia, a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein constituent variables are defined herein.

The present disclosure further provides a pharmaceutical composition comprising a compound of the disclosure, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient.

The present disclosure further provides methods of inhibiting KRAS activity, which comprises administering to an individual a compound of the disclosure, or a pharmaceutically acceptable salt thereof. The present disclosure also provides uses of the compounds described herein in the manufacture of a medicament for use in therapy. The present disclosure also provides the compounds described herein for use in therapy.

The present disclosure further provides methods of treating a disease or disorder in a patient comprising administering to the patient a therapeutically effective amount of a compound of the disclosure, or a pharmaceutically acceptable salt thereof. DETAILED DESCRIPTION

Compounds

In an aspect, provided herein is a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein:

— represents a single bond or a double bond;

-AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, OR a1 , SR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , OC(O)R b1 , OC(O)NR c1 R d1 , NR c1 R d1 , NR c1 C(O)R b1 , NR c1 C(O)OR a1 , NR c1 C(O)NR c1 R d1 , NR c1 S(O)R b1 , NR c1 S(O) 2 R b1 , NR c1 S(O) 2 NR c1 R d1 , S(O)R b1 , S(O)NR c1 R d1 , S(O) 2 R b1 , S(O) 2 NR c1 R d1 , and BR h1 R i1 ; wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ;

R 2 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a2 , SR a2 , C(O)R b2 , C(O)NR c2 R d2 , C(O)OR a2 , OC(O)R b2 , OC(O)NR c2 R d2 , NR c2 R d2 , NR c2 C(O)R b2 , NR c2 C(O)OR a2 , NR c2 C(O)NR c2 R d2 , C(=NR e2 )R b2 , C(=NOR a2 )R b2 , C(=NR e2 )NR c2 R d2 , NR c2 C(=NR e2 )NR c2 R d2 , NR c2 C(=NR e2 )R b2 , NR c2 S(O)R b2 , NR c2 S(O) 2 R b2 , NR c2 S(O) 2 NR c2 R d2 , S(O)R b2 , S(O)NR c2 R d2 , S(O)2R b2 , S(O)2NR c2 R d2 , and BR h2 R' 2 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 10 ; when R 3 N— CR 4 is a single bond, then R 4 is selected from =0 and =S; and

R 3 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, and 5-10 membered heteroaryl-Ci.3 alkylene; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; when R 3 N— CR 4 is a double bond, then R 3 is absent; and

R 4 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a3 , SR a3 , C(O)R b3 , C(O)NR c3 R d3 , C(O)OR a3 , OC(O)R b3 , OC(O)NR c3 R d3 , NR^R^, NR c3 C(O)R b3 , NR c3 C(O)OR a3 , NR c3 C(O)NR c3 R d3 , C(=NR e3 )R b3 , C(=NOR a3 )R b3 , C(=NR e3 )NR c3 R d3 , NR c3 C(=NR e3 )NR c3 R d3 , NR c3 C(=NR e3 )R b3 , NR c3 S(O)R b3 , NR c3 S(O) 2 R b3 , NR c3 S(O) 2 NR c3 R d3 , S(O)R b3 , S^NR^R^, S(O)2R b3 , S(O)2NR c3 R d3 , and BR h3 R' 3 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ;

R 5 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a5 , SR a5 , C(O)R b5 , NR c5 C(=NR e5 )R b5 , NR c5 S(O)R b5 , NR c5 S(O) 2 R b5 , NR c5 S(O) 2 NR c5 R d5 , S(O)R b5 , S^NR^R^, S(O) 2 R b5 , S(O) 2 NR c5 R d5 , and BR h5 R' 5 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ;

R 6 is selected from H, Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, NO 2 , OR a6 , SR a6 , C(O)R b6 , C(O)NR c6 R d6 , C(O)OR a6 , OC(O)R b6 , OC(O)NR c6 R d6 , NR^R^, NR c6 C(O)R b6 , NR c6 C(O)OR a6 , NR c6 C(O)NR c6 R d6 , C(=NR e6 )R b6 , C(=NOR a6 )R b6 , C(=NR e6 )NR c6 R d6 , NR c6 C(=NR e6 )NR c6 R d6 , NR c6 C(=NR e6 )R b6 , NR c6 S(O)R b6 , NR c6 S(O) 2 R b6 , NR c6 S(O) 2 NR c6 R d6 , S(O)R b6 , S^NR^R^, S(O) 2 R b6 , S(O) 2 NR c6 R d6 , and BR h6 R' 6 ; wherein said Ci-e alkyl, C 2 -6 alkenyl, C 2 -6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)OR a7 , NR c7 C(O)NR c7 R d7 , C(=NR e7 )R b7 , C(=NOR a7 )R b7 , C(=NR e7 )NR c7 R d7 , NR c7 C(=NR e7 )NR c7 R d7 , NR c7 C(=NR e7 )R b7 , NR c7 S(O)R b7 , NR c7 S(O) 2 R b7 , NR c7 S(O) 2 NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O) 2 R b7 , S(O) 2 NR c7 R d7 , and BR h7 R' 7 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 70 ;

Cy 2 is selected from C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-14 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4- 14 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a1 °, SR a1 °, C(O)R b1 °, C(O)NR c10 R d1 °, C(O)OR a1 °, OC(O)R b1 °, OC(O)NR c10 R d1 °, NR c10 R d1 °, NR c10 C(O)R b1 °, NR c10 C(O)OR a1 °, NR c10 C(O)NR c10 R d1 °, C(=NR e10 )R b1 °, C(=NOR a10 )R b1 °, C(=NR e10 )NR c10 R d1 °, NR c10 C(=NR e10 )NR c10 R d1 °, NR c10 S(O)R b1 °, NR c10 S(O) 2 R b1 °, NR c10 S(O)2NR c10 R d1 °, S(O)R b1 °, S(O)NR c10 R d1 °, S(O) 2 R b1 °, S(O) 2 NR c10 R d10 , and BR h10 R i1 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 11 ; each R 11 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a11 , SR a11 , C(O)R b11 , C(O)NR c11 R d11 , C(O)OR a11 , OC(O)R b11 , OC(O)NR c11 R d11 , NR c11 R d11 , NR c11 C(O)R b11 , NR c11 C(O)OR a11 , NR c11 C(O)NR c11 R d11 , NR c11 S(O)R b11 , NR c11 S(O) 2 R b11 , NR c11 S(O) 2 NR c11 R d11 , S(O)R b11 , S(O)NR c11 R d11 , S(O) 2 R b11 , S(O) 2 NR c11 R d11 , and BR h11 R' 11 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 12 ; each R 12 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, OR a12 , SR a12 , C(O)R b12 , C(O)NR c12 R d12 , C(O)OR a12 , OC(O)R b12 , OC(O)NR c12 R d12 , NR c12 R d12 , NR c12 C(O)R b12 , NR c12 C(O)OR a12 , NR c12 C(O)NR c12 R d12 , NR c12 S(O)R b12 , NR c12 S(O) 2 R b12 , NR c12 S(O) 2 NR c12 R d12 , S(O)R b12 , S(O)NR c12 R d12 , S(O) 2 R b12 , S(O)2NR c12 R d12 , and BR h12 R' 12 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R 20 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a2 °, SR a2 °,

C(O)R b2 °, C(O)NR c20 R d2 °, C(O)OR a2 °, OC(O)R b2 °, OC(O)NR c20 R d2 °, NR c20 R d2 °,

NR c20 C(O)R b2 °, NR c20 C(O)OR a2 °, NR c20 C(O)NR c20 R d2 °, C(=NR e20 )R b2 °, C(=NOR a20 )R b2 °,

C(=NR e20 )NR c20 R d2 °, NR c20 C(=NR e20 )NR c20 R d2 °, NR c20 S(O)R b2 °, NR c20 S(O) 2 R b2 °, NR c20 S(O) 2 NR c20 R d2 °, S(O)R b2 °, S(O)NR c20 R d2 °, S(O) 2 R b2 °, S(O) 2 NR c20 R d2 °, and BR h20 R i2 °; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a21 , SR a21 , C(O)R b21 , C(O)NR c21 R d21 , C(O)OR a21 , OC(O)R b21 , OC(O)NR c21 R d21 , NR c21 R d21 , NR c21 C(O)R b21 , NR c21 C(O)OR a21 , NR c21 C(O)NR c21 R d21 , NR c21 S(O)R b21 , NR c21 S(O) 2 R b21 , NR c21 S(O) 2 NR c21 R d21 , S(O)R b21 , S(O)NR c21 R d21 , S(O) 2 R b21 , S(O) 2 NR c21 R d21 , and BR h21 R' 21 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 22 ; each R 22 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, OR a22 , SR a22 , C(O)R b22 , C(O)NR c22 R d22 , C(O)OR a22 , OC(O)R b22 , OC(O)NR c22 R d22 , NR c22 R d22 , NR c22 C(O)R b22 , NR c22 C(O)OR a22 , NR c22 C(O)NR c22 R d22 , NR c22 S(O)R b22 , NR c22 S(O) 2 R b22 , NR c22 S(O) 2 NR c22 R d22 , S(O)R b22 , S(O)NR c22 R d22 , S(O) 2 R b22 , S(O) 2 NR c22 R d22 , and BR h22 R' 22 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R 23 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a23 , SR a23 , C(O)R b23 , C(O)NR c23 R d23 , C(O)OR a23 , OC(O)R b23 , OC(O)NR c23 R d23 , NR c23 R d23 , NR c23 C(O)R b23 , NR c23 C(O)OR a23 , NR c23 C(O)NR c23 R d23 , NR c23 S(O)R b23 , NR c23 S(O) 2 R b23 , NR c23 S(O) 2 NR c23 R d23 , S(O)R b23 , S(O)NR c23 R d23 , S(O) 2 R b23 , S(O) 2 NR c23 R d23 , and BR h23 R' 23 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 24 ; each R 24 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, OR a24 , SR a24 , C(O)R b24 , C(O)NR c24 R d24 , C(O)OR a24 , OC(O)R b24 , OC(O)NR c24 R d24 , NR c24 R d24 , NR c24 C(O)R b24 , NR c24 C(O)OR a24 , NR c24 C(O)NR c24 R d24 , NR c24 S(O)R b24 , NR c24 S(O) 2 R b24 , NR c24 S(O) 2 NR c24 R d24 , S(O)R b24 , S(O)NR c24 R d24 , S(O) 2 R b24 , S(O)2NR c24 R d24 , and BR h24 R' 24 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R 30 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a3 °, SR a3 °, C(O)R b3 °, C(O)NR c30 R d3 °, C(O)OR a3 °, OC(O)R b3 °, OC(O)NR c30 R d3 °, NR^R^ 0 , NR c30 C(O)R b3 °, NR c30 C(O)OR a3 °, NR^C^NR^R^ 0 , NR c30 S(O)R b3 °, NR c30 S(O) 2 R b3 °, NR c30 S(O)2NR c30 R d3 °, S(O)R b3 °, S^NR^R^ 0 , S(O) 2 R b3 °, S(O) 2 NR c30 R d30 , and BR h30 R i3 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R 31 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a31 , SR a31 , wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 32 ; each R 32 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, S(O)2NR c32 R d32 , and BR h32 R' 32 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R 60 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-io cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a6 °, SR a6 °, C(O)R b6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, OC(O)R b6 °, OC(O)NR c60 R d6 °, NR^R^ 0 , NR c60 C(O)R b6 °, NR c60 C(O)OR a6 °, NR c60 C(O)NR c60 R d6 °, NR c60 S(O)R b6 °, NR c60 S(O) 2 R b6 °, NR c60 S(O)2NR c60 R d6 °, S(O)R b6 °, S^NR^R^ 0 , S(O) 2 R b6 °, S(O) 2 NR c60 R d60 , and BR h60 R i6 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ciw alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 61 ; each R 61 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a61 , SR a61 , C(O)R b61 , C(O)NR c61 R d61 , C(O)OR a61 , OC(O)R b61 , OC(O)NR c61 R d61 , NR^ R^ 1 , NR c61 C(O)R b61 , NR c61 C(O)OR a61 , NR c61 C(O)NR c61 R d61 , NR c61 S(O)R b61 , NR c61 S(O) 2 R b61 , NR c61 S(O) 2 NR c61 R d61 , S(O)R b61 , S^NR^ R^ 1 , S(O) 2 R b61 , S(O) 2 NR c61 R d61 , and BR h61 R' 61 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 62 ; each R 62 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, S(O)2NR c62 R d62 , and BR h62 R' 62 ; wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R 70 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a7 °, SR a7 °,

C(O)R b7 °, C(O)NR c70 R d7 °, C(O)OR a7 °, OC(O)R b7 °, OC(O)NR c70 R d7 °, NR c70 R d7 °,

NR c70 C(O)R b70 , NR c70 C(O)OR a7 °, NR c70 C(O)NR c70 R d7 °, NR c70 S(O)R b7 °, NR c70 S(O) 2 R b7 °, NR c70 S(O)2NR c70 R d7 °, S(O)R b7 °, S(O)NR c70 R d7 °, S(O) 2 R b7 °, S(O) 2 NR c70 R d70 , and BR h70 R i7 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R a1 , R b1 , R c1 , and R d1 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c1 and R d1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 9 ; each R h1 and R i1 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h1 and R i1 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and C1.6 haloalkyl; each R a2 , R b2 , R c2 and R d2 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; or any R c2 and R d2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; each R e2 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h2 and R' 2 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h2 and R' 2 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a3 , R b3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; each R e3 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h3 and R' 3 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h3 and R' 3 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a5 , R b5 , R 05 , and R d5 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c5 and R d5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 9 ; each R e5 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h5 and R' 5 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h5 and R' 5 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a6 , R b6 , R 06 , and R d6 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ; or any R c6 and R d6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 60 ; each R e6 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h6 and R' 6 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h6 and R' 6 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a7 , R b7 , R c7 and R d7 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 70 ; or any R c7 and R d7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 70 ; each R e7 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h7 and R' 7 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h7 and R' 7 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a1 °, R b1 °, R c1 ° and R d1 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 11 ; or any R c1 ° and R d1 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 11 ; each R e1 ° is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h1 ° and R' 10 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h1 ° and R' 10 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a11 , R b11 , R c11 and R d11 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 12 ; or any R c11 and R d11 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 12 ; each R h11 and R' 11 is independently selected from OH, C1-6 alkoxy, and C1-6 haloalkoxy; or any R h11 and R' 11 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from C1-6 alkyl and C1-6 haloalkyl; each R a12 , R b12 , R c12 and R d12 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R h12 and R' 12 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h12 and R' 12 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a2 °, R b2 °, R c2 ° and R d2 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; or any R c20 and R d20 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R e20 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h20 and R' 20 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h20 and R' 20 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a21 , R b21 , R c21 and R d21 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 22 ; or any R c21 and R d21 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 22 ; each R h21 and R' 21 is independently selected from OH, C1-6 alkoxy, and C1-6 haloalkoxy; or any R h21 and R' 21 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from C1-6 alkyl and C1-6 haloalkyl; each R a22 , R b22 , R c22 and R d22 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R h22 and R' 22 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h22 and R' 22 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a23 , R b23 , R c23 and R d23 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 24 ; or any R c23 and R d23 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 24 ; each R h23 and R' 23 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h23 and R' 23 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a24 , R b24 , R c24 and R d24 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R h24 and R' 24 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h24 and R' 24 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a3 °, R b3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R h3 ° and R' 30 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h3 ° and R' 30 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a31 , R b31 , R 031 and R d31 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 32 ; or any R 031 and R d31 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 32 ; each R h31 and R' 31 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h31 and R' 31 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a32 , R b32 , R c32 and R d32 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c32 and R d32 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R h32 and R' 32 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h32 and R' 32 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a6 °, R b6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 61 ; or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 61 ; each R h6 ° and R' 60 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h6 ° and R' 60 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a61 , R b61 , R 061 and R d61 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 62 ; or any R 061 and R d61 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 62 ; each R h61 and R' 61 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h61 and R' 61 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a62 , R b62 , R c62 and R d62 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c62 and R d62 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R h62 and R' 62 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h62 and R' 62 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a7 °, R b7 °, R c7 ° and R d7 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c7 ° and R d7 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; each R h7 ° and R' 70 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h7 ° and R' 70 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; and each R 9 is independently selected from D, OH, NO2, ON, halo, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, C3-6 cycloalkyl-Ci.2 alkylene, Ci-e alkoxy, Ci-e haloalkoxy, Ci-3 alkoxy-Ci-3 alkyl, Ci-3 alkoxy-Ci-3 alkoxy, HO-C1.3 alkoxy, HO-C1.3 alkyl, cyano-Ci-3 alkyl, H2N-C1.3 alkyl, amino, Ci-e alkylamino, di(Ci-6 alkyl)amino, thio, Ci-e alkylthio, C1.6 alkylsulfinyl, Ci-e alkylsulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci-e alkylcarbonyl, Ci-e alkoxycarbonyl, Ci-e alkylcarbonylamino, Ci-e alkoxycarbonylamino, Ci-e alkylcarbonyloxy, aminocarbonyloxy, Ci-e alkylaminocarbonyloxy, di(Ci-6 alkyl)aminocarbonyloxy, Ci-e alkylsulfonylamino, aminosulfonyl, Ci-e alkylaminosulfonyl, di(Ci-6 alkyl)aminosulfonyl, aminosulfonylamino, Ci-e alkylaminosulfonylamino, di(Ci-6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci-e alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino; provided that,

(a) Cy 1 is other than 3,5-dimethylisoxazol-4-yl; and

(b) R 3 is other than CH2CF3.

In an embodiment of Formula I, or a pharmaceutically acceptable salt thereof,

— represents a single bond or a double bond;

-AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, OR a1 , SR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , OC(O)R b1 , OC(O)NR c1 R d1 , NR c1 R d1 , NR c1 C(O)R b1 , NR c1 C(O)OR a1 , NR c1 C(O)NR c1 R d1 , NR c1 S(O)R b1 , NR c1 S(O) 2 R b1 , NR c1 S(O) 2 NR c1 R d1 , S(O)R b1 , S(O)NR c1 R d1 , S(O) 2 R b1 , S(O) 2 NR c1 R d1 , and BR h1 R i1 ; wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ;

R 2 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a2 , SR a2 , C(O)R b2 , C(O)NR c2 R d2 , C(O)OR a2 , OC(O)R b2 , OC(O)NR c2 R d2 , NR c2 R d2 , NR c2 C(O)R b2 , NR c2 C(O)OR a2 , NR c2 C(O)NR c2 R d2 , C(=NR e2 )R b2 , C(=NOR a2 )R b2 , C(=NR e2 )NR c2 R d2 , NR c2 C(=NR e2 )NR c2 R d2 , NR c2 C(=NR e2 )R b2 , NR c2 S(O)R b2 , NR c2 S(O) 2 R b2 , NR c2 S(O) 2 NR c2 R d2 , S(O)R b2 , S(O)NR c2 R d2 , S(O)2R b2 , S(O)2NR c2 R d2 , and BR h2 R' 2 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 10 ; when R 3 N— CR 4 is a single bond, then R 4 is selected from =0 and =S; and

R 3 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, and 5-10 membered heteroaryl-Ci.3 alkylene; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; when R 3 N— CR 4 is a double bond, then R 3 is absent; and

R 4 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a3 , SR a3 , C(O)R b3 , C(O)NR c3 R d3 , C(O)OR a3 , OC(O)R b3 , OC(O)NR c3 R d3 , NR^R^, NR c3 C(O)R b3 , NR c3 C(O)OR a3 , NR c3 C(O)NR c3 R d3 , C(=NR e3 )R b3 , C(=NOR a3 )R b3 , C(=NR e3 )NR c3 R d3 , NR c3 C(=NR e3 )NR c3 R d3 , NR c3 C(=NR e3 )R b3 , NR c3 S(O)R b3 , NR c3 S(O) 2 R b3 , NR c3 S(O) 2 NR c3 R d3 , S(O)R b3 , S^NR^R^, S(O)2R b3 , S(O)2NR c3 R d3 , and BR h3 R' 3 ; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ;

R 5 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a5 , SR a5 , C(O)R b5 , NR c5 C(=NR e5 )R b5 , NR c5 S(O)R b5 , NR c5 S(O) 2 R b5 , NR c5 S(O) 2 NR c5 R d5 , S(O)R b5 , S^NR^R^, S(O) 2 R b5 , S(O) 2 NR c5 R d5 , and BR h5 R' 5 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ;

R 6 is selected from H, Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, NO 2 , OR a6 , SR a6 , C(O)R b6 , C(O)NR c6 R d6 , C(O)OR a6 , OC(O)R b6 , OC(O)NR c6 R d6 , NR^R^, NR c6 C(O)R b6 , NR c6 C(O)OR a6 , NR c6 C(O)NR c6 R d6 , C(=NR e6 )R b6 , C(=NOR a6 )R b6 , C(=NR e6 )NR c6 R d6 , NR c6 C(=NR e6 )NR c6 R d6 , NR c6 C(=NR e6 )R b6 , NR c6 S(O)R b6 , NR c6 S(O) 2 R b6 , NR c6 S(O) 2 NR c6 R d6 , S(O)R b6 , S^NR^R^, S(O) 2 R b6 , S(O) 2 NR c6 R d6 , and BR h6 R' 6 ; wherein said Ci-e alkyl, C 2 -6 alkenyl, C 2 -6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)OR a7 , NR c7 C(O)NR c7 R d7 , C(=NR e7 )R b7 , C(=NOR a7 )R b7 , C(=NR e7 )NR c7 R d7 , NR c7 C(=NR e7 )NR c7 R d7 , NR c7 C(=NR e7 )R b7 , NR c7 S(O)R b7 , NR c7 S(O) 2 R b7 , NR c7 S(O) 2 NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O) 2 R b7 , S(O) 2 NR c7 R d7 , and BR h7 R' 7 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 70 ;

Cy 2 is selected from C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-14 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4- 14 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-14 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a1 °, SR a1 °, C(O)R b1 °, C(O)NR c10 R d1 °, C(O)OR a1 °, OC(O)R b1 °, OC(O)NR c10 R d1 °, NR c10 R d1 °, NR c10 C(O)R b1 °, NR c10 C(O)OR a1 °, NR c10 C(O)NR c10 R d1 °, C(=NR e10 )R b1 °, C(=NOR a10 )R b1 °, C(=NR e10 )NR c10 R d1 °, NR c10 C(=NR e10 )NR c10 R d1 °, NR c10 S(O)R b1 °, NR c10 S(O) 2 R b1 °, NR c10 S(O)2NR c10 R d1 °, S(O)R b1 °, S(O)NR c10 R d1 °, S(O) 2 R b1 °, S(O) 2 NR c10 R d10 , and BR h10 R i1 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 11 ; each R 11 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a11 , SR a11 , C(O)R b11 , C(O)NR c11 R d11 , C(O)OR a11 , OC(O)R b11 , OC(O)NR c11 R d11 , NR c11 R d11 , NR c11 C(O)R b11 , NR c11 C(O)OR a11 , NR c11 C(O)NR c11 R d11 , NR c11 S(O)R b11 , NR c11 S(O) 2 R b11 , NR c11 S(O) 2 NR c11 R d11 , S(O)R b11 , S(O)NR c11 R d11 , S(O) 2 R b11 , S(O) 2 NR c11 R d11 , and BR h11 R' 11 ; each R 20 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO2, OR a2 °, SR a2 °, C(O)R b2 °, C(O)NR c20 R d2 °, C(O)OR a2 °, OC(O)R b2 °, OC(O)NR c20 R d2 °, NR c20 R d2 °, NR c20 C(O)R b2 °, NR c20 C(O)OR a2 °, NR c20 C(O)NR c20 R d2 °, C(=NR e20 )R b2 °, C(=NOR a20 )R b2 °, C(=NR e20 )NR c20 R d2 °, NR c20 C(=NR e20 )NR c20 R d20 , NR c20 S(O)R b2 °, NR c20 S(O) 2 R b2 °, NR c20 S(O)2NR c20 R d2 °, S(O)R b2 °, S(O)NR c20 R d2 °, S(O) 2 R b2 °, S(O) 2 NR c20 R d20 , and BR h20 R i2 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, OR a21 , SR a21 , C(O)R b21 , C(O)NR c21 R d21 , C(O)OR a21 , OC(O)R b21 , OC(O)NR c21 R d21 , NR c21 R d21 , NR c21 C(O)R b21 , NR c21 C(O)OR a21 , NR c21 C(O)NR c21 R d21 , NR c21 S(O)R b21 , NR c21 S(O) 2 R b21 , NR c21 S(O) 2 NR c21 R d21 , S(O)R b21 , S(O)NR c21 R d21 , S(O) 2 R b21 , S(O) 2 NR c21 R d21 , and BR h21 R' 21 ; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 22 ; each R 22 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, halo, D, CN, OR a22 , SR a22 , C(O)R b22 , C(O)NR c22 R d22 , C(O)OR a22 , OC(O)R b22 , OC(O)NR c22 R d22 , NR c22 R d22 , NR c22 C(O)R b22 , NR c22 C(O)OR a22 , NR c22 C(O)NR c22 R d22 , NR c22 S(O)R b22 , NR c22 S(O) 2 R b22 , NR c22 S(O) 2 NR c22 R d22 , S(O)R b22 , S(O)NR c22 R d22 , S(O) 2 R b22 , S(O) 2 NR c22 R d22 , and BR h22 R' 22 ; each R 23 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3- cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a23 , SR a23 , C(O)R b23 , C(O)NR c23 R d23 , C(O)OR a23 , OC(O)R b23 , OC(O)NR c23 R d23 , NR c23 R d23 , NR c23 C(O)R b23 , NR c23 C(O)OR a23 , NR c23 C(O)NR c23 R d23 , NR c23 S(O)R b23 , NR c23 S(O) 2 R b23 , NR c23 S(O) 2 NR c23 R d23 , S(O)R b23 , S(O)NR c23 R d23 , S(O) 2 R b23 , S(O) 2 NR c23 R d23 , and BR h23 R' 23 ; each R 30 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a3 °, SR a3 °, C(O)R b3 °, C(O)NR c30 R d3 °, C(O)OR a3 °, OC(O)R b3 °, OC(O)NR c30 R d3 °, NR^R^ 0 , NR c30 C(O)R b3 °, NR c30 C(O)OR a3 °, NR^C^NR^R^ 0 , NR c30 S(O)R b3 °, NR c30 S(O) 2 R b3 °, NR c30 S(O) 2 NR c30 R d3 °, S(O)R b3 °, S^NR^R^ 0 , S(O) 2 R b3 °, S(O) 2 NR c30 R d3 °, and BR h30 R i3 °; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, OR a31 , SR a31 , wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 32 ; each R 32 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, each R 60 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3- cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, NO 2 , OR a6 °, SR a6 °, C(O)R b6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, OC(O)R b6 °, OC(O)NR c60 R d6 °, NR^R^ 0 , NR c60 C(O)R b6 °, NR c60 C(O)OR a6 °, NR^C^NR^R^ 0 , NR c60 S(O)R b6 °, NR c60 S(O) 2 R b6 °, NR c60 S(O) 2 NR c60 R d6 °, S(O)R b6 °, S^NR^R^ 0 , S(O) 2 R b6 °, S(O) 2 NR c60 R d6 °, and BR h60 R i6 °; wherein said Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 61 ; each R 61 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a61 , SR a61 , C(O)R b61 , C(O)NR c61 R d61 , C(O)OR a61 , OC(O)R b61 , OC(O)NR c61 R d61 , NR^ R^ 1 , NR c61 C(O)R b61 , NR c61 C(O)OR a61 , NR c61 C(O)NR c61 R d61 , NR c61 S(O)R b61 , NR c61 S(O) 2 R b61 , NR c61 S(O) 2 NR c61 R d61 , S(O)R b61 , S^NR^ R^ 1 , S(O) 2 R b61 , S(O) 2 NR c61 R d61 , and BR h61 R' 61 each R 70 is independently selected from Ci-e alkyl, C 2 .6 alkenyl, C 2 .6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, NO2, OR a7 °, SR a7 °, C(O)R b70 , C(O)NR c70 R d7 °, C(O)OR a7 °, OC(O)R b7 °, OC(O)NR c70 R d7 °, NR c70 R d7 °, NR c70 C(O)R b7 °, NR c70 C(O)OR a7 °, NR c70 C(O)NR c70 R d7 °, NR c70 S(O)R b7 °, NR c70 S(O) 2 R b7 °, NR c70 S(O)2NR c70 R d7 °, S(O)R b7 °, S(O)NR c70 R d7 °, S(O) 2 R b7 °, S(O) 2 NR c70 R d70 , and BR h70 R i7 °; each R a1 , R b1 , R c1 , and R d1 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c1 and R d1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 9 ; each R h1 and R i1 is independently selected from OH, C1-6 alkoxy, and C1-6 haloalkoxy; or any R h1 and R i1 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from C1-6 alkyl and C1-6 haloalkyl; each R a2 , R b2 , R c2 and R d2 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; or any R c2 and R d2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; each R e2 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h2 and R' 2 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h2 and R' 2 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and C1.6 haloalkyl; each R a3 , R b3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; each R e3 is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h3 and R' 3 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h3 and R' 3 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a5 , R b5 , R 05 , and R d5 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 9 ; or any R c5 and R d5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 9 ; each R e5 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h5 and R' 5 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h5 and R' 5 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a6 , R b6 , R 06 , and R d6 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ; or any R c6 and R d6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 60 ; each R e6 is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h6 and R' 6 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h6 and R' 6 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a7 , R b7 , R c7 and R d7 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 70 ; or any R c7 and R d7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 70 ; each R e7 is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h7 and R' 7 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h7 and R' 7 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a1 °, R b1 °, R c1 ° and R d1 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 11 ; or any R c1 ° and R d1 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 11 ; each R e1 ° is independently selected from H, CN, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C1.6 alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h1 ° and R' 10 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h1 ° and R' 10 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a11 , R b11 , R c11 and R d11 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R c11 and R d11 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R h11 and R' 11 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h11 and R' 11 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a2 °, R b2 °, R c2 ° and R d2 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; or any R c2 ° and R d2 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R e2 ° is independently selected from H, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, Ci-e alkylthio, Ci-e alkylsulfonyl, Ci-e alkylcarbonyl, Ci-e alkylaminosulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, aminosulfonyl, Ci-e alkylaminosulfonyl and di(Ci-6 alkyl)aminosulfonyl; each R h2 ° and R' 20 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h2 ° and R' 20 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a21 , R b21 , R c21 and R d21 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 22 ; or any R c21 and R d21 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 22 ; each R h21 and R' 21 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h21 and R' 21 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a22 , R b22 , R c22 and R d22 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; each R h22 and R' 22 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h22 and R' 22 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a23 , R b23 , R c23 and R d23 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R c23 and R d23 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R h23 and R' 23 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h23 and R' 23 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a3 °, R b3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R h3 ° and R' 30 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h3 ° and R' 30 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a31 , R b31 , R 031 and R d31 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; wherein said Ci-e alkyl C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 32 ; or any R 031 and R d31 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 32 ; each R h31 and R' 31 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h31 and R' 31 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a32 , R b32 , R c32 and R d32 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl and Ci-e haloalkyl; or any R c32 and R d32 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R h32 and R' 32 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h32 and R' 32 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a6 °, R b6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 61 ; or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 61 ; each R h6 ° and R' 60 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h6 ° and R' 60 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a61 , R b61 , R 061 and R d61 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R 061 and R d61 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R h61 and R' 61 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h61 and R' 61 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; each R a7 °, R b7 °, R c7 ° and R d7 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl; or any R c7 ° and R d7 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R h7 ° and R' 70 is independently selected from OH, Ci-e alkoxy, and Ci-e haloalkoxy; or any R h7 ° and R' 70 attached to the same B atom, together with the B atom to which they are attached, form a 5- or 6-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from Ci-e alkyl and Ci-e haloalkyl; and each R 9 is independently selected from D, OH, NO2, ON, halo, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, C3-6 cycloalkyl-Ci.2 alkylene, Ci-e alkoxy, Ci-e haloalkoxy, Ci-3 alkoxy-Ci-3 alkyl, Ci-3 alkoxy-Ci-3 alkoxy, HO-C1.3 alkoxy, HO-C1.3 alkyl, cyano-Ci-3 alkyl, H2N-C1.3 alkyl, amino, Ci-e alkylamino, di(Ci-6 alkyl)amino, thio, Ci-e alkylthio, Ci-6 alkylsulfinyl, Ci-e alkylsulfonyl, carbamyl, Ci-e alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci-e alkylcarbonyl, Ci-e alkoxycarbonyl, Ci-e alkylcarbonylamino, Ci-e alkoxycarbonylamino, Ci-e alkylcarbonyloxy, aminocarbonyloxy, Ci-e alkylaminocarbonyloxy, di(Ci-6 alkyl)aminocarbonyloxy, Ci-e alkylsulfonylamino, aminosulfonyl, Ci-e alkylaminosulfonyl, di(Ci-6 alkyl)aminosulfonyl, aminosulfonylamino, Ci-e alkylaminosulfonylamino, di(Ci-6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci-e alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino.

In another embodiment, the compound of Formula I is a compound of Formula II: or a pharmaceutically acceptable salt thereof, wherein:

-AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, OR a1 , SR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , OC(O)R b1 , OC(O)NR c1 R d1 , NR c1 R d1 , NR c1 C(O)R b1 , NR c1 C(O)OR a1 , NR c1 C(O)NR c1 R d1 , NR c1 S(O) 2 R b1 , NR c1 S(O) 2 NR c1 R d1 , S(O) 2 R b1 , and S(O) 2 NR c1 R d1 ;

R 2 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, OR a2 , SR a2 , C(O)R b2 , C(O)NR c2 R d2 , C(O)OR a2 , OC(O)R b2 , OC(O)NR c2 R d2 , NR c2 R d2 , NR c2 C(O)R b2 , NR c2 C(O)OR a2 , NR c2 C(O)NR c2 R d2 , NR c2 S(O) 2 R b2 , NR c2 S(O) 2 NR c2 R d2 , S(O) 2 R b2 , and S(O) 2 NR c2 R d2 ; wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ;

Cy 1 is selected from C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 10 ; R 4 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a3 , SR a3 , C(O)R b3 , (O)OR a3 , wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce- aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci-3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ;

R 5 is selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a5 , SR a5 , C(O)R b5 ,

R 6 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a6 , SR a6 , C(O)R b6 , (O)OR a6 , wherein said C1.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce- w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-s alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)OR a7 , NR c7 C(O)NR c7 R d7 , NR c7 S(O) 2 R b7 , NR c7 S(O) 2 NR c7 R d7 , S(O) 2 R b7 , and S(O) 2 NR c7 R d7 ;

Cy 2 is selected from C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 5-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring- forming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 5-10 membered heteroaryl and 4-

10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a1 °, SR a1 °, C(O)R b1 °, C(O)NR c10 R d1 °, C(O)OR a1 °, OC(O)R b1 °, OC(O)NR c10 R d1 °, NR c10 R d1 °, NR c10 C(O)R b1 °, NR c10 C(O)OR a1 °, NR c10 C(O)NR c10 R d1 °, NR c10 S(O) 2 R b1 °, NR c10 S(O)2NR c10 R d1 °, S(O) 2 R b1 °, and S(O) 2 NR c10 R d10 ; each R 20 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs- cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a2 °, SR a2 °,

C(O)R b2 °, C(O)NR c20 R d2 °, C(O)OR a2 °, OC(O)R b2 °, OC(O)NR c20 R d2 °, NR c20 R d2 °,

NR c20 C(O)R b20 , NR c20 C(O)OR a2 ° , NR c20 C(O)NR c2 ° R d2 ° , NR c20 S(O) 2 R b2 °, NR c20 S(O)2NR c20 R d2 °,

S(O)2R b2 °, S(O)2NR c20 R d2 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl,

4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3- cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci-3 alkylene, Ce-w aryl-Ci-3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a21 , SR a21 , C(O)R b21 , C(O)NR c21 R d21 , C(O)OR a21 , OC(O)R b21 , OC(O)NR c21 R d21 , NR c21 R d21 , NR c21 C(O)R b21 , NR c21 C(O)OR a21 , NR c21 C(O)NR c21 R d21 , NR c21 S(O) 2 R b21 , NR c21 S(O) 2 NR c21 R d21 , S(O) 2 R b21 , and S(O) 2 NR c21 R d21 ; each R 23 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, Cs-w cycloalkyl-Ci-s alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a23 , SR a23 , C(O)R b23 , C(O)NR c23 R d23 , C(O)OR a23 , OC(O)R b23 , OC(O)NR c23 R d23 , NR c23 R d23 , NR c23 C(O)R b23 , NR c23 C(O)OR a23 , NR c23 C(O)NR c23 R d23 , NR c23 S(O) 2 R b23 , NR c23 S(O) 2 NR c23 R d23 , S(O) 2 R b23 , and S(O) 2 NR c23 R d23 ; each R 30 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a3 °, SR a3 °, S(O)2R b3 °, and S(O)2NR c30 R d3 °; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-W cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene and 5-10 membered heteroaryl-Ci.3 alkylene are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci.3 alkylene, 5-10 membered heteroaryl-Ci.3 alkylene, halo, D, CN, OR a31 , SR a31 , each R 60 is independently selected from Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, 5-10 membered heteroaryl, C3-w cycloalkyl-Ci-3 alkylene, 4-10 membered heterocycloalkyl-Ci.3 alkylene, Ce-w aryl-Ci-3 alkylene, 5-10 membered heteroaryl-Ci-3 alkylene, halo, D, CN, OR a6 °, SR a6 °, each R a1 , R b1 , R c1 , and R d1 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-

10 membered heteroaryl; or any R c1 and R d1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a2 , R b2 , R c2 and R d2 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; or any R c2 and R d2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; each R a3 , R b3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; each R a5 , R b5 , R 05 , and R d5 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5- 10 membered heteroaryl; or any R c5 and R d5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a6 , R b6 , R 06 , and R d6 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4- 10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ; or any R c6 and R d6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2 or 3 substituents independently selected from R 60 ; each R a7 , R b7 , R c7 and R d7 is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5- 10 membered heteroaryl; or any R c7 and R d7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a1 °, R b1 °, R c1 ° and R d1 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; or any R c1 ° and R d1 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a2 °, R b2 °, R c2 ° and R d2 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-W cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; or any R c20 and R d20 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R a21 , R b21 , R c21 and R d21 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R c21 and R d21 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a23 , R b23 , R c23 and R d23 , is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R c23 and R d23 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a3 °, R b3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; wherein said Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 5-10 membered heteroaryl, are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R a31 , R b31 , R 031 and R d31 , is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 4-7 membered heterocycloalkyl; or any R 031 and R d31 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; each R a6 °, R b6 °, R c6 ° and R d6 ° is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 5-10 membered heteroaryl; and or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.

In another embodiment of Formula II, or a pharmaceutically acceptable salt thereof, -AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ; R 1 is H;

R 2 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, CN, OR a2 , and NR c2 R d2 ; wherein said Ci-6 alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 23 ;

Cy 1 is selected from Ce- aryl and 6-10 membered heteroaryl; wherein the 6-10 membered heteroaryl has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 6-10 membered heteroaryl is optionally substituted by oxo to form a carbonyl group; and wherein the Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2 or 3 substituents independently selected from R 10 ;

R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, Cs- cycloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, CN, OR a3 , C(O)NR c3 R d3 and NR^R' 13 ; wherein said Ci-e alkyl, Cs-w cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ;

R 5 is H;

R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, CN, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, OR a6 , and NR c6 R d6 ; wherein said Ci-e alkyl, 4- 10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 60 ;

R 7 is halo;

Cy 2 is 4-7 membered heterocycloalkyl; wherein the 4-7 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 4-7 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, CN, and OH; each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, CN, OR a2 °, C(O)R b2 °, C(O)NR c20 R d20 and NR c20 R d2 °; each R 23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, CN, OR a23 , and NR c23 R d23 ; each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, Cs-w cycloalkyl, 4- 10 membered heterocycloalkyl, halo, CN, OR a3 °, and NR c30 R d3 °; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, CN, OR a31 , and NR^R' 131 ; each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, CN, OR a6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °; each R a2 , R c2 and R d2 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 23 ; each R a3 , R b3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl; wherein said C1.6 alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ; each R a6 , R 06 , and R d6 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 ; each R a2 °, R b2 °, R c2 ° and R d2 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R a23 , R c23 and R d23 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R a31 , R 031 and R d31 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; and each R a6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl.

In yet another embodiment of Formula II, or a pharmaceutically acceptable salt thereof,

-AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is selected from H, D, Ci-e alkyl, and Ci-e haloalkyl;

R 2 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a2 , and NR c2 R d2 ; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 23 ; Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4-10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2 or 3 substituents independently selected from R 10 ;

R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, D, CN, OR a3 , C(O)NR c3 R d3 and NR c3 R d3 ; wherein said C1-6 alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ;

R 5 is selected from H, C1-6 alkyl, C1-6 haloalkyl, halo, D, CN, OR a5 , and NR c5 R d5 ;

R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a6 , and NR^R' 16 ; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, CN, OR a7 , and NR c7 R d7 ;

Cy 2 is selected from 4-7 membered heterocycloalkyl; wherein the 4-7 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 4-7 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a1 °, C(O)NR c10 R d1 °, and NR c10 R d1 °; each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a2 °, C(O)R b2 °, C(O)NR c20 R d20 and NR c20 R d2 °; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a21 , and NR c21 R d21 ; each R 23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a23 , and NR c23 R d23 ; each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, D, CN, OR a3 °, and NR c30 R d3 °; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a31 , and NR c31 R d31 ; each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °; each R a2 , R c2 and R d2 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 23 ; each R a3 , R b3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl; wherein said C1.6 alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 30 ; each R a5 , R 05 , and R d5 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a6 , R 06 , and R d6 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 60 ; each R a7 , R c7 and R d7 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a1 °, R b1 °, R c1 ° and R d1 ° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a2 °, R b2 °, R c2 ° and R d2 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R a21 , R c21 and R d21 , is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a23 , R c23 and R d23 , is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R a31 , R 031 and R d31 , is independently selected from H, Ci-e alkyl, Ci-e haloalkyl; each R a6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; and or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group.

In still another embodiment of Formula II, or a pharmaceutically acceptable salt thereof,

-AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is H;

R 2 is selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 23 ;

Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 10 ;

R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, CN, OR a3 , and NR c3 R d3 ; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ;

R 5 is selected from H, Ci-e alkyl, and Ci-e haloalkyl;

R 6 is selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, and CN;

Cy 2 is selected from 4-6 membered heterocycloalkyl; wherein the 4-6 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 or 2 ring-forming heteroatoms independently selected from N, and O; and wherein the 4-6 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and OR a1 °; each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and C(O)R b2 °; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R 23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a3 °, and NR c30 R d3 °; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °; each R a3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 30 ; each R a1 ° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R b2 ° is independently selected from C2-6 alkenyl, and C2-6 alkynyl; wherein said C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R a3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R a6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; and or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group.

In another embodiment of Formula II, or a pharmaceutically acceptable salt thereof, -AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is H;

R 2 is C1.6 alkyl optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 23 ; Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 10 ;

R 3 is absent;

R 4 is selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, and OR a3 ; wherein said C1-6 alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ;

R 5 is H;

R 6 is C1-6 alkyl optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 60 ;

R 7 is selected from H, C1-6 alkyl, and halo;

Cy 2 is 4-14 membered heterocycloalkyl optionally substituted with 1 , 2, 3 or 4 substituents independently selected from R 20 ; each R 10 is independently selected from C1-6 alkyl, halo, CN, and OR a1 °; each R 20 is independently selected from C1-6 alkyl and C(O)R b2 °; wherein said C1-6 alkyl is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R 21 is independently selected from halo and CN; each R 23 is CN; each R 30 is independently selected from 4-10 membered heterocycloalkyl and NR c30 R d3 °; wherein said 4-10 membered heterocycloalkyl is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl and halo; each R 60 is independently selected from 4-10 membered heterocycloalkyl, C(O)NR c60 R d6 °, and C(O)OR a6 °; each R a3 is Ci-e alkyl optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 30 ; each R a1 ° is H; each R b2 ° is C2-6 alkenyl optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 21 ; each R c3 ° and R d3 ° is Ci-e alkyl optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R 31 ; and each R a6 °, R c6 ° and R d60 is Ci-e alkyl.

In another embodiment of Formula II, or a pharmaceutically acceptable salt thereof, -AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is N or CR 5 ;

Y is N or CR 6 ;

R 1 is H;

R 2 is Ci-6 alkyl optionally substituted with 1 , 2, or 3 substituents independently selected from R 23 ;

Cy 1 is selected from napthyl, benzothiazolyl, chromyl, phenyl, indolyl, indolinyl, tetrahydroquinolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally substituted with 1 , 2, or 3 substituents independently selected from R 10 ;

R 4 is selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, 6-10 membered heteroaryl, and OR a3 ; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl, and 6-10 membered heteroaryl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 30 ;

R 5 is H;

R 6 is Ci-6 alkyl, 4-10 membered heterocycloalkyl, dihydroindolizinonyl, wherein Ci-e alkyl and 4-10 membered heterocycloalkyl are optionally substituted with 1 , 2, or 3 substituents independently selected from R 60 ;

R 7 is C1.3 alkyl or halo;

Cy 2 is 4-14 membered heterocycloalkyl optionally substituted with 1 , 2, or 3 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, halo, CN, and OH; each R 20 is independently selected from Ci-e alkyl and C(O)R b2 °; wherein said Ci-e alkyl is optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R 21 is independently selected from halo and CN; each R 23 is CN; each R 30 is independently selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, C(O)NR c30 R d3 °, and NR^R' 130 ; wherein said 4-10 membered heterocycloalkyl is optionally substituted with 1 , 2, or 3 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl and halo; each R 60 is independently selected from 4-10 membered heterocycloalkyl, pyridinonyl, C(O)NR c60 R d6 °, C(O)R a6 °, and C(O)OR a6 °; each R a3 is Ci-e alkyl optionally substituted with 1 , 2, or 3 substituents independently selected from R 30 ; each R b2 ° is C2-6 alkenyl optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R c3 ° and R d3 ° is independently H or Ci-e alkyl; and each R a6 °, R c6 ° and R d6 ° is independently H or Ci-e alkyl.

In another embodiment of Formula I, or a pharmaceutically acceptable salt thereof, -AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is selected from H, D, Ci-e alkyl, and Ci-e haloalkyl;

R 2 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a2 , and NR c2 R d2 ; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 23 ;

Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4-10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 , 2 or 3 substituents independently selected from R 10 ;

R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, D, CN, OR a3 , C(O)NR c3 R d3 and NR c3 R d3 ; wherein said Ci-e alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ;

R 5 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a5 , and NR^R' 15 ;

R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 , and NR c6 R d6 ; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, CN, OR a7 , and NR c7 R d7 ;

Cy 2 is selected from 4-7 membered heterocycloalkyl; wherein the 4-7 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; wherein a ring-forming carbon atom of 4-7 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a1 °, C(O)NR c10 R d1 °, and NR c10 R d1 °; each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a20 , C(O)R b20 , C(O)NR c20 R d20 and NR c20 R d2 °; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a21 , and NR c21 R d21 ; each R 23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a23 , and NR c23 R d23 ; each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, D, CN, OR a3 °, C(O)NR c30 R d3 °, and NR c30 R d3 °; wherein said C1.6 alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a31 , and NR^R^ 1 ; each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)R b6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °; each R a2 , R c2 and R d2 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 23 ; each R a3 , R b3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl; wherein said C1.6 alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 30 ; each R a5 , R 05 , and R d5 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a6 , R 06 , and R d6 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 ; each R a7 , R c7 and R d7 is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a1 °, R b1 °, R c1 ° and R d1 ° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a2 °, R b2 °, R c2 ° and R d2 ° is independently selected from H, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, and Ci-e haloalkyl; wherein said Ci-e alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R a21 , R c21 and R d21 , is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a23 , R c23 and R d23 , is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R a3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R a31 , R 031 and R d31 , is independently selected from H, Ci-e alkyl, Ci-e haloalkyl; each R a6 °, R b6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl; and or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group.

In yet another embodiment of Formula I, or a pharmaceutically acceptable salt thereof,

-AR 1 =BR 2 - is selected from -N=CR 2 -, and -CR 1 =N-;

X is selected from N and CR 5 ;

Y is selected from N and CR 6 ;

R 1 is H;

R 2 is selected from H, Ci-e alkyl, and Ci-e haloalkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 23 ;

Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl, and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 10 ;

R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, CN, OR a3 , and NR c3 R d3 ; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 ;

R 5 is selected from H, Ci-e alkyl, and Ci-e haloalkyl; R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 ;

R 7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, and CN;

Cy 2 is selected from 4-6 membered heterocycloalkyl; wherein the 4-6 membered heterocycloalkyl has at least one ring-forming carbon atom and 1 or 2 ring-forming heteroatoms independently selected from N, and O; and wherein the 4-6 membered heterocycloalkyl, is optionally substituted with 1 or 2 substituents independently selected from R 20 ; each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and OR a1 °; each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and C(O)R b20 ; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 21 ; each R 21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R 23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a3 °, C(O)NR c30 R d3 °, and NR c30 R d3 °; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R 31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, and CN; each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)R b6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °; each R a3 , R c3 and R d3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 30 ; or any R c3 and R d3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 30 ; each R a1 ° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl; each R b2 ° is independently selected from C2-6 alkenyl, and C2-6 alkynyl; wherein said C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 ; each R a3 °, R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 31 ; or any R c3 ° and R d3 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group optionally substituted with 1 or 2 substituents independently selected from R 31 ; each R a6 °, R b6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; and or any R c6 ° and R d6 ° attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, or 6-membered heterocycloalkyl group.

In an embodiment, the compound of Formula I is a compound of Formula III: or a pharmaceutically acceptable salt thereof.

In an embodiment of Formula III, or a pharmaceutically acceptable salt thereof,

Cy 1 is selected from naphthyl, indolyl, tetrahydroquinolinyl, indolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally subsituted with 1 or 2 substituents independently selected from OH, halo, CN, and Ci-e alkyl;

R 6 is Ci-6 alkyl optionally substituted with 1 or 2 substituents independently selected from oxazolidin-2-one, CO2C1.6 alkyl, and CON(CI-6 alkyl^;

R 7 is selected from H and halo;

Cy 2 is selected from 2-azabicyclo[2.1.1]hexane and piperidine, both of which are optionally subsituted with 1 or 2 substituents independently selected from CO(C2-6 haloalkenyl) and Ci-e alkyl-CN; and

R a3 is C1.6 alkyl optionally substituted with pyrrolidine or hexahydropyrrolizine, wherein said pyrrolidine is optionally substituted with Ci-e alkyl and hexahydropyrrolizine is optionally subsituted with halo.

In another embodiment of Formula III, or a pharmaceutically acceptable salt thereof,

Cy 1 is selected from naphthyl, phenyl, benzothiazolyl, chromanyl, indolyl, tetrahydroquinolinyl, indolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally subsituted with 1 or 2 substituents independently selected from OH, halo, CN, and Ci-e alkyl; R 6 is Ci-6 alkyl or dihydroindolizinone, wherein said alkyl is optionally substituted with 1 or 2 substituents independently selected from oxazolidin-2-one, pyridin-2-one, CO2C1.6 alkyl, COC1.6 alkyl, and CON(CI-6 alkyl^;

R 7 is selected from H and halo;

Cy 2 is selected from 2-azabicyclo[2.1.1]hexane and piperidine, both of which are optionally subsituted with 1 or 2 substituents independently selected from CO(C2-6 haloalkenyl) and C1.6 alkyl-CN; and

R a3 is C1.6 alkyl optionally substituted with pyrrolidine or hexahydropyrrolizine, wherein said pyrrolidine is optionally substituted with C1.6 alkyl and hexahydropyrrolizine is optionally substituted with halo.

In another embodiment, the compound of Formula I is a compound of Formula IV: or a pharmaceutically acceptable salt thereof.

In an embodiment of Formula IV, or a pharmaceutically acceptable salt thereof,

Cy 1 is selected from naphthyl, indolyl, tetrahydroquinolinyl, indolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally subsituted with 1 or 2 substituents independently selected from OH, halo, CN, and C1.6 alkyl;

R 6 is C1.6 alkyl optionally substituted with 1 or 2 substituents independently selected from oxazolidin-2-one, CO2C1.6 alkyl, and CON(CI-6 alkyl^;

R 7 is selected from H and halo;

Cy 2 is selected from 2-azabicyclo[2.1.1]hexane and piperidine, both of which are optionally subsituted with 1 or 2 substituents independently selected from CO(C2-6 haloalkenyl) and C1.6 alkyl-CN; and

R a3 is C1.6 alkyl optionally substituted with pyrrolidine, hexahydropyrrolizine, wherein said pyrrolidine is optionally substituted with C1.6 alkyl and hexahydropyrrolizine is optionally subsituted with halo.

In yet another embodiment, the compound of Formula I is a compound of Formula V: or a pharmaceutically acceptable salt thereof.

In an embodiment of Formula V, or a pharmaceutically acceptable salt thereof,

Cy 1 is selected from naphthyl, indolyl, tetrahydroquinolinyl, indolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally subsituted with 1 or 2 substituents independently selected from OH, halo, CN, and Ci-e alkyl;

R 4 is azetidine optionally subsituted with 1 or 2 substituents independently selected from N(CI-6 alkyl)2;

R 7 is selected from H and halo; and

Cy 2 is selected from 2-azabicyclo[2.1.1]hexane and piperidine, both of which are optionally subsituted with 1 or 2 substituents independently selected from CO(C2-6 haloalkenyl) and Ci-e alkyl-CN.

In still another embodiment, the compound of Formula I is a compound of Formula or a pharmaceutically acceptable salt thereof.

In an embodiment of Formula VI, or a pharmaceutically acceptable salt thereof,

Cy 1 is selected from naphthyl, indolyl, tetrahydroquinolinyl, indolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally subsituted with 1 or 2 substituents independently selected from OH, halo, CN, and Ci-e alkyl;

R 4 is azetidine optionally subsituted with 1 or 2 substituents independently selected from N(CI-6 alkyl)2;

R 6 is Ci-6 alkyl optionally substituted with 1 or 2 substituents independently selected from oxazolidin-2-one, CO2C1.6 alkyl, and CON(CI-6 alkyl^;

R 7 is selected from H and halo; and Cy 2 is selected from 2-azabicyclo[2.1.1]hexane and piperidine, both of which are optionally subsituted with 1 or 2 substituents independently selected from CO(C2-6 haloalkenyl) and Ci-e alkyl-CN.

In another embodiment, the compound of Formula I is a compound of Formula VII: or a pharmaceutically acceptable salt thereof.

In an embodiment of Formula VI, or a pharmaceutically acceptable salt thereof,

R 2 is Ci-6 alkyl;

Cy 1 is selected from naphthyl, indolyl, tetrahydroquinolinyl, indolinyl, dihydroindenyl, quinolinyl, and indazolyl, all of which are optionally subsituted with 1 or 2 substituents independently selected from OH, halo, CN, and Ci-e alkyl;

R 4 is azetidine optionally subsituted with 1 or 2 substituents independently selected from N(CI-6 alkyl)2;

R 6 is Ci-6 alkyl optionally substituted with 1 or 2 substituents independently selected from oxazolidin-2-one, CO2C1.6 alkyl, and CON(CI-6 alkyl^;

R 7 is selected from H, halo, and Ci-e alkyl; and

Cy 2 is selected from 2-azabicyclo[2.1.1]hexane and piperidine, both of which are optionally subsituted with 1 or 2 substituents independently selected from CO(C2-6 haloalkenyl) and Ci-e alkyl-CN.

In yet another embodiment,

(a) Cy 1 is other than 3,5-dimethylisoxazol-4-yl; and

(b) R 3 is other than CH2CF3.

In still another embodiment, — represents a double bond. In another embodiment, — represents a single bond.

In an embodiment, -AR 1 =BR 2 - is -N=CR 2 -. In another embodiment, -AR 1 =BR 2 - is - CR 1 =N-

In yet another embodiment, X is CR 5 ; and Y is CR 6 . In still another embodiment, X is N; and Y is CR 6 . In an embodiment, X is N; and Y is N. In another embodiment, X is CR 5 . In yet another embodiment, X is N. In an embodiment, Y is CR 6 . In another embodiment, Y is N. In an embodiment, R 1 is selected from H, D, Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C1.6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce-w aryl, 5-10 membered heteroaryl, halo, CN, OR a1 , SR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , OC(O)R b1 , OC(O)NR c1 R d1 , NR c1 R d1 , NR c1 C(O)R b1 , NR c1 C(O)OR a1 , NR c1 C(O)NR c1 R d1 , NR c1 S(O) 2 R b1 , NR c1 S(O) 2 NR c1 R d1 , S(O) 2 R b1 , and S(O) 2 NR c1 R d1 . In an embodiment, R 1 is selected from H, D, C1.6 alkyl, and Ci-e haloalkyl. In another embodiment, R 1 is H.

In an embodiment, R 2 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a2 , and NR c2 R d2 ; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 23 . In an embodiment, R 2 is selected from H, Ci-e alkyl, and Ci- 6 haloalkyl; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 23 . In yet another embodiment, R 2 is selected from H, Ci-e alkyl, and Ci-e haloalkyl. In still another embodiment, R 2 is Ci-e alkyl optionally substituted with CN.

In an embodiment, Cy 1 is selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl; wherein the 4-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 4-10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, Ce- aryl and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 10 .

In another embodiment, Cy 1 is selected from 5-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl; wherein the 5-10 membered heterocycloalkyl and 6-10 membered heteroaryl each has at least one ring-forming carbon atom and 1 , 2, 3, or 4 ringforming heteroatoms independently selected from N, O, and S; wherein the N and S are optionally oxidized; wherein a ring-forming carbon atom of 6-10 membered heteroaryl and 5- 10 membered heterocycloalkyl is optionally substituted by oxo to form a carbonyl group; and wherein the 5-10 membered heterocycloalkyl, Ce-w aryl and 6-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 10 .

In an embodiment, R 3 N— CR 4 is a single bond, and R 4 is selected from =0 and =S. In yet another embodiment, R 3 N— CR 4 is a double bond, and R 3 is absent.

In an embodiment, R 3 is selected from H, Ci-e alkyl, C3-W cycloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, C3-W cycloalkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 30 . In an embodiment, R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, D, CN, OR a3 , C(O)NR c3 R d3 and NR^R' 13 ; wherein said Ci-e alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl, are each optionally substituted with 1 or 2 substituents independently selected from R 30 . In an embodiment, R 4 is selected from H, Ci-e alkyl, C1.6 haloalkyl, 4-10 membered heterocycloalkyl, halo, CN, OR a3 , and NR^R' 13 ; wherein said C1.6 alkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 30 . In still another embodiment, R 4 is selected from C1.6 alkyl, 4-10 membered heterocycloalkyl, and OR a3 ; wherein said 4-10 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 30 .

In an embodiment, R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4- 10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, D, CN, OR a3 , C(O)NR c3 R d3 and NR^R' 13 ; wherein said Ci-e alkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 . In an embodiment, R 4 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, CN, OR a3 , and NR c3 R d3 ; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 . In an embodiment, R 4 is selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, and OR a3 ; wherein said Ci-e alkyl, 4-10 membered heterocycloalkyl, and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 substituents independently selected from R 30 .

In an embodiment, R 5 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a5 , and NR^R' 15 . In an embodiment, R 5 is selected from H, Ci-e alkyl, and Ci-e haloalkyl. In an embodiment, R 5 is H.

In an embodiment, R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a6 , and NR^R' 16 ; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 60 . In an embodiment, R 6 is selected from H, Ci-e alkyl, and Ci- 6 haloalkyl; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 60 . In another embodiment, R 6 is Ci-e alkyl optionally substituted with 1 or 2 substituents independently selected from R 60 .

In an embodiment, R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 , and NR^R' 16 ; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 60 . In an embodiment, R 6 is selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 . In an embodiment, R 6 is selected from Ci-e alkyl and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 60 .

In an embodiment, R 7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, CN, OR a7 , and NR c7 R d7 . In an embodiment, R 7 is selected from H, Ci-e alkyl, and Ci-e haloalkyl, halo, D, and CN. In yet another embodiment, R 7 is selected from H, C1.3 alkyl, and halo.

In an embodiment, Cy 2 is selected from 4-10 membered heterocycloalkyl; wherein the 4-10 membered heterocycloalkyl, is optionally substituted with 1 , 2, or 3 substituents independently selected from R 20 . In an embodiment, Cy 2 is selected from 4-7 membered heterocycloalkyl; wherein the 4-7 membered heterocycloalkyl, is optionally substituted with 1 or 2 substituents independently selected from R 20 . In an embodiment, Cy 2 is selected from 4- 6 membered heterocycloalkyl; wherein the 4-6 membered heterocycloalkyl, is optionally substituted with 1 or 2 substituents independently selected from R 20 . In still another embodiment, Cy 2 is 5-10 membered heterocycloalkyl optionally substituted with 1 or 2 substituents independently selected from R 20 .

In yet another embodiment, Cy 2 is selected from wherein n is 0, 1 or 2.

In an embodiment, Cy 2 is Cy 2 -a. In another embodiment, Cy 2 is Cy 2 -b. In yet another embodiment, Cy 2 is Cy 2 -c. In still another embodiment, Cy 2 is Cy 2 -d. In an embodiment, Cy 2 is Cy 2 -e. In an embodiment, Cy 2 is selected from Cy 2 -a and Cy 2 -e.

In an embodiment, n is 0. In another embodiment, n is 1. In yet another embodiment, n is 2.

In an embodiment, Cy 2 is Cy 2 -e and n is 0.

In an embodiment, each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a1 °, C(O)NR c10 R d1 °, and NR c10 R d1 °. In an embodiment, each R 10 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and OR a1 °. In an embodiment, each R 10 is independently selected from Ci-e alkyl, halo, CN, and OH.

In an embodiment, each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a20 , C(O)R b20 , C(O)NR c20 R d20 and NR c20 R d2 °; wherein said Ci- 6 alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 21 . In an embodiment, each R 20 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, and C(O)R b2 °; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 21 . In an embodiment, each R 20 is independently selected from Ci-6 alkyl and C(O)R b2 °; wherein said Ci-e alkyl, is optionally substituted with 1 or 2 substituents independently selected from R 21 . In another embodiment, each R 20 is independently selected from Ci-e alkyl and C(O)C2-6 alkenyl; wherein said Ci-e alkyl and C2-6 alkenyl are each optionally substituted with 1 or 2 substituents independently selected from CN and halo.

In an embodiment, each R 21 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a21 , and NR c21 R d21 . In an embodiment, each R 21 is independently selected from C1.6 alkyl, Ci-e haloalkyl, halo, D, and CN. In an embodiment, each R 21 is independently selected from halo and CN.

In an embodiment, each R 23 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a23 , and NR c23 R d23 . In an embodiment, each R 23 is independently selected from C1.6 alkyl, Ci-e haloalkyl, halo, D, and CN. In an embodiment, each R 23 is CN.

In an embodiment, each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a3 °, and NR c30 R d3 °; wherein said C1.6 alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 . In an embodiment, each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a3 °, and NR^R' 130 ; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 . In an embodiment, each R 30 is independently selected from 4-10 membered heterocycloalkyl and NR^R' 130 ; wherein said 4-10 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from R 31 .

In an embodiment, each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a3 °, C(O)NR c30 R d3 °, and NR c30 R d3 °; wherein said Ci-e alkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 31 . In an embodiment, each R 30 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a3 °, C(O)NR c30 R d3 °, and NR^R' 130 ; wherein said C1.6 alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 . In an embodiment, each R 30 is independently selected from Ci-e alkyl, 4-10 membered heterocycloalkyl, C(O)NR c30 R d3 °, and NR c30 R d3 °; wherein said Ci-e alkyl and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 31 . In an embodiment, each R 31 is independently selected from Ci-e alkyl, Ci-e haloalkyl, halo, D, CN, OR a31 , and NR c31 R d31 . In an embodiment, each R 31 is independently selected from Ci-6 alkyl, Ci-e haloalkyl, halo, D, and CN. In another embodiment, each R 31 is independently selected from Ci-e alkyl and halo.

In an embodiment, each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °. In another embodiment, each R 60 is independently selected from C1.6 alkyl, C1.6 haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °. In yet another embodiment, each R 60 is independently selected from 4-10 membered heterocycloalkyl, C(O)NR c60 R d6 °, and C(O)OR a6 °. In still another embodiment, each R 60 is independently selected from 4-10 membered heterocycloalkyl, C(O)N(CI-6 alkyl)2, and C(O)OCi-6 alkyl.

In an embodiment, each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)R b6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °. In an embodiment, each R 60 is independently selected from Ci-e alkyl, Ci-e haloalkyl, 4-10 membered heterocycloalkyl, halo, D, CN, OR a6 °, C(O)R b6 °, C(O)NR c60 R d6 °, C(O)OR a6 °, and NR c60 R d6 °. In an embodiment, each R 60 is independently selected from 4-10 membered heterocycloalkyl, C(O)R b6 °, C^NR^R^ 0 , and C(O)OR a6 °.

In an embodiment, each R a3 is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl are each optionally substituted with 1 or 2 substituents independently selected from R 30 . In still another embodiment, each R a3 is Ci-e alkyl optionally substituted with 1or 2 substituents independently selected from R 30 ; and each R 30 is 4-10 membered heterocycloalkyl optionally substituted with 1 or 2 substituents independently selected from Ci-e alkyl.

In an embodiment, each R a1 ° is independently selected from H, Ci-e alkyl, and Ci-e haloalkyl. In an embodiment, each R a1 ° is H.

In an embodiment, each R b2 ° is independently selected from C2-6 alkenyl, and C2-6 alkynyl; wherein said C2-6 alkenyl, and C2-6 alkynyl, are each optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 . In an embodiment, each R b2 ° is C2-6 alkenyl; wherein said C2-6 alkenyl is optionally substituted with 1 , 2, or 3 substituents independently selected from R 21 .

In an embodiment, each R c3 ° and R d3 ° is independently selected from H, Ci-e alkyl, C1.6 haloalkyl, and 4-10 membered heterocycloalkyl; wherein said Ci-e alkyl, and 4-10 membered heterocycloalkyl, are each optionally substituted with 1 or 2 substituents independently selected from R 31 . In an embodiment, each R c3 ° and R d3 ° is Ci-e alkyl optionally substituted with 1 or 2 substituents independently selected from R 31 .

In an embodiment, each R c3 ° and R d3 ° is independently selected from H and Ci-e alkyl; wherein said Ci-e alkyl is optionally substituted with 1 or 2 substituents independently selected from R 31 .

In an embodiment, each R a6 °, R c6 ° and R d6 ° is independently selected from H, Ci-e alkyl, Ci-e haloalkyl, and 4-10 membered heterocycloalkyl. In another embodiment, each R a6 °, ReSt) anc | R d60 | S Q 1 6 gl yl

In still another embodiment, the compound of Formula I is selected from methyl 3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3-h ydroxynaphthalen- 1-yl)-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)propanoate; methyl 3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-4-(((2 R,7aS)-2- fluorotetrahydro-1 H-pyrrolizin-7a(5H)-yl)methoxy)-7-(3-hydroxynaphthalen-1-yl) -1 H- pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)propanoate;

4-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-m ethyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-7-yl)naphthalen-2-ol;

4-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-m ethyl-4-((tetrahydro-1 H- pyrrolizin-7a(5H)-yl)methoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-7-yl)naphthalen-2-ol;

3-((1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7- (3-hydroxynaphthalen-1-yl)-4- methyl-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)methyl)oxazolidin-2-one;

3-((1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7- (5-fluoro-1 H-indol-3-yl)-4-((S)- 1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)methyl)oxazolidin-2-one;

3-((1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(3,4-dihy droquinolin-1 (2H)-yl)-6-fluoro- 4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-2- yl)methyl)oxazolidin-2-one;

1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(ind olin-1-yl)-2-methyl-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridine;

3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-( 3-hydroxynaphthalen-1-yl)-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-2-yl)-N,N- dimethylpropanamide;

3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(3-cyano-2 ,3-dihydro-1 H-inden-4-yl)-4- (3-(dimethylamino)azetidin-1-yl)-6-fluoro-1 H-imidazo[4,5-c][1 ,6]naphthyridin-2-yl)-N,N- dimethylpropanamide; methyl 3-(1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-4-(3-(dimethylam ino)azetidin-1- yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-2- yl)propanoate;

2-((2S,4S)-4-(4-(3-(ethyl(methyl)amino)azetidin-1-yl)-6-f luoro-7-(5-fluoroquinolin-8- yl)-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)-1-((E)-4-fluorobut-2-enoyl)piperidin-2 - yl)acetonitrile;

2-((2S,4S)-4-(7-(5,6-dimethyl-1 H-indazol-3-yl)-4-(3-(ethyl(methyl)amino)azetidin-1- yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)-1-(2-fluoroacryloyl)piperidin-2- yl)acetonitrile;

8-(1-((2S,4S)-2-(cyanomethyl)-1-(2-fluoroacryloyl)piperid in-4-yl)-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-7-yl)-1- naphthonitrile;

8-(1-((2S,4S)-2-(cyanomethyl)-1-((E)-4-fluorobut-2-enoyl) piperidin-4-yl)-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-7-yl)-1- naphthonitrile;

3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-4-(3-(dimeth ylamino)azetidin-1-yl)-7-(3- hydroxynaphthalen-1-yl)-6,8-dimethyl-1/7-imidazo[4,5-c][1 ,5]naphthyridin-2-yl)-N,N- dimethylpropanamide; and

3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-8-(2-cyanoet hyl)-4-(3- (dimethylamino)azetidin-1-yl)-7-(3-hydroxynaphthalen-1-yl)-1 /7-imidazo[4,5- c][1 ,5]naphthyridin-2-yl)-N,N-dimethylpropanamide; or a pharmaceutically acceptable salt thereof.

In another embodiment, the compound of Formula I is selected from:

4-(1-((7/?,4/?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-6-fluor o-2-methyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-7-yl)-6-fluoronaphthalen-2-ol;

1-((/?)-2-(7-(Benzo[d]thiazol-4-yl)-1-((7/?,4/?,5S)-2-aza bicyclo[2.1.1]hexan-5-yl)-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyr rolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidin-1-yl)ethan-1-one;

3-(1-((7 ?,4 ?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-7-(chroman-8-yl)-6-fluo ro-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)-2,3- dihydroindolizin-5(1/7)-one;

1-(( ?)-1-(1-((7 ?,4 ?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-7-(chroman-8-yl)-6-fluo ro-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2- c][1 ,6]naphthyridin-2- y l)ethy I) py rid i n-2( 1 /-/)-one;

Methyl (F?)-2-(1-((7F?,4F?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2, 3-dichlorophenyl)-4- (3-(dimethylamino)-3-methylazetidin-1-yl)-6-fluoro-1/7-pyrro lo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidine-1 -carboxylate; Methyl (R)-2-(1-((7R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2,3-d ichlorophenyl)-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyr rolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidine-1 -carboxylate; and

Methyl (R)-2-(1-((7R,4F?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2-ch loro-3- methylphenyl)-6-fluoro-4-(6-(methylcarbamoyl)pyridin-2-yl)-1 /7-pyrrolo[3,2- c][1 ,6]naphthyridin-2-yl)pyrrolidine-1 -carboxylate; or a pharmaceutically acceptable salt thereof.

In yet another embodiment, the compound of Formula I is selected from: methyl (F?)-2-(7-(benzo[d]thiazol-4-yl)-1-((1F?,4F?,5S)-2-azabicycl o[2.1.1]hexan-5-yl)- 6-fluoro-4-(((S)-1-(pyrrolidin-1-yl)propan-2-yl)oxy)-1/7-pyr rolo[3,2-c][1 ,6]naphthyridin-2- yl)pyrrolidine-1 -carboxylate;

(7aS)-5-(1-((1R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-( chroman-8-yl)-6-fluoro-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3,2- c][1 ,6]naphthyridin-2-yl)tetrahydro- 1/7,3/7-pyrrolo[1 ,2-c]oxazol-3-one; and

4-(1-((1R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(chroma n-8-yl)-6-fluoro-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 /7-pyrrolo[3,2-c][1 ,6]naphthyridin-2-yl)-/V,/\/, 1 -trimethyl- 1 /7-pyrazole-3-carboxamide; or a pharmaceutically acceptable salt thereof.

In an embodiment, compounds of the Formulae herein are compounds of the Formulae or pharmaceutically acceptable salts thereof.

In another aspect, provided herein is a pharmaceutical composition comprising the compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

In an aspect, provided herein is a method of inhibiting a KRAS protein harboring a G12C mutation, said method comprising contacting a compound of the instant disclosure with KRAS.

In another aspect, provided herein is a method of inhibiting a KRAS protein harboring a G12D mutation, said method comprising contacting a compound of the instant disclosure with KRAS.

In yet another aspect, provided herein is a method of inhibiting a KRAS protein harboring a G12V mutation, said method comprising contacting a compound of the instant disclosure with KRAS.

It is further appreciated that certain features of the present disclosure, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment (while the embodiments are intended to be combined as if written in multiply dependent form). Conversely, various features of the present disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination. Thus, it is contemplated as features described as embodiments of the compounds of Formula I can be combined in any suitable combination.

At various places in the present specification, certain features of the compounds are disclosed in groups or in ranges. It is specifically intended that such a disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term “Ci-e alkyl” is specifically intended to individually disclose (without limitation) methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl and Ce alkyl.

The term “n-membered,” where n is an integer, typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n. For example, piperidinyl is an example of a 6-membered heterocycloalkyl ring, pyrazolyl is an example of a 5-membered heteroaryl ring, pyridyl is an example of a 6-membered heteroaryl ring and 1 ,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.

At various places in the present specification, variables defining divalent linking groups may be described. It is specifically intended that each linking substituent include both the forward and backward forms of the linking substituent. For example, -NR(CR'R") n - includes both -NR(CR'R") n - and -(CR'R") n NR- and is intended to disclose each of the forms individually. Where the structure requires a linking group, the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists “alkyl” or “aryl” then it is understood that the “alkyl” or “aryl” represents a linking alkylene group or arylene group, respectively.

The term “substituted” means that an atom or group of atoms formally replaces hydrogen as a “substituent” attached to another group. The term “substituted,” unless otherwise indicated, refers to any level of substitution, e.g., mono-, di-, tri-, tetra- or penta-substitution, where such substitution is permitted. The substituents are independently selected, and substitution may be at any chemically accessible position. It is to be understood that substitution at a given atom is limited by valency. It is to be understood that substitution at a given atom results in a chemically stable molecule. The phrase “optionally substituted” means unsubstituted or substituted. The term “substituted” means that a hydrogen atom is removed and replaced by a substituent. A single divalent substituent, e.g., oxo, can replace two hydrogen atoms.

The term “C n -m” indicates a range which includes the endpoints, wherein n and m are integers and indicate the number of carbons. Examples include C1.4, Ci-e and the like.

The term “alkyl” employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chained or branched. The term “C n -m alkyl,” refers to an alkyl group having n to m carbon atoms. An alkyl group formally corresponds to an alkane with one C-H bond replaced by the point of attachment of the alkyl group to the remainder of the compound. In some embodiments, the alkyl group contains from 1 to 6 carbon atoms, from 1 to 4 carbon atoms, from 1 to 3 carbon atoms, or 1 to 2 carbon atoms. Examples of alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, ferf-butyl, isobutyl, sec-butyl; higher homologs such as 2-methyl-1 -butyl, n-pentyl, 3-pentyl, n-hexyl, 1,2,2-trimethylpropyl and the like.

The term “alkenyl” employed alone or in combination with other terms, refers to a straight-chain or branched hydrocarbon group corresponding to an alkyl group having one or more double carbon-carbon bonds. An alkenyl group formally corresponds to an alkene with one C-H bond replaced by the point of attachment of the alkenyl group to the remainder of the compound. The term “C n -m alkenyl” refers to an alkenyl group having n to m carbons. In some embodiments, the alkenyl moiety contains 2 to 6, 2 to 4, or 2 to 3 carbon atoms. Example alkenyl groups include, but are not limited to, ethenyl, n-propenyl, isopropenyl, n- butenyl, sec-butenyl and the like.

The term “alkynyl” employed alone or in combination with other terms, refers to a straight-chain or branched hydrocarbon group corresponding to an alkyl group having one or more triple carbon-carbon bonds. An alkynyl group formally corresponds to an alkyne with one C-H bond replaced by the point of attachment of the alkyl group to the remainder of the compound. The term “C n -m alkynyl” refers to an alkynyl group having n to m carbons. Example alkynyl groups include, but are not limited to, ethynyl, propyn-1-yl, propyn-2-yl and the like. In some embodiments, the alkynyl moiety contains 2 to 6, 2 to 4, or 2 to 3 carbon atoms.

The term “alkylene,” employed alone or in combination with other terms, refers to a divalent alkyl linking group. An alkylene group formally corresponds to an alkane with two C-H bond replaced by points of attachment of the alkylene group to the remainder of the compound. The term “C n -m alkylene” refers to an alkylene group having n to m carbon atoms. Examples of alkylene groups include, but are not limited to, ethan-1 ,2-diyl, ethan-1 ,1- diyl, propan-1 , 3-diyl, propan-1 , 2-diyl, propan-1, 1-diyl, butan-1 ,4-diyl, butan-1 ,3-diyl, butan- 1 ,2-diyl, 2-methyl-propan-1 , 3-diyl and the like.

The term “alkoxy,” employed alone or in combination with other terms, refers to a group of formula -O-alkyl, wherein the alkyl group is as defined above. The term “C n -m alkoxy” refers to an alkoxy group, the alkyl group of which has n to m carbons. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t- butoxy and the like. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms. The term “C n -m dialkoxy” refers to a linking group of formula -0-(C n -m alkyl)-O-, the alkyl group of which has n to m carbons. Example dialkyoxy groups include -OCH2CH2O- and OCH2CH2CH2O-. In some embodiments, the two O atoms of a C n -m dialkoxy group may be attached to the same B atom to form a 5- or 6- membered heterocycloalkyl group.

The term “alkylthio,” employed alone or in combination with other terms, refers to a group of formula -S-alkyl, wherein the alkyl group is as defined above.

The term “amino,” employed alone or in combination with other terms, refers to a group of formula -NH2, wherein the hydrogen atoms may be substituted with a substituent described herein. For example, “alkylamino” can refer to -NH(alkyl) and -N(alkyl)2.

The term “carbonyl,” employed alone or in combination with other terms, refers to a -C(=O)- group, which also may be written as C(O).

The term “cyano” or “nitrile” refers to a group of formula -C=N, which also may be written as -CN.

The term “carbamyl,” as used herein, refers to a -NHC(O)O- or -OC(O)NH- group, wherein the carbon atom is doubly bound to one oxygen atom, and singly bound to a nitrogen and second oxygen atom.

The terms “halo” or “halogen,” used alone or in combination with other terms, refers to fluoro, chloro, bromo and iodo. In some embodiments, “halo” refers to a halogen atom selected from F, Cl, or Br. In some embodiments, halo groups are F.

The term “haloalkyl” as used herein refers to an alkyl group in which one or more of the hydrogen atoms has been replaced by a halogen atom. The term “C n -m haloalkyl” refers to a Cn-m alkyl group having n to m carbon atoms and from at least one up to {2(n to m)+1} halogen atoms, which may either be the same or different. In some embodiments, the halogen atoms are fluoro atoms. In some embodiments, the haloalkyl group has 1 to 6 or 1 to 4 carbon atoms. Example haloalkyl groups include CF3, C2F5, CHF2, CH2F, CC , CHCI2, C2CI5 and the like. In some embodiments, the haloalkyl group is a fluoroalkyl group.

The term “haloalkoxy,” employed alone or in combination with other terms, refers to a group of formula -O-haloalkyl, wherein the haloalkyl group is as defined above. The term “Cn-m haloalkoxy” refers to a haloalkoxy group, the haloalkyl group of which has n to m carbons. Example haloalkoxy groups include trifluoromethoxy and the like. In some embodiments, the haloalkoxy group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.

The term “oxo” or “oxy” refers to an oxygen atom as a divalent substituent, forming a carbonyl group when attached to carbon, or attached to a heteroatom forming a sulfoxide or sulfone group, or an /V-oxide group. In some embodiments, heterocyclic groups may be optionally substituted by 1 or 2 oxo (=0) substituents.

The term “sulfonyl” refers to a -SO2- group wherein a sulfur atom is doubly bound to two oxygen atoms.

The term “sulfinyl” refers to a -SO- group wherein a sulfur atom is doubly bound to one oxygen atom. The term “oxidized” in reference to a ring-forming N atom refers to a ring-forming N- oxide.

The term “oxidized” in reference to a ring-forming S atom refers to a ring-forming sulfonyl or ring-forming sulfinyl.

The term “aromatic” refers to a carbocycle or heterocycle having one or more polyunsaturated rings having aromatic character (/.e., having (4n + 2) delocalized (pi) electrons where n is an integer).

The term “aryl,” employed alone or in combination with other terms, refers to an aromatic hydrocarbon group, which may be monocyclic or polycyclic (e.g., having 2 fused rings). The term “C n -m aryl” refers to an aryl group having from n to m ring carbon atoms. Aryl groups include, e.g., phenyl, naphthyl, and the like. In some embodiments, aryl groups have from 6 to about 10 carbon atoms. In some embodiments, aryl groups have 6 carbon atoms. In some embodiments, aryl groups have 10 carbon atoms. In some embodiments, the aryl group is phenyl. In some embodiments, the aryl group is naphthyl.

The term “heteroaryl” or “heteroaromatic,” employed alone or in combination with other terms, refers to a monocyclic or polycyclic aromatic heterocycle having at least one heteroatom ring member selected from sulfur, oxygen and nitrogen. In some embodiments, the heteroaryl ring has 1 , 2, 3 or 4 heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, any ring-forming N in a heteroaryl moiety can be an N-oxide. In some embodiments, the heteroaryl has 5-14 ring atoms including carbon atoms and 1 , 2, 3 or 4 heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl has 5-10 ring atoms including carbon atoms and 1 , 2, 3 or 4 heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl has 5-6 ring atoms and 1 or 2 heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl is a five-membered or six-membered heteroaryl ring. In other embodiments, the heteroaryl is an eight-membered, nine-membered or ten-membered fused bicyclic heteroaryl ring. Example heteroaryl groups include, but are not limited to, pyridinyl (pyridyl), pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, pyrazolyl, azolyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, furanyl, thio-phenyl, quinolinyl, isoquinolinyl, naphthyridinyl (including 1 ,2-, 1 ,3-, 1 ,4-, 1 ,5-, 1 ,6-, 1 ,7-, 1 ,8-, 2,3- and 2,6-naphthyridine), indolyl, isoindolyl, benzothiophenyl, benzofuranyl, benzisoxazolyl, imidazo[1 ,2-b]thiazolyl, purinyl, and the like. In some embodiments, the heteroaryl group is pyridone (e.g., 2- pyridone). In some embodiments, the heteroaryl group is chromanyl.

A five-membered heteroaryl ring is a heteroaryl group having five ring atoms wherein one or more (e.g., 1 , 2 or 3) ring atoms are independently selected from N, O and S. Exemplary five-membered ring heteroaryls include thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1 ,2,3-triazolyl, tetrazolyl, 1 ,2,3- thiadiazolyl, 1 ,2,3-oxadiazolyl, 1 ,2,4-triazolyl, 1 ,2,4-thiadiazolyl, 1 ,2,4-oxadiazolyl, 1 ,3,4- triazolyl, 1 ,3,4-thiadiazolyl and 1 ,3,4-oxadiazolyl.

A six-membered heteroaryl ring is a heteroaryl group having six ring atoms wherein one or more (e.g., 1 , 2 or 3) ring atoms are independently selected from N, O and S. Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl, isoindolyl, and pyridazinyl.

The term “cycloalkyl,” employed alone or in combination with other terms, refers to a non-aromatic hydrocarbon ring system (monocyclic, bicyclic or polycyclic), including cyclized alkyl and alkenyl groups. The term “C n -m cycloalkyl” refers to a cycloalkyl that has n to m ring member carbon atoms. Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) groups and spirocycles. Cycloalkyl groups can have 3, 4, 5, 6 or 7 ringforming carbons (C3-7). In some embodiments, the cycloalkyl group has 3 to 6 ring members, 3 to 5 ring members, or 3 to 4 ring members. In some embodiments, the cycloalkyl group is monocyclic. In some embodiments, the cycloalkyl group is monocyclic or bicyclic. In some embodiments, the cycloalkyl group is a C3-6 monocyclic cycloalkyl group. Ring-forming carbon atoms of a cycloalkyl group can be optionally oxidized to form an oxo or sulfido group. Cycloalkyl groups also include cycloalkylidenes. In some embodiments, cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings fused (/.e., having a bond in common with) to the cycloalkyl ring, e.g., benzo or thienyl derivatives of cyclopentane, cyclohexane and the like. A cycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, bicyclo[1.1.1]pentanyl, bicyclo[2.1.1]hexanyl, and the like. In some embodiments, the cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

The term “heterocycloalkyl,” employed alone or in combination with other terms, refers to a non-aromatic ring or ring system, which may optionally contain one or more alkenylene groups as part of the ring structure, which has at least one heteroatom ring member independently selected from nitrogen, sulfur, oxygen and phosphorus, and which has 4-10 ring members, 4-7 ring members, or 4-6 ring members. Included within the term “heterocycloalkyl” are monocyclic 4-, 5-, 6- and 7-membered heterocycloalkyl groups. Heterocycloalkyl groups can include mono- or bicyclic (e.g., having two fused or bridged rings) or spirocyclic ring systems. In some embodiments, the heterocycloalkyl group is a monocyclic group having 1 , 2 or 3 heteroatoms independently selected from nitrogen, sulfur and oxygen. Ring-forming carbon atoms and heteroatoms of a heterocycloalkyl group can be optionally oxidized to form an oxo or sulfido group or other oxidized linkage (e.g., C(O), S(O), C(S) or S(O) 2 , /V-oxide etc.) or a nitrogen atom can be quaternized. The heterocycloalkyl group can be attached through a ring-forming carbon atom or a ringforming heteroatom. In some embodiments, the heterocycloalkyl group contains 0 to 3 double bonds. In some embodiments, the heterocycloalkyl group contains 0 to 2 double bonds. Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings fused (/.e., having a bond in common with) to the heterocycloalkyl ring, e.g., benzo or thienyl derivatives of piperidine, morpholine, azepine, etc. A heterocycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring. Examples of heterocycloalkyl groups include 2,5-diazabicyclo[2.2.1]heptanyl; pyrrolidinyl; hexahydropyrrolo[3,4-b]pyrrol-1(2/7)-yl; 1,6- dihydropyridinyl; morpholinyl; azetidinyl; piperazinyl; and 4,7-diazaspiro[2.5]octan-7-yl.

At certain places, the definitions or embodiments refer to specific rings (e.g., an azetidine ring, a pyridine ring, etc.). Unless otherwise indicated, these rings can be attached to any ring member provided that the valency of the atom is not exceeded. For example, an azetidine ring may be attached at any position of the ring, whereas an azetidin-3-yl ring is attached at the 3-position.

The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.

Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art. One method includes fractional recrystallization using a chiral resolving acid which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, e.g., optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as - camphorsulfonic acid. Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a-methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, /V- methylephedrine, cyclohexylethylamine, 1 ,2-diaminocyclohexane and the like.

Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.

In some embodiments, the compounds disclosed herein have the (^-configuration. In other embodiments, the compounds have the (S)-configuration. In compounds with more than one chiral centers, each of the chiral centers in the compound may be independently (/?) or (S), unless otherwise indicated.

Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, e.g., H- and 3/7-imidazole, 1/7-, 2/7- and 4/7- 1 ,2,4-triazole, 1/7- and 2/7- isoindole and 1/7- and 2/7-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.

Compounds disclosed herein can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium. One or more constituent atoms of the compounds of the present disclosure can be replaced or substituted with isotopes of the atoms in natural or non-natural abundance. In some embodiments, the compound includes at least one deuterium atom. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced or substituted by deuterium. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 deuterium atoms. Synthetic methods for including isotopes into organic compounds are known in the art (Deuterium Labeling in Organic Chemistry by Alan F. Thomas (New York, N.Y., Appleton-Century-Crofts, 1971; The Renaissance of H/D Exchange by Jens Atzrodt, Volker Derdau, Thorsten Fey and Jochen Zimmermann, Angew. Chem. Int. Ed. 2007, 7744-7765; The Organic Chemistry of Isotopic Labelling by James R. Hanson, Royal Society of Chemistry, 2011). Isotopically labeled compounds can used in various studies such as NMR spectroscopy, metabolism experiments, and/or assays.

Substitution with heavier isotopes such as deuterium, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. (A. Kerekes et.al. J. Med. Chem. 2011, 54, 201-210; R. Xu et.al. J. Label Compd. Radiopharm. 2015, 58, 308-312).

The term “compound” as used herein is meant to include all stereoisomers, geometric isomers, tautomers and isotopes of the structures depicted. The term is also meant to refer to compounds provided herein, regardless of how they are prepared, e.g., synthetically, through biological process (e.g., metabolism or enzyme conversion), or a combination thereof.

All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g., hydrates and solvates) or can be isolated. When in the solid state, the compounds described herein and salts thereof may occur in various forms and may, e.g., take the form of solvates, including hydrates. The compounds may be in any solid state form, such as a polymorph or solvate, so unless clearly indicated otherwise, reference in the specification to compounds and salts thereof should be understood as encompassing any solid state form of the compound.

In some embodiments, the compounds disclosed herein, or salts thereof, are substantially isolated. By “substantially isolated” is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, e.g., a composition enriched in the present compounds. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds disclosed herein, or salt thereof.

The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

The expressions “ambient temperature” and “room temperature,” as used herein, are understood in the art, and refer generally to a temperature, e.g., a reaction temperature, that is about the temperature of the room in which the reaction is carried out, e.g., a temperature from about 20 °C to about 30 °C.

The present disclosure also includes pharmaceutically acceptable salts of the compounds described herein. The term “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present disclosure include the non-toxic salts of the parent compound formed, e.g., from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol or butanol) or acetonitrile (MeCN) are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17 th Ed., (Mack Publishing Company, Easton, 1985), p. 1418, Berge et al., J. Pharm. Sci., 1977, 66(1), 1-19 and in Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, (Wiley, 2002). In some embodiments, the compounds described herein include the N-oxide forms.

Compounds provided herein, including salts thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes, such as those in the Schemes below.

The reactions for preparing compounds of the present disclosure can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.

Preparation of compounds provided herein can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups is described, e.g., in Kocienski, Protecting Groups, (Thieme, 2007); Robertson, Protecting Group Chemistry, (Oxford University Press, 2000); Smith et al., March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 6 th Ed. (Wiley, 2007); Peturssion et al., “Protecting Groups in Carbohydrate Chemistry,” J. Chem. Educ., 1997, 74(11), 1297; and Wuts et al., Protective Groups in Organic Synthesis, 4th Ed., (Wiley, 2006).

Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry or by chromatographic methods such as high-performance liquid chromatography (HPLC) or thin layer chromatography (TLC). The Schemes below provide general guidance in connection with preparing the compounds of the present disclosure. One skilled in the art would understand that the preparations shown in the Schemes can be modified or optimized using general knowledge of organic chemistry to prepare various compounds provided herein.

Scheme 1 Compounds of formula 1-22 and 1-23 can be prepared via the synthetic route outlined in Scheme 1. Starting material 1-1 can undergo iodination with /V-iodo-succinimide (NIS) to form intermediate 1-2, which can then be subjected to carbonylation conditions with palladium catalyst and carbon monoxide in methanol to give intermediate 1-3. Treating intermediate 1-3 with ethyl malonyl chloride can afford compound 1-4, which can cyclize to give intermediate 1-5 in the presence of sodium ethoxide. Compound 1-6 can be obtained by refluxing intermediate 1-5 in a mixture of concentrated hydrochloride acid and 1 ,4-dioxane. Nitration of compound 1-6 with nitric acid in acetic acid can deliver the nitro compound 1-7, which can be converted into the common intermediate 1-8 by POCh. A SNAr reaction of intermediate 1-8 with amine 1-9 (PG is an appropriate protecting group, such as Boc) can be carried out to generate compound 1-10, which can undergo another SNAr reaction with sodium thiomethoxide to yield intermediate 1-11. Compound 1-13 can be prepared from intermediate 1-11 by Boc protection and subsequent reduction of the nitro group to NH2 with reducing agents (e.g. Fe powder or sodium dithionite). The NH2 group of compound 1-13 can be converted to iodo atom when subjected to Sandmeyer reaction conditions to give intermediate 1-14, of which the Boc group can be removed by trifluoroacetic acid to deliver compound 1-15. A cross-coupling reaction between 1-15 and alkynes of formula 1-16, in which X is a hydrogen atom or an appropriately substituted metal [e.g. Sn(Alkyl)s] under standard Sonogashira coupling conditions (e.g., in the presence of a palladium catalyst), or standard Stille cross-coupling conditions (e.g., in the presence of a palladium catalyst), can generate intermediate 1-17, which can cyclize to form the pyrrole ring in the presence of a gold catalyst to deliver compound 1-18. Intermediate 1-20 can be prepared by a crosscoupling reaction between 1-18 and an adduct of formula 1-19, in which Cy 1 -M is a boronic acid, boronic ester or an appropriately substituted metal reagent [e.g., M is B(OR)2, Sn(Alkyl)s, or Zn-Hal], or a NH-containing heterocycle, under standard Suzuki cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base), or standard Stille cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Negishi cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Buchwald-Hartwig cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base). Intermediate 1-20 can be converted to intermediate 1-21 either through oxidation of the sulfur group with a suitable oxidant (e.g. m-CPBA) followed by an SNAr reaction, or a cross-coupling reaction (Org. Lett. 2002, 4, 979-981). Removal of the protecting group in 1-21 can afford the desired product 1-22. Alternatively, after removing the protecting group in 1-21 , the free amine can be capped (by various moieties) through electrophilic substitution, or reductive amination, or amide coupling, or cross-coupling reactions, to deliver 1-23. The order of the above described chemical reactions can be rearranged or omitted as appropriate to suit the preparation of different analogues. Scheme 2

Compounds of formula 2-10 can be prepared via the synthetic route outlined in Scheme 2. A SNAr reaction of intermediate 1-8 with amine 2-1 (PG is an appropriate protecting group, such as Boc) can be carried out to generate compound 2-2, which can undergo another SNAr reaction with sodium thiomethoxide to yield intermediate 2-3. Compound 2-4 can be prepared from intermediate 2-3 by reduction of the nitro group to NH2 with reducing agents (e.g. Fe powder or sodium dithionite). Cyclization between 2-4 and an aldehyde 2-5 can generate intermediate 2-6, which can undergo a cross-coupling reaction with an adduct of formula 2-7, in which Cy 1 -M is a boronic acid, boronic ester or an appropriately substituted metal reagent [e.g., M is B(OR)2, Sn(Alkyl)s, or Zn-Hal], or a NH- containing heterocycle, under standard Suzuki cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base), or standard Stille cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Negishi cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Buchwald-Hartwig cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base), to yield compound 2-8. Intermediate 2-9 can be prepared from 2-8 either through oxidation of the sulfur group with a suitable oxidant (e.g. m-CPBA) followed by an SNAr reaction, or a cross-coupling reaction (Org. Lett. 2002, 4, 979-981). Removal of the protecting group in 2-9 can afford the desired product 2-10. The free NH group in 2-10 can also be capped by various moieties as illustrated in Scheme 1. The order of the above described chemical reactions can be rearranged or omitted as appropriate to suit the preparation of different analogues.

Scheme 3

Compounds of formula 3-9 can be prepared via the synthetic route outlined in Scheme 3. A SNAr reaction of intermediate 1-8 with amine 3-1 (PG is an appropriate protecting group, such as Boc) can be carried out to generate compound 3-2, which can undergo another SNAr reaction with a nucleophile Nu-H [e.g. amine, alcohol, or phenol] to yield intermediate 3-3. Compound 3-4 can be prepared from intermediate 3-3 by reduction of the nitro group to NH2 with reducing agents (e.g. Fe powder or sodium dithionite). Cyclization between 3-4 and an aldehyde 3-5 can generate intermediate 3-6, which can undergo a cross-coupling reaction with an adduct of formula 3-7, in which Cy 1 -M is a boronic acid, boronic ester or an appropriately substituted metal reagent [e.g., M is B(OR)2, Sn(Alkyl)s, or Zn-Hal], or a NH-containing heterocycle, under standard Suzuki cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base), or standard Stille cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Negishi cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Buchwald-Hartwig cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base), to yield compound 3-8. Removal of the protecting group in 3-8 can afford the desired product 3-9. The free NH group in 3-9 can also be capped by various moieties as illustrated in Scheme 1. The order of the above described chemical reactions can be rearranged or omitted as appropriate to suit the preparation of different analogues. Scheme 4

Compounds of formula 4-9 can be prepared via the synthetic route outlined in Scheme 4. A SNAr reaction of intermediate 1-8 with amine 4-1 (PG is an appropriate protecting group, such as Boc) can be carried out to generate compound 4-2, which can undergo another SNAr reaction with sodium thiomethoxide to yield intermediate 4-3. Compound 4-4 can be prepared from intermediate 4-3 by reduction of the nitro group to NH2 with reducing agents (e.g. Fe powder or sodium dithionite). Treating intermediate 4-4 with NaNC>2 in acetic acid can generate intermediate 4-5. Compound 4-7 can be prepared by a cross-coupling reaction between 4-5 and an adduct of formula 4-6, in which Cy 1 -M is a boronic acid, boronic ester or an appropriately substituted metal reagent [e.g., M is B(OR)2, Sn(Alkyl)s, or Zn-Hal], or a NH-containing heterocycle, under standard Suzuki cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base), or standard Stille cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Negishi cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Buchwald-Hartwig cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base). Compound 4-7 can be converted to 4-8 either through oxidation of the sulfur group with a suitable oxidant (e.g. m-CPBA) followed by an SNAr reaction, or a crosscoupling reaction (Org. Lett. 2002, 4, 979-981). Removal of the protecting group in 4-8 can afford the intermediate 4-9. The free NH group in 4-9 can also be capped by various moieties as illustrated in Scheme 1 . The order of the above described chemical reactions can be rearranged or omitted as appropriate to suit the preparation of different analogues.

Scheme 5 Compounds of formula 5-20 can be prepared via the synthetic route outlined in

Scheme 5. Halogenation of starting material 5-1 with an appropriate reagent, such as N- chloro-succinimide (NCS), /V-Bromo-succinimide (NBS), or /V-lodo-succinimide (NIS), affords intermediate 5-2 (Hal is a halide, such as F, Cl, Br, or I). Compound 5-4 can then be prepared by a amide coupling reaction of 5-2 with appropriate acyl chloride 5-3. Condensation of 5-4 followed by decarboxylation can deliver intermediate 5-6. Nitration of intermediate 5-6 gives the nitro adduct 5-7, which upon treatment with a reagent, such as POCh, yields intermediate 5-8. A SNAr reaction of 5-8 with an amine of formula 5-9 (PG is an appropriate protecting group, such as Boc) followed by a second SNAr with another nucleophile generate compound 5-11. The nitro functionality in 5-11 can be reduced to -NH2 under suitable conditions (such as using Fe in acetic acid), followed by a cyclization reaction to deliver compound 5-13. Compound 5-19 can be prepared via sequential coupling reactions (and/or SNAr reactions) of intermediate 5-13 using appropriate coupling partners, in which Cy 1 -M is a boronic acid, boronic ester or an appropriately substituted metal reagent [e.g., M is B(0R)2, Sn(Alkyl)3, or Zn-Hal, Li, Na, K], or a NH-containing heterocycle, under standard Suzuki Cross-Coupling conditions {e.g., in the presence of a palladium catalyst and a suitable base), or standard Stille cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Negishi cross-coupling conditions (e.g., in the presence of a palladium catalyst), or standard Buchwald-Hartwig cross-coupling conditions (e.g., in the presence of a palladium catalyst and a suitable base). Removal of the protecting group in 5- 19 affords the desired product 5-20. The free NH group in 5-20 can also be capped by various moieties as illustrated in Scheme 1. The order of the above described chemical reactions can be rearranged or omitted as appropriate to suit the preparation of different analogues.

KRAS Protein

The Ras family is comprised of three members: KRAS, NRAS and HRAS. RAS mutant cancers account for about 25% of human cancers. KRAS is the most frequently mutated isoform in human cancers: 85% of all RAS mutations are in KRAS, 12% in NRAS, and 3% in HRAS (Simanshu, D. et al. Cell 170.1 (2017):17-33). KRAS mutations are prevalent amongst the top three most deadly cancer types: pancreatic (97%), colorectal (44%), and lung (30%) (Cox, A.D. et al. Nat Rev Drug Discov (2014) 13:828-51). The majority of RAS mutations occur at amino acid residues/codons 12, 13, and 61; Codon 12 mutations are most frequent in KRAS. The frequency of specific mutations varied between RAS genes and G12D mutations are most predominant in KRAS whereas Q61R and G12R mutations are most frequent in NRAS and HRAS. Furthermore, the spectrum of mutations in a RAS isoform differs between cancer types. For example, KRAS G12D mutations predominate in pancreatic cancers (51%), followed by colorectal adenocarcinomas (45%) and lung cancers (17%) (Cox, A.D. et al. Nat Rev Drug Discov (2014) 13:828-51). In contrast, KRAS G12C mutations predominate in non-small cell lung cancer (NSCLC) comprising 11-16% of lung adenocarcinomas (nearly half of mutant KRAS is G12C), as well as 2-5% of pancreatic and colorectal adenocarcinomas, respectively (Cox, A.D. et al. Nat. Rev. Drug Discov. (2014) 13:828-51). Using shRNA knockdown thousands of genes across hundreds of cancer cell lines, genomic studies have demonstrated that cancer cells exhibiting KRAS mutations are highly dependent on KRAS function for cell growth (McDonald, R. et al. Cell 170 (2017): 577-592). Taken together, these findings suggested that KRAS mutations play a critical role in human cancers, therefore development of the inhibitors targeting mutant KRAS may be useful in the clinical treatment of diseases that have characterized by a KRAS mutation. Methods of Use

The cancer types in which KRAS harboring G12C, G12V, and G12D mutations are implicated include, but are not limited to: carcinomas (e.g., pancreatic, colorectal, lung, bladder, gastric, esophageal, breast, head and neck, cervical skin, thyroid); hematopoietic malignancies (e.g., myeloproliferative neoplasms (MPN), myelodysplastic syndrome (MDS), chronic and juvenile myelomonocytic leukemia (CMML and JMML), acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL) and multiple myeloma (MM)); and other neoplasms (e.g., glioblastoma and sarcomas). In addition, KRAS mutations were found in acquired resistance to anti-EGFR therapy (Knickelbein, K.et al. Genes & Cancer, (2015): 4- 12). KRAS mutations were found in immunological and inflammatory disorders (Fernandez- Medarde, A. et al. Genes & Cancer, (2011): 344-358) such as Ras-associated lymphoproliferative disorder (RALD) or juvenile myelomonocytic leukemia (JMML) caused by somatic mutations of KRAS or NRAS.

Compounds of the present disclosure can inhibit the activity of the KRAS protein. For example, compounds of the present disclosure can be used to inhibit activity of KRAS in a cell or in an individual or patient in need of inhibition of the enzyme by administering an inhibiting amount of one or more compounds of the present disclosure to the cell, individual, or patient.

As KRAS inhibitors, the compounds of the present disclosure are useful in the treatment of various diseases associated with abnormal expression or activity of KRAS. Compounds which inhibit KRAS will be useful in providing a means of preventing the growth or inducing apoptosis in tumors, or by inhibiting angiogenesis. It is therefore anticipated that compounds of the present disclosure will prove useful in treating or preventing proliferative disorders such as cancers. In particular, tumors with activating mutants of receptor tyrosine kinases or upregulation of receptor tyrosine kinases may be particularly sensitive to the inhibitors.

In an aspect, provided herein is a method of inhibiting KRAS activity, said method comprising contacting a compound of the instant disclosure with KRAS. In an embodiment, the contacting comprises administering the compound to a patient.

In another aspect, provided herein a is method of treating a disease or disorder associated with inhibition of KRAS interaction, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any of the formulae disclosed herein, or pharmaceutically acceptable salt thereof.

In an embodiment, the disease or disorder is an immunological or inflammatory disorder. In another embodiment, the immunological or inflammatory disorder is Ras- associated lymphoproliferative disorder and juvenile myelomonocytic leukemia caused by somatic mutations of KRAS.

In an aspect, provided herein is a method of treating a disease or disorder associated with inhibiting a KRAS protein harboring a G12C mutation, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any of the formulae disclosed herein, or pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of treating cancer in a patient in need thereof comprising administering to the patient a therapeutically effective amount of the compounds disclosed herein wherein the cancer is characterized by an interaction with a KRAS protein harboring a G12C mutation.

In another aspect, provided herein is a method of treating a disease or disorder associated with inhibiting a KRAS protein harboring a G12D mutation, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any of the formulae disclosed herein, or pharmaceutically acceptable salt thereof.

In yet another aspect, provided herein is also a method of treating cancer in a patient in need thereof comprising administering to the patient a therapeutically effective amount of the compounds disclosed herein wherein the cancer is characterized by an interaction with a KRAS protein harboring a G12D mutation.

In another aspect, provided herein is a method of treating a disease or disorder associated with inhibiting a KRAS protein harboring a G12V mutation, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any of the formulae disclosed herein, or pharmaceutically acceptable salt thereof.

In yet another aspect, provided herein is also a method of treating cancer in a patient in need thereof comprising administering to the patient a therapeutically effective amount of the compounds disclosed herein wherein the cancer is characterized by an interaction with a KRAS protein harboring a G12V mutation.

In yet another aspect, provided herein is a method for treating a cancer in a patient, said method comprising administering to the patient a therapeutically effective amount of any one of the compounds disclosed herein, or pharmaceutically acceptable salt thereof.

In an embodiment, the cancer is selected from carcinomas, hematological cancers, sarcomas, and glioblastoma.

In another embodiment, the hematological cancer is selected from myeloproliferative neoplasms, myelodysplastic syndrome, chronic and juvenile myelomonocytic leukemia, acute myeloid leukemia, acute lymphocytic leukemia, and multiple myeloma. In yet another embodiment, the carcinoma is selected from pancreatic,] colorectal, lung, bladder, gastric, esophageal, breast, head and neck, cervical, skin, and thyroid.

In another aspect, provided herein is a method for treating a disease or disorder associated with inhibition of KRAS interaction or a mutant thereof in a patient in need thereof comprising the step of administering to the patient a compound disclosed herein, or a pharmaceutically acceptable salt thereof, or a composition comprising a compound disclosed herein or a pharmaceutically acceptable salt thereof, in combination with another therapy or therapeutic agent as described herein.

In an embodiment, the cancer is selected from hematological cancers, sarcomas, lung cancers, gastrointestinal cancers, genitourinary tract cancers, liver cancers, bone cancers, nervous system cancers, gynecological cancers, and skin cancers.

In another embodiment, the lung cancer is selected from non-small cell lung cancer (NSCLC), small cell lung cancer, bronchogenic carcinoma, squamous cell bronchogenic carcinoma, undifferentiated small cell bronchogenic carcinoma, undifferentiated large cell bronchogenic carcinoma, adenocarcinoma, bronchogenic carcinoma, alveolar carcinoma, bronchiolar carcinoma, bronchial adenoma, chondromatous hamartoma, mesothelioma, pavicellular and non-pavicellular carcinoma, bronchial adenoma, and pleuropulmonary blastoma.

In yet another embodiment, the lung cancer is non-small cell lung cancer (NSCLC). In still another embodiment, the lung cancer is adenocarcinoma.

In an embodiment, the gastrointestinal cancer is selected from esophagus squamous cell carcinoma, esophagus adenocarcinoma, esophagus leiomyosarcoma, esophagus lymphoma, stomach carcinoma, stomach lymphoma, stomach leiomyosarcoma, exocrine pancreatic carcinoma, pancreatic ductal adenocarcinoma, pancreatic insulinoma, pancreatic glucagonoma, pancreatic gastrinoma, pancreatic carcinoid tumors, pancreatic vipoma, small bowel adenocarcinoma, small bowel lymphoma, small bowel carcinoid tumors, Kaposi's sarcoma, small bowel leiomyoma, small bowel hemangioma, small bowel lipoma, small bowel neurofibroma, small bowel fibroma, large bowel adenocarcinoma, large bowel tubular adenoma, large bowel villous adenoma, large bowel hamartoma, large bowel leiomyoma, colorectal cancer, gall bladder cancer, and anal cancer.

In an embodiment, the gastrointestinal cancer is colorectal cancer.

In another embodiment, the cancer is a carcinoma. In yet another embodiment, the carcinoma is selected from pancreatic carcinoma, colorectal carcinoma, lung carcinoma, bladder carcinoma, gastric carcinoma, esophageal carcinoma, breast carcinoma, head and neck carcinoma, cervical skin carcinoma, and thyroid carcinoma. In still another embodiment, the cancer is a hematopoietic malignancy. In an embodiment, the hematopoietic malignancy is selected from multiple myeloma, acute myelogenous leukemia, and myeloproliferative neoplasms.

In another embodiment, the cancer is a neoplasm. In yet another embodiment, the neoplasm is glioblastoma or sarcomas.

In certain embodiments, the disclosure provides a method for treating a KRAS- mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to the present disclosure, or a pharmaceutically acceptable composition thereof.

In some embodiments, diseases and indications that are treatable using the compounds of the present disclosure include, but are not limited to hematological cancers, sarcomas, lung cancers, gastrointestinal cancers, genitourinary tract cancers, liver cancers, bone cancers, nervous system cancers, gynecological cancers, and skin cancers.

Exemplary hematological cancers include lymphomas and leukemias such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, Non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular), Hodgkin lymphoma, myeloproliferative diseases (e.g., primary myelofibrosis (PMF), polycythemia vera (PV), essential thrombocytosis (ET), 8p11 myeloproliferative syndrome, myelodysplasia syndrome (MDS), T-cell acute lymphoblastic lymphoma (T-ALL), multiple myeloma, cutaneous T-cell lymphoma, adult T-cell leukemia, Waldenstrom's Macroglubulinemia, hairy cell lymphoma, marginal zone lymphoma, chronic myelogenic lymphoma and Burkitt's lymphoma.

Exemplary sarcomas include chondrosarcoma, Ewing’s sarcoma, osteosarcoma, rhabdomyosarcoma, angiosarcoma, fibrosarcoma, liposarcoma, myxoma, rhabdomyoma, rhabdosarcoma, fibroma, lipoma, harmatoma, lymphosarcoma, leiomyosarcoma, and teratoma.

Exemplary lung cancers include non-small cell lung cancer (NSCLC), small cell lung cancer, bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, chondromatous hamartoma, mesothelioma, pavicellular and non-pavicellular carcinoma, bronchial adenoma and pleuropulmonary blastoma.

Exemplary gastrointestinal cancers include cancers of the esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (exocrine pancreatic carcinoma, ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colorectal cancer, gall bladder cancer and anal cancer.

Exemplary genitourinary tract cancers include cancers of the kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], renal cell carcinoma), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma) and urothelial carcinoma.

Exemplary liver cancers include hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.

Exemplary bone cancers include, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma, and giant cell tumors

Exemplary nervous system cancers include cancers of the skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, meduoblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma, glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors, neuro-ectodermal tumors), and spinal cord (neurofibroma, meningioma, glioma, sarcoma), neuroblastoma, Lhermitte-Duclos disease and pineal tumors.

Exemplary gynecological cancers include cancers of the breast (ductal carcinoma, lobular carcinoma, breast sarcoma, triple-negative breast cancer, HER2-positive breast cancer, inflammatory breast cancer, papillary carcinoma), uterus (endometrial carcinoma), cervix (cervical carcinoma, pre -tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), and fallopian tubes (carcinoma).

Exemplary skin cancers include melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, Merkel cell skin cancer, moles dysplastic nevi, lipoma, angioma, dermatofibroma, and keloids. Exemplary head and neck cancers include glioblastoma, melanoma, rhabdosarcoma, lymphosarcoma, osteosarcoma, squamous cell carcinomas, adenocarcinomas, oral cancer, laryngeal cancer, nasopharyngeal cancer, nasal and paranasal cancers, thyroid and parathyroid cancers, tumors of the eye, tumors of the lips and mouth and squamous head and neck cancer.

The compounds of the present disclosure can also be useful in the inhibition of tumor metastases.

In addition to oncogenic neoplasms, the compounds of the present disclosure are useful in the treatment of skeletal and chondrocyte disorders including, but not limited to, achrondroplasia, hypochondroplasia, dwarfism, thanatophoric dysplasia (TD) (clinical forms TD I and TD II), Apert syndrome, Crouzon syndrome, Jackson- Weiss syndrome, Beare- Stevenson cutis gyrate syndrome, Pfeiffer syndrome, and craniosynostosis syndromes. In some embodiments, the present disclosure provides a method for treating a patient suffering from a skeletal and chondrocyte disorder.

In some embodiments, compounds described herein can be used to treat Alzheimer’s disease, HIV, or tuberculosis.

As used herein, the term “8p11 myeloproliferative syndrome” is meant to refer to myeloid/lymphoid neoplasms associated with eosinophilia and abnormalities of FGFR1.

As used herein, the term “cell” is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal.

As used herein, the term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, “contacting” KRAS with a compound described herein includes the administration of a compound described herein to an individual or patient, such as a human, having KRAS, as well as, for example, introducing a compound described herein into a sample containing a cellular or purified preparation containing KRAS.

As used herein, the term “individual,” “subject,” or “patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.

As used herein, the phrase “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent such as an amount of any of the solid forms or salts thereof as disclosed herein that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. An appropriate “effective” amount in any individual case may be determined using techniques known to a person skilled in the art.

The phrase “pharmaceutically acceptable” is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, immunogenicity or other problem or complication, commensurate with a reasonable benefit/risk ratio.

As used herein, the phrase “pharmaceutically acceptable carrier or excipient” refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material. Excipients or carriers are generally safe, non-toxic and neither biologically nor otherwise undesirable and include excipients or carriers that are acceptable for veterinary use as well as human pharmaceutical use. In one embodiment, each component is “pharmaceutically acceptable” as defined herein. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, Pa., 2005; Handbook of Pharmaceutical Excipients, 6th ed.; Rowe et al., Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed.; Gibson Ed.; CRC Press LLC: Boca Raton, Fla., 2009.

As used herein, the term “treating” or “treatment” refers to inhibiting a disease; for example, inhibiting a disease, condition, or disorder in an individual who is experiencing or displaying the pathology or symptomology of the disease, condition, or disorder (/.e., arresting further development of the pathology and/or symptomology) or ameliorating the disease; for example, ameliorating a disease, condition, or disorder in an individual who is experiencing or displaying the pathology or symptomology of the disease, condition, or disorder (/.e., reversing the pathology and/or symptomology) such as decreasing the severity of the disease.

The term “prevent,” “preventing,” or “prevention” as used herein, comprises the prevention of at least one symptom associated with or caused by the state, disease or disorder being prevented.

It is appreciated that certain features of the present disclosure, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment (while the embodiments are intended to be combined as if written in multiply dependent form). Conversely, various features of the present disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination. Combination Therapies I. Cancer therapies

Cancer cell growth and survival can be impacted by dysfunction in multiple signaling pathways. Thus, it is useful to combine different enzyme/protein/receptor inhibitors, exhibiting different preferences in the targets which they modulate the activities of, to treat such conditions. Targeting more than one signaling pathway (or more than one biological molecule involved in a given signaling pathway) may reduce the likelihood of drug-resistance arising in a cell population, and/or reduce the toxicity of treatment.

One or more additional pharmaceutical agents such as, for example, chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, immune- oncology agents, metabolic enzyme inhibitors, chemokine receptor inhibitors, and phosphatase inhibitors, as well as targeted therapies such as Bcr-Abl, Flt-3, EGFR, HER2, JAK, c-MET, VEGFR, PDGFR, c-Kit, IGF-1 R, RAF, FAK, and CDK4/6 kinase inhibitors such as, for example, those described in WO 2006/056399 can be used in combination with the compounds of the present disclosure for treatment of CDK2-associated diseases, disorders or conditions. Other agents such as therapeutic antibodies can be used in combination with the compounds of the present disclosure for treatment of CDK2-associated diseases, disorders or conditions. The one or more additional pharmaceutical agents can be administered to a patient simultaneously or sequentially.

In some embodiments, the CDK2 inhibitor is administered or used in combination with a BCL2 inhibitor or a CDK4/6 inhibitor.

The compounds as disclosed herein can be used in combination with one or more other enzyme/protein/receptor inhibitors therapies for the treatment of diseases, such as cancer and other diseases or disorders described herein. Examples of diseases and indications treatable with combination therapies include those as described herein. Examples of cancers include solid tumors and non-solid tumors, such as liquid tumors, blood cancers. Examples of infections include viral infections, bacterial infections, fungus infections or parasite infections. For example, the compounds of the present disclosure can be combined with one or more inhibitors of the following kinases for the treatment of cancer: Akt1 , Akt2, Akt3, BCL2, CDK4/6, TGF-pR, PKA, PKG, PKC, CaM-kinase, phosphorylase kinase, MEKK, ERK, MAPK, mTOR, EGFR, HER2, HER3, HER4, INS-R, IDH2, IGF-1 R, IR- R, PDGFaR, PDGFpR, PI3K (alpha, beta, gamma, delta, and multiple or selective), CSF1R, KIT, FLK-II, KDR/FLK-1, FLK-4, flt-1 , FGFR1, FGFR2, FGFR3, FGFR4, c-Met, PARP, Ron, Sea, TRKA, TRKB, TRKC, TAM kinases (Axl, Mer, Tyro3), FLT3, VEGFR/Flt2, Flt4, EphA1, EphA2, EphA3, EphB2, EphB4, Tie2, Src, Fyn, Lek, Fgr, Btk, Fak, SYK, FRK, JAK, ABL, ALK and B-Raf. In some embodiments, the compounds of the present disclosure can be combined with one or more of the following inhibitors for the treatment of cancer or infections. Non-limiting examples of inhibitors that can be combined with the compounds of the present disclosure for treatment of cancer and infections include an FGFR inhibitor (FGFR1, FGFR2, FGFR3 or FGFR4, e.g., pemigatinib (INCB54828), INCB62079), an EGFR inhibitor (also known as ErB-1 or HER-1; e.g., erlotinib, gefitinib, vandetanib, orsimertinib, cetuximab, necitumumab, or panitumumab), a VEGFR inhibitor or pathway blocker (e.g. bevacizumab, pazopanib, sunitinib, sorafenib, axitinib, regorafenib, ponatinib, cabozantinib, vandetanib, ramucirumab, lenvatinib, ziv-aflibercept), a PARP inhibitor (e.g., olaparib, rucaparib, veliparib or niraparib), a JAK inhibitor (JAK1 and/or JAK2; e.g., ruxolitinib or baricitinib; or JAK1; e.g., itacitinib (INCB39110), INCB052793, or INCB054707), an IDO inhibitor (e.g., epacadostat, NLG919, or BMS-986205, MK7162), an LSD1 inhibitor (e.g., GSK2979552, INCB59872 and INCB60003), a TDO inhibitor, a PI3K-delta inhibitor (e.g., parsaclisib (INCB50465) or INCB50797), a PI3K-gamma inhibitor such as PI3K-gamma selective inhibitor, a Pirn inhibitor (e.g., INCB53914), a CSF1 R inhibitor, a TAM receptor tyrosine kinases (Tyro-3, Axl, and Mer; e.g., INCB081776), an adenosine receptor antagonist (e.g., A2a/A2b receptor antagonist), an HPK1 inhibitor, a chemokine receptor inhibitor (e.g., CCR2 or CCR5 inhibitor), a SHP1/2 phosphatase inhibitor, a histone deacetylase inhibitor (HDAC) such as an HDAC8 inhibitor, an angiogenesis inhibitor, an interleukin receptor inhibitor, bromo and extra terminal family members inhibitors (for example, bromodomain inhibitors or BET inhibitors such as INCB54329 and INCB57643), c- MET inhibitors (e.g., capmatinib), an anti-CD19 antibody (e.g., tafasitamab), an ALK2 inhibitor (e.g., INCB00928); or combinations thereof.

In some embodiments, the compound or salt described herein is administered with a PI3K5 inhibitor. In some embodiments, the compound or salt described herein is administered with a JAK inhibitor. In some embodiments, the compound or salt described herein is administered with a JAK1 or JAK2 inhibitor (e.g., baricitinib or ruxolitinib). In some embodiments, the compound or salt described herein is administered with a JAK1 inhibitor. In some embodiments, the compound or salt described herein is administered with a JAK1 inhibitor, which is selective over JAK2.

In addition, for treating cancer and other proliferative diseases, compounds described herein can be used in combination with targeted therapies such as, e.g., c-MET inhibitors (e.g., capmatinib), an anti-CD19 antibody (e.g., tafasitamab), an ALK2 inhibitor (e.g., INCB00928); or combinations thereof.

Example antibodies for use in combination therapy include, but are not limited to, trastuzumab (e.g., anti-HER2), ranibizumab (e.g., anti-VEGF-A), bevacizumab (AVASTIN™, e.g., anti-VEGF), panitumumab (e.g., anti-EGFR), cetuximab (e.g., anti-EGFR), rituxan (e.g., anti-CD20), and antibodies directed to c-MET. One or more of the following agents may be used in combination with the compounds of the present disclosure and are presented as a non-limiting list: a cytostatic agent, cisplatin, doxorubicin, taxotere, taxol, etoposide, irinotecan, camptosar, topotecan, paclitaxel, docetaxel, epothilones, tamoxifen, 5-fluorouracil, methotrexate, temozolomide, cyclophosphamide, SCH 66336, R115777, L778.123, BMS 214662, IRESSA™ (gefitinib), TARCEVA™ (erlotinib), antibodies to EGFR, intron, ara-C, adriamycin, cytoxan, gemcitabine, uracil mustard, chlormethine, ifosfamide, melphalan, chlorambucil, pipobroman, triethylenemelamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, floxuridine, cytarabine, 6-mercaptopurine, 6- thioguanine, fludarabine phosphate, oxaliplatin, leucovirin, ELOXATIN™ (oxaliplatin), pentostatine, vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mithramycin, deoxycoformycin, mitomycin-C, L- asparaginase, teniposide 17. alpha. -ethinylestradiol, diethylstilbestrol, testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, testolactone, m eg estro I acetate, methylprednisolone, methyltestosterone, prednisolone, triamcinolone, chlorotrianisene, hydroxyprogesterone, aminoglutethimide, estramustine, medroxyprogesteroneacetate, leuprolide, flutamide, toremifene, goserelin, carboplatin, hydroxyurea, amsacrine, procarbazine, mitotane, mitoxantrone, levamisole, navelbene, anastrazole, letrazole, capecitabine, reloxafine, droloxafine, hexamethylmelamine, avastin, HERCEPTIN™ (trastuzumab), BEXXAR™ (tositumomab), VELCADE™ (bortezomib), ZEVALIN™ (ibritumomab tiuxetan), TRISENOX™ (arsenic trioxide), XELODA™ (capecitabine), vinorelbine, porfimer, ERBITUX™ (cetuximab), thiotepa, altretamine, melphalan, trastuzumab, lerozole, fulvestrant, exemestane, ifosfomide, rituximab, C225 (cetuximab), Campath (alemtuzumab), clofarabine, cladribine, aphidicolon, rituxan, sunitinib, dasatinib, tezacitabine, Sml1 , fludarabine, pentostatin, triapine, didox, trimidox, amidox, 3-AP, and MDL-101 ,731.

The compounds of the present disclosure can further be used in combination with other methods of treating cancers, for example by chemotherapy, irradiation therapy, tumor- targeted therapy, adjuvant therapy, immunotherapy or surgery. Examples of immunotherapy include cytokine treatment (e.g., interferons, GM-CSF, G-CSF, IL-2), CRS-207 immunotherapy, cancer vaccine, monoclonal antibody, bispecific or multi-specific antibody, antibody drug conjugate, adoptive T cell transfer, Toll receptor agonists, RIG-I agonists, oncolytic virotherapy and immunomodulating small molecules, including thalidomide or JAK1/2 inhibitor, PI3K5 inhibitor and the like. The compounds can be administered in combination with one or more anti-cancer drugs, such as a chemotherapeutic agent. Examples of chemotherapeutics include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, baricitinib, bleomycin, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin, dromostanolone propionate, eculizumab, epirubicin, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, fentanyl citrate, filgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, goserelin acetate, histrelin acetate, ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib mesylate, interferon alfa 2a, irinotecan, lapatinib ditosylate, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, meclorethamine, megestrol acetate, melphalan, mercaptopurine, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone phenpropionate, nelarabine, nofetumomab, oxaliplatin, paclitaxel, pamidronate, panitumumab, pegaspargase, pegfilgrastim, pemetrexed disodium, pentostatin, pipobroman, plicamycin, procarbazine, quinacrine, rasburicase, rituximab, ruxolitinib, sorafenib, streptozocin, sunitinib, sunitinib maleate, tamoxifen, temozolomide, teniposide, testolactone, thalidomide, thioguanine, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, vorinostat, and zoledronate.

Additional examples of chemotherapeutics include proteasome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.

Example steroids include corticosteroids such as dexamethasone or prednisone. Example Bcr-Abl inhibitors include imatinib mesylate (GLEEVAC™), nilotinib, dasatinib, bosutinib, and ponatinib, and pharmaceutically acceptable salts. Other example suitable Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521 ,184, WO 04/005281 , and U.S. Ser. No. 60/578,491.

Example suitable Flt-3 inhibitors include midostaurin, lestaurtinib, linifanib, sunitinib, sunitinib, maleate, sorafenib, quizartinib, crenolanib, pacritinib, tandutinib, PLX3397 and ASP2215, and their pharmaceutically acceptable salts. Other example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771 , and WO 04/046120.

Example suitable RAF inhibitors include dabrafenib, sorafenib, and vemurafenib, and their pharmaceutically acceptable salts. Other example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444. Example suitable FAK inhibitors include VS-4718, VS-5095, VS-6062, VS-6063, BI853520, and GSK2256098, and their pharmaceutically acceptable salts. Other example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402.

Example suitable CDK4/6 inhibitors include palbociclib, ribociclib, trilaciclib, lerociclib, and abemaciclib, and their pharmaceutically acceptable salts. Other example suitable CDK4/6 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 09/085185, WO 12/129344, WO 11/101409, WO 03/062236, WO 10/075074, and WO 12/061156.

In some embodiments, the compounds of the disclosure can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.

In some embodiments, the compounds of the disclosure can be used in combination with a chemotherapeutic in the treatment of cancer, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects. In some embodiments, the compounds of the disclosure can be used in combination with a chemotherapeutic provided herein. For example, additional pharmaceutical agents used in the treatment of multiple myeloma, can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib). Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. In some embodiments, the agent is an alkylating agent, a proteasome inhibitor, a corticosteroid, or an immunomodulatory agent. Examples of an alkylating agent include cyclophosphamide (CY), melphalan (MEL), and bendamustine. In some embodiments, the proteasome inhibitor is carfilzomib. In some embodiments, the corticosteroid is dexamethasone (DEX). In some embodiments, the immunomodulatory agent is lenalidomide (LEN) or pomalidomide (POM). Additive or synergistic effects are desirable outcomes of combining a CDK2 inhibitor of the present disclosure with an additional agent.

The agents can be combined with the present compound in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.

The compounds of the present disclosure can be used in combination with one or more other inhibitors or one or more therapies for the treatment of infections. Examples of infections include viral infections, bacterial infections, fungus infections or parasite infections. In some embodiments, a corticosteroid such as dexamethasone is administered to a patient in combination with the compounds of the disclosure where the dexamethasone is administered intermittently as opposed to continuously.

The compounds of Formula (I) or any of the formulas as described herein, a compound as recited in any of the claims and described herein, or salts thereof can be combined with another immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines. Non-limiting examples of tumor vaccines that can be used include peptides of melanoma antigens, such as peptides of gp100, MAGE antigens, Trp-2, MARTI and/or tyrosinase, or tumor cells transfected to express the cytokine GM-CSF.

The compounds of Formula (I) or any of the formulas as described herein, a compound as recited in any of the claims and described herein, or salts thereof can be used in combination with a vaccination protocol for the treatment of cancer. In some embodiments, the tumor cells are transduced to express GM-CSF. In some embodiments, tumor vaccines include the proteins from viruses implicated in human cancers such as Human Papilloma Viruses (HPV), Hepatitis Viruses (HBV and HCV) and Kaposi's Herpes Sarcoma Virus (KHSV). In some embodiments, the compounds of the present disclosure can be used in combination with tumor specific antigen such as heat shock proteins isolated from tumor tissue itself. In some embodiments, the compounds of Formula (I) or any of the formulas as described herein, a compound as recited in any of the claims and described herein, or salts thereof can be combined with dendritic cells immunization to activate potent anti-tumor responses.

The compounds of the present disclosure can be used in combination with bispecific macrocyclic peptides that target Fe alpha or Fe gamma receptor-expressing effectors cells to tumor cells. The compounds of the present disclosure can also be combined with macrocyclic peptides that activate host immune responsiveness.

In some further embodiments, combinations of the compounds of the disclosure with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant. The compounds of the present disclosure can be used in combination with bone marrow transplant for the treatment of a variety of tumors of hematopoietic origin.

The compounds of Formula (I) or any of the formulas as described herein, a compound as recited in any of the claims and described herein, or salts thereof can be used in combination with vaccines, to stimulate the immune response to pathogens, toxins, and self-antigens. Examples of pathogens for which this therapeutic approach may be particularly useful, include pathogens for which there is currently no effective vaccine, or pathogens for which conventional vaccines are less than completely effective. These include, but are not limited to, HIV, Hepatitis (A, B, & C), Influenza, Herpes, Giardia, Malaria, Leishmania, Staphylococcus aureus, Pseudomonas Aeruginosa.

Viruses causing infections treatable by methods of the present disclosure include, but are not limit to human papillomavirus, influenza, hepatitis A, B, C or D viruses, adenovirus, poxvirus, herpes simplex viruses, human cytomegalovirus, severe acute respiratory syndrome virus, Ebola virus, measles virus, herpes virus (e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr virus), flaviviruses, echovirus, rhinovirus, coxsackie virus, cornovirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus and arboviral encephalitis virus.

Pathogenic bacteria causing infections treatable by methods of the disclosure include, but are not limited to, chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, legionella, diphtheria, salmonella, bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lyme's disease bacteria.

Pathogenic fungi causing infections treatable by methods of the disclosure include, but are not limited to, Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.

Pathogenic parasites causing infections treatable by methods of the disclosure include, but are not limited to, Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi, and Nippostrongylus brasiliensis.

When more than one pharmaceutical agent is administered to a patient, they can be administered simultaneously, separately, sequentially, or in combination (e.g., for more than two agents).

Methods for the safe and effective administration of most of these chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, the administration of many of the chemotherapeutic agents is described in the “Physicians' Desk Reference” (PDR, e.g., 1996 edition, Medical Economics Company, Montvale, NJ), the disclosure of which is incorporated herein by reference as if set forth in its entirety. II. Immune-checkpoint therapies

Compounds of the present disclosure can be used in combination with one or more immune checkpoint inhibitors for the treatment of diseases, such as cancer or infections. Exemplary immune checkpoint inhibitors include inhibitors against immune checkpoint molecules such as CBL-B, CD20, CD28, CD40, CD70, CD122, CD96, CD73, CD47, CDK2, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, HPK1 , CD137 (also known as 4-1 BB), ICOS, A2AR, B7-H3, B7-H4, BTLA, CTLA-4, LAG3, TIM3, TLR (TLR7/8), TIGIT, CD112R, VISTA, PD-1 , PD-L1 and PD-L2. In some embodiments, the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, 0X40, GITR and CD137. In some embodiments, the immune checkpoint molecule is an inhibitory checkpoint molecule selected from A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM3, TIGIT, and VISTA. In some embodiments, the compounds provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.

In some embodiments, the compounds provided herein can be used in combination with one or more agonists of immune checkpoint molecules, e.g., 0X40, CD27, GITR, and CD137 (also known as 4-1 BB).

In some embodiments, the inhibitor of an immune checkpoint molecule is anti-PD1 antibody, anti-PD-L1 antibody, or anti-CTLA-4 antibody.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1 or PD-L1 , e.g., an anti-PD-1 or anti-PD-L1 monoclonal antibody. In some embodiments, the anti-PD-1 or anti-PD-L1 antibody is nivolumab, pembrolizumab, atezolizumab, durvalumab, avelumab, cemiplimab, atezolizumab, avelumab, tislelizumab, spartalizumab (PDR001), cetrelimab (JNJ-63723283), toripalimab (JS001), camrelizumab (SHR-1210), sintilimab (IBI308), AB122 (GLS-010), AMP-224, AMP-514/MEDI-0680, BMS936559, JTX-4014, BGB-108, SHR-1210, MEDI4736, FAZ053, BCD-100, KN035, CS1001, BAT1306, LZM009, AK105, HLX10, SHR-1316, CBT-502 (TQB2450), A167 (KL- A167), STI-A101 (ZKAB001), CK-301 , BGB-A333, MSB-2311, HLX20, TSR-042, or LY3300054. In some embodiments, the inhibitor of PD-1 or PD-L1 is one disclosed in U.S. Pat. Nos. 7,488,802, 7,943,743, 8,008,449, 8,168,757, 8,217, 149, or 10,308,644; U.S. Publ. Nos. 2017/0145025, 2017/0174671 , 2017/0174679, 2017/0320875, 2017/0342060, 2017/0362253, 2018/0016260, 2018/0057486, 2018/0177784, 2018/0177870, 2018/0179179, 2018/0179201 , 2018/0179202, 2018/0273519, 2019/0040082, 2019/0062345, 2019/0071439, 2019/0127467, 2019/0144439, 2019/0202824, 2019/0225601, 2019/0300524, or 2019/0345170; or PCT Pub. Nos. WO 03042402, WO 2008156712, WO 2010089411 , WO 2010036959, WO 2011066342, WO 2011159877, WO 2011082400, or WO 2011161699, which are each incorporated herein by reference in their entirety. In some embodiments, the inhibitor of PD-L1 is INCB086550.

In some embodiments, the antibody is an anti-PD-1 antibody, e.g., an anti-PD-1 monoclonal antibody. In some embodiments, the anti-PD-1 antibody is nivolumab, pembrolizumab, cemiplimab, spartalizumab, camrelizumab, cetrelimab, toripalimab, sintilimab, AB122, AMP-224, JTX-4014, BGB-108, BCD-100, BAT1306, LZM009, AK105, HLX10, or TSR-042. In some embodiments, the anti-PD-1 antibody is nivolumab, pembrolizumab, cemiplimab, spartalizumab, camrelizumab, cetrelimab, toripalimab, or sintilimab. In some embodiments, the anti-PD-1 antibody is pembrolizumab. In some embodiments, the anti-PD-1 antibody is nivolumab. In some embodiments, the anti-PD-1 antibody is cemiplimab. In some embodiments, the anti-PD-1 antibody is spartalizumab. In some embodiments, the anti-PD-1 antibody is camrelizumab. In some embodiments, the anti-PD-1 antibody is cetrelimab. In some embodiments, the anti-PD-1 antibody is toripalimab. In some embodiments, the anti-PD-1 antibody is sintilimab. In some embodiments, the anti-PD-1 antibody is AB122. In some embodiments, the anti-PD-1 antibody is AMP-224. In some embodiments, the anti-PD-1 antibody is JTX-4014. In some embodiments, the anti-PD-1 antibody is BGB-108. In some embodiments, the anti-PD-1 antibody is BCD-100. In some embodiments, the anti-PD-1 antibody is BAT1306. In some embodiments, the anti-PD-1 antibody is LZM009. In some embodiments, the anti-PD-1 antibody is AK105. In some embodiments, the anti-PD-1 antibody is HLX10. In some embodiments, the anti-PD-1 antibody is TSR-042. In some embodiments, the anti-PD-1 monoclonal antibody is nivolumab or pembrolizumab. In some embodiments, the anti-PD-1 monoclonal antibody is MGA012 (INCMGA0012; retifanlimab). In some embodiments, the anti-PD1 antibody is SHR-1210. Other anti-cancer agent(s) include antibody therapeutics such as 4-1 BB (e.g., urelumab, utomilumab). In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-L1 monoclonal antibody. In some embodiments, the anti-PD-L1 monoclonal antibody is atezolizumab, avelumab, durvalumab, tislelizumab, BMS-935559, MEDI4736, atezolizumab (MPDL3280A; also known as RG7446), avelumab (MSB0010718C), FAZ053, KN035, CS1001 , SHR-1316, CBT-502, A167, STI-A101 , CK-301, BGB-A333, MSB-2311 , HLX20, or LY3300054. In some embodiments, the anti-PD-L1 antibody is atezolizumab, avelumab, durvalumab, or tislelizumab. In some embodiments, the anti-PD-L1 antibody is atezolizumab. In some embodiments, the anti-PD-L1 antibody is avelumab. In some embodiments, the anti-PD-L1 antibody is durvalumab. In some embodiments, the anti-PD-L1 antibody is tislelizumab. In some embodiments, the anti-PD-L1 antibody is BMS-935559. In some embodiments, the anti-PD-L1 antibody is MEDI4736. In some embodiments, the anti-PD-L1 antibody is FAZ053. In some embodiments, the anti-PD-L1 antibody is KN035. In some embodiments, the anti-PD-L1 antibody is CS1001. In some embodiments, the anti-PD-L1 antibody is SHR- 1316. In some embodiments, the anti-PD-L1 antibody is CBT-502. In some embodiments, the anti-PD-L1 antibody is A167. In some embodiments, the anti-PD-L1 antibody is STI- A101. In some embodiments, the anti-PD-L1 antibody is CK-301. In some embodiments, the anti-PD-L1 antibody is BGB-A333. In some embodiments, the anti-PD-L1 antibody is MSB-2311. In some embodiments, the anti-PD-L1 antibody is HLX20. In some embodiments, the anti-PD-L1 antibody is LY3300054.

In some embodiments, the inhibitor of an immune checkpoint molecule is a small molecule that binds to PD-L1, or a pharmaceutically acceptable salt thereof. In some embodiments, the inhibitor of an immune checkpoint molecule is a small molecule that binds to and internalizes PD-L1, or a pharmaceutically acceptable salt thereof. In some embodiments, the inhibitor of an immune checkpoint molecule is a compound selected from those in US 2018/0179201 , US 2018/0179197, US 2018/0179179, US 2018/0179202, US 2018/0177784, US 2018/0177870, US Ser. No. 16/369,654 (filed Mar. 29, 2019), and US Ser. No. 62/688,164, or a pharmaceutically acceptable salt thereof, each of which is incorporated herein by reference in its entirety.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of KIR, TIGIT, LAIR1 , CD160, 2B4 and TGFR beta.

In some embodiments, the inhibitor is MCLA-145.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody. In some embodiments, the anti-CTLA-4 antibody is ipilimumab, tremelimumab, AGEN1884, or CP-675,206.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody. In some embodiments, the anti-LAG3 antibody is BMS-986016, LAG525, INCAGN2385, or eftilagimod alpha (IMP321).

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CD73. In some embodiments, the inhibitor of CD73 is oleclumab.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of TIGIT. In some embodiments, the inhibitor of TIGIT is OMP-31M32.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of VISTA. In some embodiments, the inhibitor of VISTA is JNJ-61610588 or CA-170.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of B7-H3. In some embodiments, the inhibitor of B7-H3 is enoblituzumab, MGD009, or 8H9.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of KIR. In some embodiments, the inhibitor of KIR is lirilumab or IPH4102.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of A2aR. In some embodiments, the inhibitor of A2aR is CPI-444. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of TGF-beta. In some embodiments, the inhibitor of TGF-beta is trabedersen, galusertinib, or M7824.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PI3K-gamma. In some embodiments, the inhibitor of PI3K-gamma is I PI-549.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CD47. In some embodiments, the inhibitor of CD47 is Hu5F9-G4 or TTI-621.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CD73. In some embodiments, the inhibitor of CD73 is MEDI9447.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CD70. In some embodiments, the inhibitor of CD70 is cusatuzumab or BMS-936561.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of TIM3, e.g., an anti-TIM3 antibody. In some embodiments, the anti-TIM3 antibody is INCAGN2390, MBG453, or TSR-022.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CD20, e.g., an anti-CD20 antibody. In some embodiments, the anti-CD20 antibody is obinutuzumab or rituximab.

In some embodiments, the agonist of an immune checkpoint molecule is an agonist of 0X40, CD27, CD28, GITR, ICOS, CD40, TLR7/8, and CD137 (also known as 4-1 BB).

In some embodiments, the agonist of CD137 is urelumab. In some embodiments, the agonist of CD137 is utomilumab.

In some embodiments, the agonist of an immune checkpoint molecule is an inhibitor of GITR. In some embodiments, the agonist of GITR is TRX518, MK-4166, INCAGN1876, MK-1248, AMG228, BMS-986156, GWN323, MEDI1873, or MEDI6469.ln some embodiments, the agonist of an immune checkpoint molecule is an agonist of 0X40, e.g., 0X40 agonist antibody or OX40L fusion protein. In some embodiments, the anti-OX40 antibody is INCAGN01949, MEDI0562 (tavolimab), MOXR-0916, PF-04518600, GSK3174998, BMS-986178, or 9B12. In some embodiments, the OX40L fusion protein is MEDI6383.

In some embodiments, the agonist of an immune checkpoint molecule is an agonist of CD40. In some embodiments, the agonist of CD40 is CP-870893, ADC-1013, CDX-1140, SEA-CD40, RG7009789, JNJ-64457107, APX-005M, or Chi Lob 7/4.

In some embodiments, the agonist of an immune checkpoint molecule is an agonist of ICOS. In some embodiments, the agonist of ICOS is GSK-3359609, JTX-2011 , or MEDI- 570.

In some embodiments, the agonist of an immune checkpoint molecule is an agonist of CD28. In some embodiments, the agonist of CD28 is theralizumab. In some embodiments, the agonist of an immune checkpoint molecule is an agonist of CD27. In some embodiments, the agonist of CD27 is varlilumab.

In some embodiments, the agonist of an immune checkpoint molecule is an agonist of TLR7/8. In some embodiments, the agonist of TLR7/8 is MEDI9197.

The compounds of the present disclosure can be used in combination with bispecific antibodies. In some embodiments, one of the domains of the bispecific antibody targets PD- 1 , PD-L1, CTLA-4, GITR, 0X40, TIM3, LAG3, CD137, ICOS, CD3 or TGFp receptor. In some embodiments, the bispecific antibody binds to PD-1 and PD-L1. In some embodiments, the bispecific antibody that binds to PD-1 and PD-L1 is MCLA-136. In some embodiments, the bispecific antibody binds to PD-L1 and CTLA-4. In some embodiments, the bispecific antibody that binds to PD-L1 and CTLA-4 is AK104.

In some embodiments, the compounds of the disclosure can be used in combination with one or more metabolic enzyme inhibitors. In some embodiments, the metabolic enzyme inhibitor is an inhibitor of I DO1 , TDO, or arginase. Examples of IDO1 inhibitors include epacadostat, NLG919, BMS-986205, PF-06840003, IOM2983, RG-70099 and LY338196. Inhibitors of arginase inhibitors include INCB1158.

As provided throughout, the additional compounds, inhibitors, agents, etc. can be combined with the present compound in a single or continuous dosage form, or they can be administered simultaneously or sequentially as separate dosage forms.

Formulation, Dosage Forms and Administration

When employed as pharmaceuticals, the compounds of the present disclosure can be administered in the form of pharmaceutical compositions. Thus, the present disclosure provides a composition comprising a compound of Formula I, II, or any of the formulas as described herein, a compound as recited in any of the claims and described herein, or a pharmaceutically acceptable salt thereof, or any of the embodiments thereof, and at least one pharmaceutically acceptable carrier or excipient. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is indicated and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, e.g., by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.

This disclosure also includes pharmaceutical compositions which contain, as the active ingredient, the compound of the present disclosure or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers or excipients. In some embodiments, the composition is suitable for topical administration. In making the compositions of the present disclosure, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, e.g., a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, e.g., up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.

In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.

The compounds provided herein may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. Finely divided (nanoparticulate) preparations of the compounds presented herein can be prepared by processes known in the art see, e.g., WO 2002/000196.

Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the present disclosure can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.

In some embodiments, the pharmaceutical composition comprises silicified microcrystalline cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the silicified microcrystalline cellulose comprises about 98% microcrystalline cellulose and about 2% silicon dioxide w/w.

In some embodiments, the composition is a sustained release composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one component selected from microcrystalline cellulose, lactose monohydrate, hydroxypropyl methylcellulose and polyethylene oxide. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate and hydroxypropyl methylcellulose. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate and polyethylene oxide. In some embodiments, the composition further comprises magnesium stearate or silicon dioxide. In some embodiments, the microcrystalline cellulose is Avicel PH102™. In some embodiments, the lactose monohydrate is Fast-flo 316™. In some embodiments, the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M Premier™) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel K00LV™). In some embodiments, the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Polyox WSR 1105™).

In some embodiments, a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.

The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient. In some embodiments, each dosage contains about 10 mg of the active ingredient. In some embodiments, each dosage contains about 50 mg of the active ingredient. In some embodiments, each dosage contains about 25 mg of the active ingredient. The term “unit dosage forms” refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.

The components used to formulate the pharmaceutical compositions are of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Particularly for human consumption, the composition is preferably manufactured or formulated under Good Manufacturing Practice standards as defined in the applicable regulations of the U.S. Food and Drug Administration. For example, suitable formulations may be sterile and/or substantially isotonic and/or in full compliance with all Good Manufacturing Practice regulations of the U.S. Food and Drug Administration.

The active compound may be effective over a wide dosage range and is generally administered in a therapeutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms and the like.

The therapeutic dosage of a compound of the present disclosure can vary according to, e.g., the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound provided herein in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds disclosed herein can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 pg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.

For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present disclosure. When referring to these preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, e.g., about 0.1 to about 1000 mg of the active ingredient of the present disclosure.

The tablets or pills of the present disclosure can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.

The liquid forms in which the compounds and compositions of the present disclosure can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.

Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.

Topical formulations can contain one or more conventional carriers. In some embodiments, ointments can contain water and one or more hydrophobic carriers selected from, e.g., liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like. Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g., glycerinemonostearate, PEG- glycerinemonostearate and cetylstearyl alcohol. Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, e.g., glycerol, hydroxyethyl cellulose, and the like. In some embodiments, topical formulations contain at least about 0.1 , at least about 0.25, at least about 0.5, at least about 1 , at least about 2 or at least about 5 wt % of the compound disclosed herein. The topical formulations can be suitably packaged in tubes of, e.g., 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.

The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient and the like.

The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11 , more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers or stabilizers will result in the formation of pharmaceutical salts.

The therapeutic dosage of a compound of the present disclosure can vary according to, e.g., the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound provided herein in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the present disclosure can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 pg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from doseresponse curves derived from in vitro or animal model test systems.

Labeled Compounds and Assay Methods

Another aspect of the present disclosure relates to labeled compounds of the disclosure (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating KRAS protein in tissue samples, including human, and for identifying KRAS ligands by inhibition binding of a labeled compound. Substitution of one or more of the atoms of the compounds of the present disclosure can also be useful in generating differentiated ADME (Adsorption, Distribution, Metabolism and Excretion). Accordingly, the present disclosure includes KRAS binding assays that contain such labeled or substituted compounds. The present disclosure further includes isotopically-labeled compounds of the disclosure. An “isotopically” or “radio-labeled” compound is a compound of the disclosure where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present disclosure include but are not limited to 2 H (also written as D for deuterium), 3 H (also written as T for tritium), 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 0, 18 F, 35 S, 36 CI, 82 Br, 75 Br, 76 Br, 77 Br, 123 l, 124 l, 125 l and 131 1. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced by deuterium atoms (e.g., one or more hydrogen atoms of a Ci-6 alkyl group of Formula I, II, or any formulae provided herein can be optionally substituted with deuterium atoms, such as -CDs being substituted for -CHs). In some embodiments, alkyl groups in Formula I, II, or any formulae provided herein can be perdeuterated.

One or more constituent atoms of the compounds presented herein can be replaced or substituted with isotopes of the atoms in natural or non-natural abundance. In some embodiments, the compound includes at least one deuterium atom. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1-2, 1-3, 1-4, 1-5, or 1-6 deuterium atoms. In some embodiments, all of the hydrogen atoms in a compound can be replaced or substituted by deuterium atoms.

Synthetic methods for including isotopes into organic compounds are known in the art (Deuterium Labeling in Organic Chemistry by Alan F. Thomas (New York, N.Y., Appleton- Century-Crofts, 1971 ; The Renaissance of H/D Exchange by Jens Atzrodt, Volker Derdau, Thorsten Fey and Jochen Zimmermann, Angew. Chem. Int. Ed. 2007, 7744-7765; The Organic Chemistry of Isotopic Labelling by James R. Hanson, Royal Society of Chemistry, 2011). Isotopically labeled compounds can be used in various studies such as NMR spectroscopy, metabolism experiments, and/or assays.

Substitution with heavier isotopes, such as deuterium, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances, (see e.g., A. Kerekes et. al. J. Med. Chem. 2011 , 54, 201-210; R. Xu et. al. J. Label Compd. Radiopharm. 2015, 58, 308-312). In particular, substitution at one or more metabolism sites may afford one or more of the therapeutic advantages.

The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro adenosine receptor labeling and competition assays, compounds that incorporate 3 H, 14 C, 82 Br, 125 l, 131 l or 35 S can be useful. For radio-imaging applications 11 C, 18 F, 125 l, 123 l, 124 l, 131 l, 75 Br, 76 Br or 77 Br can be useful. It is understood that a “radio-labeled” or “labeled compound” is a compound that has incorporated at least one radionuclide. In some embodiments, the radionuclide is selected from 3 H, 14 C, 125 l, 35 S and 82 Br.

The present disclosure can further include synthetic methods for incorporating radioisotopes into compounds of the disclosure. Synthetic methods for incorporating radioisotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of disclosure.

A labeled compound of the present disclosure can be used in a screening assay to identify and/or evaluate compounds. For example, a newly synthesized or identified compound (/.e., test compound) which is labeled can be evaluated for its ability to bind a KRAS protein by monitoring its concentration variation when contacting with the KRAS, through tracking of the labeling. For example, a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a KRAS protein (/.e., standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to the KRAS protein directly correlates to its binding affinity. Conversely, in some other screening assays, the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained.

Kits

The present disclosure also includes pharmaceutical kits useful, e.g., in the treatment or prevention of diseases or disorders associated with the activity of KRAS, such as cancer or infections, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, II, or any of the embodiments thereof. Such kits can further include one or more of various conventional pharmaceutical kit components, such as, e.g., containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.

The present disclosure will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the disclosure in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results. The compounds of the Examples have been found to inhibit the activity of KRAS according to at least one assay described herein.

EXAMPLES

Experimental procedures for compounds provided herein are provided below. Preparatory LC-MS purifications of some of the compounds prepared were performed on Waters mass directed fractionation systems. The basic equipment setup, protocols, and control software for the operation of these systems have been described in detail in the literature. See e.g. “Two-Pump At Column Dilution Configuration for Preparative LC-MS”, K. Blom, J. Combi. Chem., 4, 295 (2002); “Optimizing Preparative LC-MS Configurations and Methods for Parallel Synthesis Purification”, K. Blom, R. Sparks, J. Doughty, G. Everlof, T. Haque, A. Combs, J. Combi. Chem., 5, 670 (2003); and “Preparative LC-MS Purification: Improved Compound Specific Method Optimization”, K. Blom, B. Glass, R. Sparks, A. Combs, J. Combi. Chem., 6, 874-883 (2004). The compounds separated were typically subjected to analytical liquid chromatography mass spectrometry (LCMS) for purity check.

The compounds separated were typically subjected to analytical liquid chromatography mass spectrometry (LCMS) for purity check under the following conditions: Instrument; Agilent 1100 series, LC/MSD, Column: Waters Sunfire™ C 5 pm particle size, 2.1 x 5.0 mm, Buffers: mobile phase A: 0.025% TFA in water and mobile phase B: acetonitrile; gradient 2% to 80% of B in 3 minutes with flow rate 2.0 mL/minute.

Some of the compounds prepared were also separated on a preparative scale by reverse-phase high performance liquid chromatography (RP-HPLC) with MS detector or flash chromatography (silica gel) as indicated in the Examples. Typical preparative reversephase high performance liquid chromatography (RP-HPLC) column conditions are as follows: pH = 2 purifications: Waters Sunfire™ C 5 pm particle size, 19 x 100 mm column, eluting with mobile phase A: 0.1% TFA (trifluoroacetic acid) in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature [see “Preparative LCMS Purification: Improved Compound Specific Method Optimization”, K. Blom, B. Glass, R. Sparks, A. Combs, J. Comb. Chem., 6, 874-883 (2004)]. Typically, the flow rate used with the 30 x 100 mm column was 60 mL/minute. pH = 10 purifications: Waters XBridge C 5 pm particle size, 19 x 100 mm column, eluting with mobile phase A: 0.15% NH4OH in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature [See “Preparative LCMS Purification: Improved Compound Specific Method Optimization”, K. Blom, B. Glass, R. Sparks, A. Combs, J. Comb. Chem., 6, 874-883 (2004)]. Typically, the flow rate used with 30 x 100 mm column was 60 mL/minute.”

The following abbreviations may be used herein: AcOH (acetic acid); AC2O (acetic anhydride); aq. (aqueous); atm. (atmosphere(s)); Boc (t-butoxycarbonyl); br (broad); Cbz (carboxybenzyl); calc, (calculated); d (doublet); dd (doublet of doublets); DBU (1 ,8- diazabicyclo[5.4.0]undec-7-ene); DCM (dichloromethane); DIAD (A/, N -diisopropyl azidodicarboxylate); DIEA (N,N-diisopropylethylamine); DIBAL-H (diisobutylaluminium hydride); DMF (A/, /V-dimethylformamide); EtOH (ethanol); EtOAc (ethyl acetate); FCC (flash column chromatography); g (gram(s)); h (hour(s)); HATLI (A/, N, N', /V'-tetramethyl-O-(7- azabenzotriazol-1-yl)uronium hexafluorophosphate); HCI (hydrochloric acid); HPLC (high performance liquid chromatography); Hz (hertz); J (coupling constant); LCMS (liquid chromatography - mass spectrometry); LDA (lithium diisopropylamide); m (multiplet); M (molar); mCPBA (3-chloroperoxybenzoic acid); MS (Mass spectrometry); Me (methyl); MeCN (acetonitrile); MeOH (methanol); mg (milligram(s)); min. (minutes(s)); mL (milliliter(s)); mmol (millimole(s)); N (normal); NCS (N-chlorosuccinimide); NEts (triethylamine); nM (nanomolar); NMP (N-methylpyrrolidinone); NMR (nuclear magnetic resonance spectroscopy); OTf (trifluoromethanesulfonate); Ph (phenyl); pM (picomolar); PPT(precipitate); RP-HPLC (reverse phase high performance liquid chromatography); r.t. (room temperature), s (singlet); t (triplet or tertiary); TBS (tert-butyldimethylsilyl); tert (tertiary); tt (triplet of triplets); TFA (trifluoroacetic acid); THF (tetrahydrofuran); pg (microgram (s)); pL (microliter(s)); pM (micromolar); wt % (weight percent). Brine is saturated aqueous sodium chloride. In vacuo is under vacuum.

The compounds of the present disclosure can be isolated in free-base or pharmaceutical salt form.

Intermediate 1. 2,4,7-trichloro-8-fluoro-3-nitro-1 ,6-naphthyridine

Step 1. 2-chloro-3-fluoro-5-iodopyridin-4-amine p-Toluenesulfonic acid monohydrate (1.1 g, 5.8 mmol) was added to a solution of 2- chloro-3-fluoropyridin-4-amine (17 g, 116 mmol) and NIS (31.3 g, 139 mmol) in acetonitrile (150 ml), then the resulting mixture was stirred at 70 °C for 16 h. The reaction mixture was cooled to room temperature, diluted with EtOAc, and washed subsequently with water, saturated NaHCCh solution, and brine. After drying over sodium sulfate, the organic layer was concentrated in vacuo to give the crude product (31 g, 98% yield), which was used in the next step without further purification. LCMS calculated for C5H4CIFIN2 (M+H) + : m/z = 272.9; found 272.9.

Step 2. methyl 4-amino-6-chloro-5-fluoronicotinate

A mixture of 2-chloro-3-fluoro-5-iodopyridin-4-amine (20 g, 73.4 mmol), Et 3 N (36.8 ml, 264 mmol), and bis(triphenylphosphine)palladium(ll) chloride (5.15 g, 7.34 mmol) in MeOH (250 ml) was degassed with N2 for 10 minutes, then vacuumed and backfilled with CO gas for 3 times. The reaction was stirred at 60 °C for 18 h to reach full conversion. After cooling to room temperature, the desired product crashed out. The crude product was collected by filtration then air dried (14 g, 93% yield), and used in the next step without further purification. LCMS calculated for C7H7CIFN2O2 (M+H) + : m/z = 205.0; found 205.0. Step 3. methyl 6-chloro-4-(3-ethoxy-3-oxopropanamido)-5-fluoronicotinate

A solution of pyridine (11.82 ml, 146 mmol) in 20 mL DCM was added dropwise to a solution of methyl 4-amino-6-chloro-5-fluoronicotinate (23 g, 112 mmol) and ethyl 3-chloro-3- oxopropanoate (18.71 ml, 146 mmol) in DCM (600 mL) at room temperature. The resulting mixture was stirred for 2 h then quenched with water. The organic phase was washed with saturated NaHCCh solution, brine, and dried over sodium sulfate. The solution was concentrated in vacuo, and the crude product was purified by recrystallization in EtOAc and hexanes (25.3 g, 71% yield). LCMS calculated for C12H13CIFN2O5 (M+H) + : m/z = 319.0; found 319.1.

Step 4. ethyl 7-chloro-8-fluoro-2,4-dihydroxy-1,6-naphthyridine-3-carboxyl ate

A solution of sodium ethoxide (32.2 ml, 86 mmol) in EtOH was added dropwise to a suspension of methyl 6-chloro-4-(3-ethoxy-3-oxopropanamido)-5-fluoronicotinate (25 g, 78 mmol) in EtOH (150 ml) at room temperature. The reaction was stirred for 1 h, and the precipitates were collected by filtration and air dried. The crude product (26 g, 116% yield) was used in the next step without further purification. LCMS calculated for C11H9CIFN2O4 (M+H) + : m/z = 287.0; found 287.1.

Step 5. 7-chloro-8-fluoro-1,6-naphthyridine-2,4-diol

A suspension of crude ethyl 7-chloro-8-fluoro-2,4-dihydroxy-1,6-naphthyridine-3- carboxylate (30 g, 105 mmol) in concentrated hydrochloric acid (50 mL, 600 mmol) and 1 ,4- dioxane (250 mL) was refluxed at 100 °C overnight. Upon completion, the reaction mixture was poured into 500 mL ice water, and the precipitates were collected by filtration, washed with ether (3 x 100 mL), then air dried. The crude product (23.8 g, 105% yield) was used in the next step without further purification. LCMS calculated for C8H5CIFN2O2 (M+H) + : m/z = 215.0; found 215.0.

Step 6. 7-chloro-8-fluoro-3-nitro- 1, 6-naphthyridine-2,4-diol

Nitric acid (90% wt, 12 mL, 254 mmol) was added dropwise to a suspension of 7- chloro-8-fluoro-1 ,6-naphthyridine-2,4-diol (9.5 g, 44 mmol) in acetic acid (100 ml) at room temperature. The reaction was stirred for 3h and monitored by LC-MS to avoid over reaction. The desired product crashed out and was collected by filtration, washed with ether (3 x 20 mL) and air dried. The crude product (6.4 g, 56% yield) was used in the next step without further purification. LCMS calculated for C8H4CIFN3O4 (M+H) + : m/z = 260.0; found 260.0 Step 7. 2,4, 7-trichloro-8-fluoro-3-nitro-1,6-naphthyridine

DIPEA (20.9 g, 162 mmol) was added to a suspension of 7-chloro-8-fluoro-3-nitro- 1 ,6-naphthyridine-2,4-diol (14 g, 54 mmol) in POCh (100 ml) at room temperature. The reaction mixture was then stirred at 100 °C for 1 h. After cooling down to room temperature, excess POCh was removed in vacuo then azeotroped with toluene. The residue was dissolved in DCM, washed with saturated NaHCOs solution, dried over sodium sulfate, and then concentrated in vacuo. The crude product was purified by flash chromatography, eluting with a gradient of 0-35% DCM in hexanes to give intermediate 1 as white solid (12.9 g, 81% yield). LCMS calculated for C8H2CI3FN3O2 (M+H) + : m/z = 295.9/297.9; found 295.9/297.9.

Intermediate 2. 2,3,4,6,8-pentachloro-7-nitro-1,5-naphthyridine

Step 1. methyl 3-amino-4, 5, 6-trichloropicolinate

NCS (88 g, 662 mmol) was added to a solution of methyl 3-aminopicolinate (25.2 g, 166 mmol) in DMF (350 mL) and then the reaction was stirred for 16 h. The mixture was cooled with ice water and then water (400 mL) was added and stirred for 20 min, the precipitate was filtered and washed with water, dried under reduced pressure to provide the desired product as a solid (23.6 g, 56% yield). The crude product was used directly for next step without further purification. LCMS calculated for C7H6CI3N2O2 (M+H) + : m/z = 254.9/256.9; found 254.9/256.9.

Step 2. methyl 4,5,6-trichloro-3-(3-methoxy-3-oxopropanamido)picolinate

Methyl 3-chloro-3-oxopropanate (15.1 g, 111 mmol) was added to methyl 3-amino- 4, 5, 6-trichloropicolinate (23.6, 92 mmol) in 1 ,2-dichloroethane (250 mL). The reaction was heated at 85 °C for 2 h. After cooling to room temperature, saturated NaHCCh solution was added and the organic layer separated. The aqueous layer was extracted three times with DCM. The organic layers were combined, dried over Na2SO4 and concentrated. The crude product was used directly for next step without further purification. LCMS calculated for C11H10CI3N2O5 (M+H) + : m/z = 355.0/357.0; found 354.9/356.9.

Step 3. 6, 7,8-trichloro-4-hydroxy-1 ,5-naphthyridin-2(1 H)-one

To a solution of the crude product from step 2 in methanol (300 mL) was added 25%

NaOMe (69.6 g, 322 mmol). The reaction mixture was vigorously stirred for 1 h. The precipitates were collected by filtration, washed with methanol and dried under reduced pressure.

The solids was dissolved in trifluoroacetic acid (200 mL), and 12 N HCI solution (100 mL) was added. The resulting mixture was stirred at 100 °C for 16 h. After removal of volatiles, water (200 mL) was added and the mixture stirred for 0.5 h. The precipitates were filtered and washed with water, dried under reduced pressure to provide the desired product as a solid (21 .0 g, 86%). The crude product was used directly for next step without further purification. LCMS calculated for C8H4CI3N2O2 (M+H) + : m/z = 264.9/266.9; found 264.9/266.9.

Step 4. 6, 7,8-trichloro-4-hydroxy-3-nitro-1 ,5-naphthyridin-2(1 H)-one

Potassium nitrate (12.0 g, 119 mmol) was added to 6,7,8-trichloro-4-hydroxy-1 ,5- naphthyridin-2(1/7)-one (21.0 g, 79 mmol) suspended in sulfuric acid (cone., 60 mL) and the mixture was heated to 50 °C for 6 h. After cooling to ambient temperature, the mixture was slowly poured into a water/ice mixture and stirred vigorously for 1 h. The resulted yellow precipitate was filtered and washed with minimum amount of water, dried under reduced pressure to provide the desired product as a solid (16.5 g, 67%). The crude product was used directly for next step without further purification. LCMS calculated for C8H3CI3N3O4 (M+H) + : m/z = 309.9/311.9; found 309.9/311.9.

Step 5. 2,3,4,6,8-pentachloro-7-nitro-1,5-naphthyridine

DIPEA (27.8 mL, 159 mmol) was added to a mixture of 6,7,8-trichloro-4-hydroxy-3- nitro-1 ,5-naphthyridin-2(1/-/)-one (16.5 g, 53.1 mmol) in POCI3 (99 mL, 1063 mmol) and then the reaction was stirred at 110 °C for 2 h. The solvent was removed under vacuum and then azeotroped with toluene 3 times. The residue was poured into saturated aqueous solution of NaHCOs and stirred vigorously for 0.5 h. The resulted precipitate was filtered, dried under reduced pressure to provide the desired product as a solid. The crude product was used directly for next step without further purification. LCMS calculated for C8HCI5N3O2 (M+H) + : m/z = 345.8/347.8/349.8; found 345.9/347.9/349.9

Intermediate 3. 7-bromo-2,4,6-trichloro-3-nitro-1,5-naphthyridine

Step 1. methyl 3-amino-5-bromo-6-chloropicolinate

NCS (9.36 g, 70.1 mmol) was added to a solution of methyl 3-amino-5- bromopicolinate (16.2 g, 70.1 mmol) in DMF (140 mL) and then the reaction was stirred for 16 h. The mixture was cooled with ice water and then water (200 mL) was added and stirred for 20 min, the precipitate was filtered and washed with water, dried under reduced pressure to provide the desired product as a solid. The crude product was used directly for next step without further purification. LCMS calculated for CyHyBrCIIX C^ (M+H) + : m/z = 264.9/266.9; found 264.9/266.9.

Step 2. methyl 5-bromo-6-chloro-3-(3-methoxy-3-oxopropanamido)picolinate

This compound was prepared according to the procedure described in Intermediate 2 Step 2, replacing methyl 3-amino-4,5,6-trichloropicolinate with methyl 3-amino-5-bromo-6- chloropicolinate. LCMS calculated for CnHioBrCIIXkOs (M+H) + : m/z = 365.0/367.0; found 364.9/366.9.

Step 3. 7-bromo-6-chloro-4-hydroxy-1 ,5-naphthyridin-2(1 H)-one

This compound was prepared according to the procedure described in Intermediate 2 Step 3, replacing methyl 4,5,6-trichloro-3-(3-methoxy-3-oxopropanamido)picolinate with methyl 5-bromo-6-chloro-3-(3-methoxy-3-oxopropanamido)picolinate. LCMS calculated for C8H 5 BrCIN 2 O2 (M+H) + : m/z = 274.9/276.9; found 274.9/276.9.

Step 4. 7-bromo-6-chloro-4-hydroxy-3-nitro- 1, 5-naphthyridin-2( 1H)-one

This compound was prepared according to the procedure described in Intermediate 2 Step 4, replacing 6,7,8-trichloro-4-hydroxy-1 ,5-naphthyridin-2(1/7)-one with 7-bromo-6- chloro-4-hydroxy-1 ,5-naphthyridin-2(1/-/)-one. LCMS calculated for Cst^BrCINsC (M+H) + : m/z = 319.9/321.9; found 319.9/321.9.

Step 5. 7-bromo-2,4,6-trichloro-3-nitro-1,5-naphthyridine

This compound was prepared according to the procedure described in Intermediate 2 Step 5, replacing 6,7,8-trichloro-4-hydroxy-3-nitro-1 ,5-naphthyridin-2(1 /7)-one with 7- bromo-6-chloro-4-hydroxy-3-nitro-1 ,5-naphthyridin-2(1/-/)-one. LCMS calculated for C 8 H 2 BrCI 3 N 2 O 2 (M+H) + : m/z = 355.8/357.8/359.8 ; found 355.9/357.9/359.9.

Intermediate 4. tert-butyl (encfo)-5-((7-chloro-8-fluoro-2-(methylthio)-3-nitro-1,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late

Step 1. tert-butyl (endo)-5-((2, 7-dichloro-8-fluoro-3-nitro-1,6-naphthyridin-4-yl)amino)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

DI PEA (2.503 ml, 14.33 mmol) was added dropwise to a suspension of 2,4,7- trichloro-8-fluoro-3-nitro-1 ,6-naphthyridine (intermediate 1 , 4.72g, 14.33 mmol) and tertbutyl (encfo)-5-amino-2-azabicyclo[2.1.1]hexane-2-carboxylate (2.84 g, 14.33 mmol) in acetonitrile (80 ml) at 0 °C. The mixture was stirred at 0 °C for 1 h, and the precipitates were collected by filtration to give the crude product as yellow solid (5.97 g, 91% yield), which was used in the next step without further purification. LCMS calculated for C HigChFNsC (M+H) + : m/z = 458.1 ; found 458.1.

Step 2. tert-butyl (endo)-5-((7-chloro-8-fluoro-2-(methylthio)-3-nitro-1,6-naph thyridin-4- yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate Sodium thiomethoxide (1.698 g, 24.22 mmol) was added to a suspension of tert-butyl (endo)-5-((2,7-dichloro-8-fluoro-3-nitro-1 ,6-naphthyridin-4-yl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (7.4 g, 16.15 mmol) in MeOH (150 ml) at 0 °C in one portion, the reaction mixture was stirred at 0 °C for 3h. Upon completion, the reaction was poured into water (200 mL), and the precipitates were collected by filtration and air dried, which was used in the next step without further purification. LCMS calculated for C19H22CIFN5O4S (M+H) + : m/z = 470.1 ; found 470.1

Intermediate 5. tert-butyl (encfo)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late

4-Dimethylaminopyridine (0.395 g, 3.23 mmol) was added to a solution of tert-butyl (endo)-5-((7-chloro-8-fluoro-2-(methylthio)-3-nitro-1 ,6-naphthyridin-4-yl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (intermediate 4, 7.8 g, 16.60 mmol), di-tert-butyl dicarbonate (8.8 g, 40.4 mmol), and triethylamine (4.50 ml, 32.3 mmol) in THF (150 ml), and the resulting mixture was stirred at 60 °C for 2h. The reaction was cooled down, concentrated and purified by flash chromatography, eluting with 0-25% EtOAc in hexanes to give the product (6.5 g, 71% yield over two steps). LCMS calculated for C24H29CIFN50eSNa (M+Na) + : m/z = 592.1 ; found 592.1.

Step 2. tert-butyl (endo)-5-((3-amino-7-chloro-8-fluoro-2-(methylthio)-1,6-naph thyridin-4-

A mixture of ammonium chloride (5.91 g, 111 mmol), tert-butyl (encfo)-5-((tert- butoxycarbonyl)(7-chloro-8-fluoro-2-(methylthio)-3-nitro-1 ,6-naphthyridin-4-yl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (6 g, 10.53 mmol), and iron (5.88 g, 105 mmol) in THF (15 ml),MeOH (15.00 ml) and Water (15.00 ml) was stirred at 60 °C for 2h with LCMS monitoring. When the reaction was completed, the mixture was filter through celite, and the filtrate was extracted with EtOAc. The organic layer was washed with brine, dried over sodium sulfate, concentrated to give the crude product (5.7 g, 100% yield), which was used in the next step without further purification. LCMS calculated for C24H32CIFN5O4S (M+H) + : m/z = 540.2; found 540.1.

Step 3. tert-butyl (endo)-5-((tert-butoxycarbonyl)(7-chloro-8-fluoro-3-iodo-2-( methylthio)-1,6-

To a solution of tert-butyl (endo)-5-((3-amino-7-chloro-8-fluoro-2-(methylthio)-1 ,6- naphthyridin-4-yl)(tert-butoxycarbonyl)amino)-2-azabicyclo[2 .1.1]hexane-2-carboxylate (0.5 g, 0.926 mmol) in Acetonitrile (37.5 ml) at -10 °C was added a solution of sulfuric acid (0.123 ml, 2.315 mmol) in Water (0.562 ml), followed by the addition of potassium iodide (0.615 g, 3.70 mmol) in Water (0.562 ml). After stirring for 5 min, a solution of sodium nitrite (0.128 g, 1.852 mmol) in Water (0.562 ml) was slowly added to maintain internal T< 0 °C. The reaction mixture was stirred at -10 °C for 30 minutes, then gradually warmed up to room temperature and continued stirring for another 30 minutes with LCMS monitoring. The reaction mixture was extracted with EtOAc, washed with brine, then purified by flash chromatography (0-25% EtOAc in hexanes) to give the desire product (0.5 g, 83% yield). LCMS calculated for C24H30CIFN4O4S (M+H) + : m/z = 651.1 ; found 651.0.

Step 4. tert-butyl (endo)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1,6-napht hyridin-4- yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

A solution of tert-butyl (encfo)-5-((tert-butoxycarbonyl)(7-chloro-8-fluoro-3-iodo-2- (methylthio)-1 ,6-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate (4 g, 6.1 mmol) in DCM (20 mL) and TFA (20 mL) was stirred at room temperature for 3h, then the mixture was concentrated in vacuo to remove volatiles. The residue was dissolved in THF (40 mL), and di-tert-butyl dicarbonate (2.0 g, 9.2 mmol), and TEA (8.57 ml, 61.5 mmol) were added, followed by addition of 4-dimethylaminopyridine (150 mg, 1.23 mmol). The mixture was stirred at room temperature for 1h with LCMS monitoring. Most of the product crashed out from the reaction mixture and was collected by filtration, the filtrate was concentrated and purified by flash chromatography (0-40% EtOAc in hexanes), yielding intermediate 3 as a white solid (2.9 g, 86% yield). LCMS calculated for C19H22CIFIN4O2S (M+H) + : m/z = 551.0; found 551.0.

Intermediate 6. 2-(3-(Methoxymethoxy)naphthalen-1-yl)-4,4,5,5-tetramethyl-1, 3,2- dioxaborolane

Step 1: 1-bromo-3-(methoxymethoxy) naphthalene

A sample of 4-bromonaphthalen-2-ol (1.57 g, 7.04 mmol) was dissolved in DCM (14 mL) and stirred at room temperature. The solution was treated with N,N- diisopropylethylamine (1.4 mL, 7.8 mmol) and chloromethyl methyl ether (0.6 mL, 7.8 mmol). After 20 min, LCMS indicated complete conversion to the desired product. The reaction was quenched with saturated aq. NH4CI and diluted with DCM. The layers were separated, and the aqueous layer was extracted with additional DCM. The combined organic layers were dried over MgSC , filtered, and concentrated in vacuo. The crude material was dissolved in 50% DCM in hexanes and filtered through a silica plug. The filtrate was concentrated in vacuo to provide 1-bromo-3-(methoxymethoxy)naphthalene (1.76 g, 6.59 mmol, 94% yield). LCMS calculated for CnH 8 BrO 4 (M-MeOH) + : m/z = 235.0, 237.0; found: 235.0, 237.0. Step 2: 2-(3-(Methoxymethoxy)naphthalen- 1-yl)-4,4, 5, 5-tetra methyl- 1, 3, 2-dioxaborolane

A sample of 1-bromo-3-(methoxymethoxy)naphthalene (1.76 g, 6.59 mmol) was dissolved in dioxane (19 mL) and stirred at room temperature. The solution was treated with potassium acetate (1.9 g, 19.8 mmol) and bis(pinacolato)diboron (2.5 g, 9.9 mmol). Finally, [1 ,T-bis(diphenylphosphino)ferrocene]dichloropalladium(ll), DCM complex (0.270 g, 0.329 mmol) was added to the solution, which was then stirred at 100 °C. After 16 hours, LCMS indicated complete conversion to the product. The reaction mixture was diluted with EtOAc, filtered to remove solids, and concentrated in vacuo. The crude material was purified by flash column chromatography (0-40% EtOAc/hexanes) to give 2-(3- (methoxymethoxy)naphthalen-1-yl)-4, 4, 5, 5-tetramethyl-1 , 3, 2-dioxaborolane (1.74 g, 5.53 mmol, 84% yield). LCMS calculated for Ci?H2oB03 (M-MeOH) + : m/z = 283.2; found: 283.1. Intermediate 7. tert-butyl (encfo)-5-(7-chloro-6-fluoro-2-(3-methoxy-3-oxopropyl)-4- (methylthio)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-azab icyclo[2.1.1]hexane-2- carboxylate

A mixture of tert-butyl (endo)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1 ,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (Intermediate 5, 45 mg, 0.082 mmol), DIPEA (143 pl, 0.817 mmol), methyl pent-4-ynoate (27.5 mg, 0.245 mmol), Pd(PPhs)4 (9.4 mg, 0.008 mmol) and copper(l) iodide (3.11 mg, 0.016 mmol) in DMF (1 ml) was stirred at 70 °C for 2h. The reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The crude was purified by flash chromatography to give the desired product (39 mg, 89%). LCMS calculated for C25H29CIFN4O4S (M+H) + : m/z = 535.2; found 535.1.

Step 2. tert-butyl (endo)-5-(7-chloro-6-fluoro-2-(3-methoxy-3-oxopropyl)-4-(met hylthio)-1H- pyrrolo[3,2-c][1 , 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

A solution of tert-butyl (encfo)-5-((7-chloro-8-fluoro-3-(5-methoxy-5-oxopent-1-yn-1- yl)-2-(methylthio)-1 ,6-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate (78 mg, 0.146 mmol), silver tetrafluoroborate (42.6 mg, 0.219 mmol), and (1 ,3-bis(2,6- diisopropylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene)gold(lll) chloride (18.11 mg, 0.029 mmol) in THF (2 ml) was stirred at 70 °C for 2h with LCMS monitoring. The reaction mixture was diluted with EtOAc and filtered through celite, the filtrate was concentrated, and purified by chromatography to give the product (46 mg, 59%). LCMS calculated for C25H29CIFN4O4S (M+H) + : m/z = 535.2; found 535.1.

Intermediate 8. tert-butyl (encfo)-5-(7-chloro-6-fluoro-4-(methylthio)-2-((2- oxooxazolidin-3-yl)methyl)-1 H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate

A mixture of tert-butyl (endo)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (Intermediate 5, 230 mg, 0.42 mmol), DIPEA (0.73 mL, 4.18 mmol), 3-(prop-2-yn-1-yl)oxazolidin-2-one (104 mg, 0.0.84 mmol), Pd(PPhs)4 (48 mg, 0.04 mmol) and copper(l) iodide (16 mg, 0.08 mmol) in DMF (4 ml) was stirred at 70 °C for 2h. The reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The crude was purified by flash chromatography to give the desired product (217 mg, 95%). LCMS calculated for C25H28CIFN5O4S (M+H) + : m/z = 548.2; found 548.1.

Step 2. tert-butyl (endo)-5-(7-chloro-6-fluoro-4-(methylthio)-2-((2-oxooxazolid in-3-yl)methyl)- 1H-pyrrolo[3,2-c][1, 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

A solution of tert-butyl (endo)-5-((7-chloro-8-fluoro-2-(methylthio)-3-(3-(2- oxooxazolidin-3-yl)prop-1-yn-1-yl)-1,6-naphthyridin-4-yl)ami no)-2-azabicyclo[2.1.1]hexane-2- carboxylate (170 mg, 0.31 mmol), silver tetrafluoroborate (91 mg, 0.46 mmol), and (1,3- bis(2,6-diisopropylphenyl)-1,3-dihydro-2H-imidazol-2-ylidene )gold(lll) chloride (38.5 mg, 0.062 mmol) in THF (2 ml) was stirred at 70 °C for 2h with LCMS monitoring. The reaction mixture was diluted with EtOAc and filtered through celite, the filtrate was concentrated, and purified by flash chromatography (0-10% MeOH in DCM) to give the product (93 mg, 55%). LCMS calculated for C25H28CIFN5O4S (M+H) + : m/z = 548.2; found 548.1.

Intermediate 9. tert-butyl (encfo)-5-(7-chloro-6-fluoro-4-((S)-1-((S)-1-methylpyrrolidi n-2- yl)ethoxy)-2-((2-oxooxazolidin-3-yl)methyl)-1H-pyrrolo[3,2-c ][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate

Step 1. tert-butyl (endo)-5-(7-chloro-6-fluoro-4-(methylsulfinyl)-2-((2-oxooxaz olidin-3- yl)methyl)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-azabic yclo[2. 1. 1]hexane-2-carboxylate m-CPBA (236 mg, 1.37 mmol) was added to a solution of tert-butyl (encfo)-5-(7- chloro-6-fluoro-4-(methylthio)-2-((2-oxooxazolidin-3-yl)meth yl)-1 H-pyrrolo[3,2- c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carbox ylate (intermediate 8, 500 mg, 0.91 mmol) in DCM (10 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCCh aq. solution, dried over MgSC and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C25H28CIFN5O5S (M+H) + : m/z = 564.1; found 564.1.

Step 2. tert- butyl (endo)-5- (7-chloro-6-fluoro-4-( (S)-1-((S)- 1-methylpyrrolidin-2-yl) ethoxy) -2- ((2-oxooxazolidin-3-yl)methyl)-1H-pyrrolo[3,2-c][1,6]naphthy ridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

□HMDS (1M in THF, 1.35 mL, 1.35 mmol) was added dropwise to a solution of the crude product from step 1 (380 mg, 0.67 mmol) and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan- 1-ol (174 mg, 1.35 mmol) in THF (10 mL) at 0 °C, and the reaction was stirred at 0 °C for 30 minutes with LCMS monitoring. Upon completion, the reaction mixture was concentrated in vacuo, and the residue was purified by flash chromatography (0-20% MeOH in DCM) to give the product (231 mg, 55% yield). LCMS calculated for C31H39CIFN6O5 (M+H) + : m/z = 629.3; found 629.2.

Intermediate 10. tert-butyl (enc/o)-5-(7-chloro-6-fluoro-2-methyl-4-(methylthio)-1H- pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hex ane-2-carboxylate

A mixture of tert-butyl (endo)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1 ,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (intermediate 5, 275 mg, 0.5 mmol), tributyl(prop-1-yn-1-yl)stannane (0.30 ml, 1.0 mmol), Pd(PPhs)2Cl2 (32 mg, 0.05 mmol) and CsF (304 mg, 2 mmol) in THF (4 ml) was stirred at 70 °C for 2h. The reaction mixture was filtered through Celite and washed with EtOAc, and the combined filtrate was concentrated in vacuo. The residue was purified by flash chromatography (0-50% EtOAc in hexanes) to give the product (178 mg, 77% yield). LCMS calculated for C22H25CIFN4O2S (M+H) + : m/z = 463.1 ; found 463.1. Step 2. tert-butyl (endo)-5-(7-chloro-6-fluoro-2-methyl-4-(methylthio)-1H-pyrro lo[3,2- c][1, 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

A solution of tert-butyl (endo)-5-((7-chloro-8-fluoro-2-(methylthio)-3-(prop-1-yn-1-y l)- 1 ,6-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate (66 mg, 0.143 mmol), silver tetrafluoroborate (41.6 mg, 0.21 mmol)and (1 ,3-bis(2,6-diisopropylphenyl)-1 ,3-dihydro- 2H-imidazol-2-ylidene)gold(lll) chloride (8.8 mg, 0.014 mmol) in THF (2 ml) was stirred at 70 °C for 2h with LCMS monitoring. The reaction mixture was diluted with EtOAc and filtered through celite, the filtrate was concentrated, and purified by chromatography to give the product as a solid. LCMS calculated for C22H25CIFN4O2S (M+H) + : m/z = 463.1 ; found 463.1. Intermediate 11. tert-butyl (enc/o)-5-(7-chloro-6-fluoro-2-methyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1 -y I )-2- azabicyclo[2.1.1]hexane-2-carboxylate

Step 1. tert-butyl (endo)-5-(7-chloro-6-fluoro-2-methyl-4-(methylsulfinyl)-1H-p yrrolo[3,2- c][1, 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate m-CPBA (123 mg, 0.713 mmol) was added to a solution of tert-butyl (encfo)-5-(6- fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-2-methyl-4-(met hylthio)-1H-pyrrolo[3,2- c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carbox ylate (intermediate 10, 220 mg, 0.475 mmol) in DCM (5 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCCh solution, dried over MgSC and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C22H25CIFN4O3S (M+H) + : m/z = 479.1; found 479.1.

Step 2. tert-butyl (endo)-5-(7-chloro-6-fluoro-2-methyl-4-((S)-1-((S)-1-methylp yrrolidin-2- yl)ethoxy)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-azabic yclo[2. 1. 1]hexane-2-carboxylate

□HMDS (1M in THF, 0.95 mL, 0.95 mmol) was added dropwise to a solution of the crude product from step 1 and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (123 mg, 0.95 mmol) in THF (5 mL) at 0 °C, and the reaction was then warmed up to room temperature and stirred for 30 minutes with LCMS monitoring. The reaction mixture was concentrated in vacuo and the residue was purified by flash chromatography (eluting with 0-15% MeOH in DCM) to give the desired product as an oil (164 mg, 63% yield over two steps). LCMS calculated for C28H36CIFN5O3 (M+H) + : m/z = 544.2; found 544.2.

Intermediate 12. tert-butyl (enc/o)-5-(7-chloro-2-(3-(dimethylamino)-3-oxopropyl)-6- fluoro-4-(methylthio)-1H-imidazo[4,5-c][1,6]naphthyridin-1-y l)-2- azabicyclo[2.1.1]hexane-2-carboxylate

Sodium hydrosulfite (395 mg, 1.92 mmol) in water (1 mL) was added to a suspension of tert-butyl (endo)-5-((7-chloro-8-fluoro-2-(methylthio)-3-nitro-1,6-naph thyridin-4-yl)amino)- 2-azabicyclo[2.1.1]hexane-2-carboxylate (intermediate 4, 300 mg, 0.64 mmol ) and 30% aq. ammonium hydroxide (0.46 mL, 6.4 mmol) in MeOH (5 mL) at room temperature. After 10 min, the reaction was diluted with water and extracted with DCM for 3 times. The combined organic layers were dried Na2SO4, filtered and concentrated to give the crude product (250 mg, 89% yield), which was used in the next step without further purification. LCMS calculated for C19H24CIFN5O2S (M+H) + : m/z = 440.1 ; found 440.2.

Step 2. tert-butyl (endo)-5-(7-chloro-2-(3-(dimethylamino)-3-oxopropyl)-6-fluor o-4- (methylthio)- 1H-imidazo[4,5-c][1, 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2- carboxylate

A solution of tert-butyl (endo)-5-((3-amino-7-chloro-8-fluoro-2-(methylthio)-1 ,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (250 mg, 0.57 mmol) and N,N-dimethyl-4-oxobutanamide (220 mg, 1.7 mmol) in i-PrOH (4 mL) was stirred at 80 °C overnight. The reaction mixture was concentrated and the residue was purified by flash chromatography (0-10% MeOH in DCM) to give the product (273 mg, 87% yield). LCMS calculated for C25H31CIFN6O3S (M+H) + : m/z = 549.2; found 549.2.

Intermediate 13. tert-butyl (2S,4S)-4-amino-2-(cyanomethyl)piperidine-1-carboxylate To a solution of 2.0 M LDA (100 mL, 200 mmol) in anhydrous THF (223 mL) was cooled to -78°C for 1 h, and then fert-butyl acetate (26.9 mL, 200 mmol) was added dropwise with stirring over 20 min. After an additional 40 minutes maintained at -78°C, a solution of ethyl (R)-4-cyano-3-hydroxybutanoate (10.5 g, 66.8 mmol) was added dropwise. The mixture was allowed to stir at -40°C for 4 h, and then an appropriate amount of HCI (2 M) was added to the mixture, keeping pH ~6. During this quench, the temperature of the mixture was maintained at -10°C. Upon completion, the temperature of the mixture was cooled to 0°C. The mixture was extracted with ethyl acetate (3 x 100 mL). The combined organic layer was washed with NaHCOs (100 mL) and brine (100 mL), dried over anhydrous Na2SC>4, and evaporated to provide the material as yellow oil (15.0 g, 99% yield). LCMS calculated for C11H18NO4 (M+H) + : m/z = 228.2; found: 228.2

A solution of tert-butyl (R)-6-cyano-5-hydroxy-3-oxohexanoate (15.0 g, 66.0 mmol) in acetic acid (110 ml) was treated with platinum (IV) oxide hydrate (0.868 g, 3.30 mmol). The Parr bottle was evacuated and backfilled with H2 three times and stirred under a H2 atmosphere (45 psi, recharged 4 times) at 22 °C for 3h. The mixture was filtered through Celite and the filter cake was washed with EtOH. The filtrate was concentrated to yield product with a ~9:1 cis:trans diastereomer ratio.

The residue was dissolved in methanol (100 mL) then Boc-anhydride (15.32 ml, 66.0 mmol), sodium carbonate (13.99 g, 132 mmol) was added. The reaction mixture was stirred at room temperature overnight. The mixture was filtered and concentrated. The residue was purified with silica gel column to give the desired product (11.7 g, 56% yield). LCMS calculated for C ^gNNaOs (M+Na) + : m/z = 338.2; found: 338.2.

To a solution of terf-butyl (2S,4R)-2-(2-(terf-butoxy)-2-oxoethyl)-4-hydroxypiperidine- 1-carboxylate (2.10 g, 6.66 mmol) in DCM (33 ml) at 0 °C was added Ms-CI (0.67 mL, 8.66 mmol), After stirring for 1 h, The reaction was diluted with water and organic layer was separated and dried over Na2SC>4, filtered and concentrated. The resulting residue was dissolved in DMF and sodium azide (1.3 g, 20 mmol) was added and the reaction mixture was heated at 70 °C for 5 h. After cooling to rt, the reaction was diluted with EtOAc and water. The organic layer was separated and dried over Na2SO4, filtered and concentrated. The residue was purified with silica gel column to give the desired product (1.90 g, 84% yield). LCMS calculated for (Product-Boc) C11H21N4O2 (M+H) + : m/z = 241.2; found: 241.2.

To a solution of terf-butyl (2S,4S)-4-azido-2-(2-(terf-butoxy)-2-oxoethyl)piperidine-1- carboxylate (1.9 g, 5.58 mmol) in methanol (27.9 ml) was added 10 % palladium on carbon (0.594 g, 0.558 mmol). The reaction mixture was evacuated under vacuum and refilled with H2, stirred at rt for 2 h. The reaction mixture was filtered through a pad of Celite and washed with methanol. The filtrate was concentrated and used as is (1.5 g, 85% yield). LCMS calculated for C16H31N2O4 (M+H) + : m/z = 315.2; found: 315.2.

Step 5. tert-butyl (2S,4S)-4-(((benzyloxy)carbonyl)amino)-2-(2-(tert-butoxy)-2- oxoethyl)piperidine-1 -carboxylate

Cbz

To a stirred solution of tert-butyl (2S,4S)-4-amino-2-(2-(tert-butoxy)-2- oxoethyl)piperidine-1-carboxylate (5.2 g, 16.54 mmol) in CH2CI2 (80.0 mL) was added N- (benzyloxy-carbonyloxy)succinimide (4.95 g, 19.85 mmol) followed by DIPEA (4.33 mL, 24.81 mmol). The mixture stirred at room temperature for 2 hours, then diluted with water. The mixture was extracted with water and brine. The combined organic layers were dried over MgSC , filtered, concentrated, and used directly in the next step without further purification. LCMS calculated for C24H37N2O6 (M+H) + : m/z = 449.2; found: 449.2.

The concentrated residue from Step 5 was taken up in CH2CI2 (80.0 mL) and TFA (50.0 mL). The mixture was stirred at room temperature overnight then concentrated to dryness and used directly in the next step without further purification. LCMS calculated for C15H21N2O4 (M+H) + : m/z = 293.2; found: 293.2. Step 7. 2-((2S,4S)-4-(((benzyloxy)carbonyl)amino)-1-(tert-butoxycarb onyl)piperidin-2- yl)acetic acid

The concentrated residue from Step 6 was taken up in CH2CI2 (80.0 mL) and triethylamine (23.1 mL, 165 mmol) was added slowly. The mixture was stirred at room temperature for 5 minutes, then Boc-anhydride (4.33 g, 19.85 mmol) was added. The mixture was stirred at room temperature for another 30 minutes. Additional Boc-anhydride was added as necessary. Upon full conversion, the mixture was acidified to pH 4-5 then extracted with EtOAc. The combined organic layers were dried over MgSCU, filtered, concentrated, and used directly in the next step without further purification. LCMS calculated for C20H29N2O6 (M+H) + : m/z = 393.2; found: 393.2.

Step 8. tert-butyl (2S,4S)-2-(2-amino-2-oxoethyl)-4-(((benzyloxy)carbonyl)amino )piperidine- 1-carboxylate

The concentrated residue from Step 7 was taken up in THF (80.0 mL) and DIPEA (8.67 mL, 49.6 mmol). The mixture was cooled to 0 °C, then isobutyl chloroformate (5.43 mL, 41.3 mmol) was added slowly. The mixture was stirred at 0 °C for another 20 minutes before ammonium hydroxide (28% in water, 23.0 mL, 165 mmol) was added to the mixture. After stirring for another 5 minutes, the mixture was diluted with brine and extracted with EtOAc. The combined organic layers were dried over MgSO4, filtered, concentrated, and purified by column chromatography 0-8% MeOH in DCM. LCMS calculated for C20H30N3O5 (M+H) + : m/z = 392.2; found: 392.2.

Step 9. tert-butyl (2S,4S)-4-(((benzyloxy)carbonyl)amino)-2-(cyanomethyl)piperi dine-1- carboxylate

Cbz

The purified product from Step 8 was taken up in THF (80.0 mL) and triethylamine (6.0 mL, 43 mmol). The mixture was cooled to 0 °C, then TFAA (3.5 mL, 24.8 mmol) was added slowly. The mixture was stirred at 0 °C for another 30 minutes before diluting with EtOAc and extracting with brine. The combined organic layers were dried over MgSO4, filtered, concentrated to dryness, and purified on silica gel to yield the desired product (5.12 g, 83% over 5 steps). LCMS calculated for C16H20N3O4 (M+H-tert-butyl) + : m/z = 318.1 ; found: 318.1. Step 10. tert-butyl (2S,4S)-4-amino-2-(cyanomethyl)piperidine-1-carboxylate

A round-bottom flask containing a stir bar was charged with terf-butyl (2S,4S)-4- (((benzyloxy)carbonyl)amino)-2-(cyanomethyl)piperidine-1-car boxylate (5.12 g, 13.71 mmol), palladium on carbon (10 wt%, 2.92 g, 2.74 mmol), and MeOH (45 mL). The round-bottom was evacuated and backfilled with H2 (this process was repeated a total of three times) and the mixture was stirred vigorously at room temperature for 1.5 hours with a balloon of H2 attached. The mixture was then filtered over celite and the solids were washed with EtOAc. The filtrate was concentrated and used directly in the next step without further purification. LCMS calculated for C12H22N3O2 (M+H) + : m/z = 240.1; found: 318.1.

Intermediate 14. tert-butyl (2S,4S)-4-(7-chloro-6-fluoro-4-(methylthio)-1H- [1, 2, 3]triazolo[4,5-c][1,6]naphthyridin-1-yl)-2-(cyanomethyl)pipe ridine-1 -carboxylate

Step 1. tert-butyl (2S,4S)-2-(cyanomethyl)-4-((2, 7-dichloro-8-fluoro-3-nitro-1,6-naphthyridin- 4-yl) amino) piperidine- 1 -carboxylate

DIPEA (0.14 ml, 0.79 mmol) was added to a suspension of 2,4,7-trichloro-8-fluoro-3- nitro-1,6-naphthyridine (intermediate 1 , 223 mg, 0.75 mmol) and tert-butyl (2S,4S)-4-amino- 2-(cyanomethyl)piperidine-1-carboxylate (intermediate 13, 180 mg, 0.75 mmol) in DCM (4 ml). The mixture was stirred at room temperature for 1 h, then the reaction mixture was concentrated in vacuo. The residue was purified by flash chromatography (0-50% EtOAc in hexanes) to give the product (220 mg, 58.6% yield). LCMS calculated for C20H22CI2FN6O4 (M+H) + : m/z = 499.1; found 499.1.

Step 2. tert-butyl (2S,4S)-4-((3-amino-7-chloro-8-fluoro-2-(methylthio)-1,6-nap hthyridin-4- yl)amino)-2-(cyanomethyl)piperidine-1 -carboxylate

Sodium thiomethoxide (46 mg, 0.66 mmol) was added to a suspension of tert-butyl (2S,4S)-2-(cyanomethyl)-4-((2,7-dichloro-8-fluoro-3-nitro-1 ,6-naphthyridin-4- yl)amino)piperidine-1-carboxylate (220 mg, 0.44 mmol) in MeOH (4 ml) at 0 °C in one portion, the reaction mixture was stirred at 0 °C for 1 h. After LCMS indicated full conversion, 30% aq. ammonium hydroxide (0.32 mL, 4.4 mmol) was added, followed by the addition of sodium hydrosulfite (272 mg, 1.32 mmol) in water (1 mL), and the resulting mixture was stirred for another 10 minutes. The reaction was diluted with water, extracted with DCM for 3 times, and the combined organic layers were dried Na2SO4, filtered and concentrated to give the crude product (168 mg, 79% yield), which was used in the next step without further purifications. LCMS calculated for C21H27CIFN6O2S (M+H) + : m/z = 481.2; found 481.2. Step 3. tert-butyl (2S,4S)-4-(7-chloro-6-fluoro-4-(methylthio)-1H-[1,2,3]triazo lo[4,5- c][1, 6]naphthyridin- 1-yl)-2-(cyanomethyl) piperidine- 1 -carboxylate

Sodium nitrite (21.52 mg, 0.312 mmol) was added to a solution of tert-butyl (2S,4S)- 4-((3-amino-7-chloro-8-fluoro-2-(methylthio)-1 ,6-naphthyridin-4-yl)amino)-2- (cyanomethyl)piperidine-l-carboxylate (100 mg, 0.208 mmol) in acetic acid (1.5 mL) at 0 °C, and the reaction was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed subsequently with 30% aq. ammonium hydroxide and brine, the organic layer was dried over Na2SO4, concentrated and purified by flash chromatography to give the product (100 mg, 98% yield). LCMS calculated for C21H24CIFN7O2S (M+H) + : m/z = 492.2; found 492.2

Intermediate 15. tert-Butyl (7/?,4/?,5S)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1,6 - naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late

This compound was prepared according to the procedure described for intermediate

5 by using chiral amine tert-butyl (7 ?,4 ?,5S)-5-amino-2-azabicyclo[2.1.1]hexane-2- carboxylate. LCMS calculated for C19H21CIFIN4O2S (M+H) + : m/z = 551.0; found 551 Intermediate 16. tert-butyl (7/?,4/?,5S)-5-(7-chloro-6-fluoro-2-methyl-4-(methylthio)-1H - pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hex ane-2-carboxylate

This compound was prepared according to the procedure described for intermediate 10 by using chiral amine tert-butyl (7R,4R,5S)-5-amino-2-azabicyclo[2.1.1]hexane-2- carboxylate. LCMS calculated for C22H25CIFN4O2S (M+H) + : m/z = 463.1; found 463.1.

Intermediate 17. (/?)-1-(2-Ethynylpyrrolidin-1-yl)ethan-1-one

Acetic anhydride (1.72 ml, 18.2 mmol) was added dropwise to a solution of (R)-2- ethynylpyrrolidine hydrochloride (2 g, 15.2 mmol) and triethylamine (4.66 ml, 33.4 mmol) in DCM (20 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was quenched with water and extracted with ethyl acetate. The organic layer was washed with 1 N HCI, 1 N NaOH, water and brine, dried over sodium sulfate and concentrated. The crude product was used in the next step without further purification. LCMS calculated for C 8 HI 2 NO (M+H)+: m/z = 138.2; found 138.2

Intermediate 18. tert-Butyl (7/?,4/?,5S)-5-(2-((/?)-1-acetylpyrrolidin-2-yl)-7-chloro-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrr olo[3,2-c][1,6]naphthyridin- 1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylate A mixture of tert-butyl (7F?,4F?,5S)-5-((7-chloro-8-fluoro-3-iodo-2-(methylthio)-1 ,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (intermediate 15, 450 mg, 0.82 mmol), DIPEA (1.43 ml, 8.2 mmol), (F?)-1-(2-ethynylpyrrolidin-1-yl)ethan-1-one (intermediate 17, 224 mg, 1.64 mmol), Pd(PPhs)4 (190 mg, 0.16 mmol) and copper(l) iodide (62 mg, 0.33 mmol) in DMF (10 ml) was stirred at 70 °C for 2h. The reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The crude was purified by flash chromatography (eluting with 0-10% MeOH in DCM) to give the desired product (333 mg, 73%). LCMS calculated for C27H32CIFN5O3S (M+H) + : m/z = 560.2; found 560.1.

Step 2. tert-Butyl (1R,4R,5S)-5-(2-((R)-1-acetylpyrrolidin-2-yl)-7-chloro-6-flu oro-4- (methylthio)- 1H-pyrrolo[3, 2-c][ 1, 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

A mixture of tert-butyl (7F?,4F?,5S)-5-(2-((F?)-1-acetylpyrrolidin-2-yl)-7-chloro-6- fluoro- 4-(methylthio)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (333 mg, 0.60 mmol) and CS2CO3 (581 mg, 1.78 mmol) in DMF (10 ml) was stirred at 100 °C for 1h. After cooled down to room temperature, the reaction mixture was diluted with ethyl acetate, washed subsequently with water (3 times) and brine. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to give the crude product (313 mg, 94% yield), which was used in the next step without further purification. LCMS calculated for C27H32CIFN5O3S (M+H) + : m/z = 560.2; found 560.1.

Step 3. tert-butyl (1R,4R,5S)-5-(2-((R)-1-acetylpyrrolidin-2-yl)-7-chloro-6-flu oro-4- (methylsulfinyl)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2. 1. 1]hexane-2- carboxylate m-CPBA (145 mg, 0.84 mmol) was added to a solution of tert-butyl (1R,4R,5S)-5-(2- ((F?)-1-acetylpyrrolidin-2-yl)-7-chloro-6-fluoro-4-(methylth io)-1 H-pyrrolo[3,2- c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (313 mg, 0.56 mmol) in DCM (5 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCCh solution, dried over anhydrous MgSC and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C27H32CIFN5O4S (M+H) + : m/z = 576.2; found 576.1.

Step 4. tert-Butyl (1R,4R,5S)-5-(2-((R)-1-acetylpyrrolidin-2-yl)-7-chloro-6-flu oro-4-((S)-1-((S)- 1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrrolo[3,2-c][1,6]naphth yridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

□HMDS (1M in THF, 1.68 ml, 1.68 mmol) was added dropwise to a solution of the crude product from step 3 and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (217 mg, 1.68 mmol) in THF (8 ml) at 0 °C, and the reaction was then warmed up to room temperature and stirred for 30 minutes with LCMS monitoring. Upon completion, the reaction mixture was concentrated in vacuo and the residue was purified by flash chromatography (eluting with 0-20% MeOH in DCM) to give the desired product (283 mg, 79% yield over two steps). LCMS calculated for C33H43CIFN6O4 (M+H) + : m/z = 641.3; found 641.3.

Intermediate 19. tert-Butyl (7/?,4/?,5S)-5-((7-(chroman-8-yl)-8-fluoro-3-iodo-2-((S)-1-( (S)- 1-methylpyrrolidin-2-yl)ethoxy)-1,6-naphthyridin-4-yl)amino) -2- azabicyclo[2.1.1]hexane-2-carboxylate

Step 1. tert-butyl ( 1R,4R,5S)-5-((3-amino-7-chloro-8-fluoro-2-((S)-1-((S)-1-meth ylpyrrolidin-2- yl)ethoxy)-1,6-naphthyridin-4-yl)(tert-butoxycarbonyl)amino) -2-azabicyclo[2. 1. 1]hexane-2- carboxylate

To a mixture of 2,4,7-trichloro-8-fluoro-3-nitro-1,6-naphthyridine (Intermediate 1 , 500 mg, 1.687 mmol) in THF (8.43 ml) was added tert-butyl (1 ?,4 ?,5S)-5-amino-2- azabicyclo[2.1.1]hexane-2-carboxylate (334 mg, 1.687 mmol) and DI PEA (309 pl, 1.771 mmol) in THF dropwise. The reaction was stirred at 0 °C for 15 min. Then, (S)-1-((S)-1- methylpyrrolidin-2-yl)ethan-1-ol (283 mg, 2.193 mmol) and potassium tert-butoxide (1.0 M in THF) (5060 pl, 5.06 mmol) were added. After stirring at 0 °C for another 30 min, water was added to the reaction mixture followed by extraction with ethyl acetate. The combined organic layers were dried over sodium sulfate, filtered and concentrated. The crude residue was dissolved into THF (8.43 ml), and boc-anhydride (587 pl, 2.53 mmol), followed by TEA (470 pl, 3.37 mmol) and DMAP (41.2 mg, 0.337 mmol) were added. The reaction mixture was heated to 60 °C for 2 hours. Then HOAc (5 mL) and iron (942 mg, 16.87 mmol) were added to the reaction mixture, and heating continued at 60 °C for 30 minutes. Water was added to the reaction mixture followed by extraction with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. The crude product (681 mg, 65% yield) was used in the next step directly. LCMS calculated for C30H43CIFN6O5 (M+H) + : m/z = 621.3; found 621.3.

Step 2. tert-butyl (1R,4R,5S)-5-((3-amino-7-(chroman-8-yl)-8-fluoro-2-((S)-1-(( S)-1- methylpyrrolidin-2-yl)ethoxy)-1, 6-naphthyridin-4-yl) (tert-butoxycarbonyl)amino)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

A vial was charged with terf-butyl 5-((3-amino-7-chloro-8-fluoro-2-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1 ,6-naphthyridin-4-yl)(tert-butoxycarbonyl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (4.15 g, 6.68 mmol), chroman-8-ylboronic acid (1.308 g, 7.35 mmol), chloro(2-dicyclohexylphosphino-2',4',6'-triisopropyl-1 ,T-biphenyl)[2-(2'-amino- 1 ,1 '-biphenyl)]palladium(ll) (0.526 g, 0.668 mmol), potassium phosphate tribasic (2.84 g, 13.36 mmol) and 5:1 dioxane/water mixture (22 ml). The reaction mixture was heated to 100 °C for 3 hours. Water was added to the reaction mixture followed by extraction with dichloromethane. The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated. The crude residue was purified by automated flash column chromatography (0-100% DCM/EtOAc) to afford the product (3.43 g, 71% yield). LCMS calculated for C39H52FN6O6 (M+H) + : m/z = 719.4; found 719.4.

Step 3. tert-butyl (1R,4R,5S)-5-((tert-butoxycarbonyl)(7-(chroman-8-yl)-8-fluor o-3-iodo-2- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1,6-naphthyridin -4-yl)amino)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

To a solution of terf-butyl (1R,4R,5S)-5-((3-amino-7-(chroman-8-yl)-8-fluoro-2-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1,6-naphthyridin-4-yl)( tert-butoxycarbonyl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (3.43 g, 4.77 mmol) in acetonitrile (47.7 ml) was added 40% sulfuric acid (2.54 ml, 19.09 mmol) and potassium iodide (3.17 g, 19.09 mmol) at 0 °C. An aqueous solution of sodium nitrite (0.658 g, 9.54 mmol) was added slowly. After 1 h, saturated aqueous sodium thiosulfate and sodium bicarbonate solutions were added to the reaction mixture followed by extraction with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered and concentrated. The crude product was purified by automated flash column chromatography (0-10% MeOH in DCM) to afford the desired product (1 g, 25% yield). LCMS calculated for C39H50FIN5O6 (M+H) + : m/z = 830.3; found 830.2.

Step 4. tert-Butyl (1R,4R,5S)-5-((7-(chroman-8-yl)-8-fluoro-3-iodo-2-((S)-1-((S )-1- methylpyrrolidin-2-yl) ethoxy) - 1, 6-naphthyridin-4-yl) amino)-2-azabicyclo[2. 1. 1 ]hexane-2- carboxylate

A vial was charged with terf-butyl (1R,4R,5S)-5-((terf-butoxycarbonyl)(7-(chroman-8- yl)-8-fluoro-3-iodo-2-((S)-1-((S)-1-methylpyrrolidin-2-yl)et hoxy)-1,6-naphthyridin-4-yl)amino)- 2-azabicyclo[2.1.1]hexane-2-carboxylate (1 g, 1.21 mmol), DCM (5 ml), and TFA (5 ml). The reaction mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated and the crude residue was charged with THF (5 ml), TEA (1.7 ml, 12 mmol), and boc-anhydride (0.284 g, 1.3 mmol). After 30 minutes, saturated aqueous sodium bicarbonate solution was added to the reaction mixture followed by extraction with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered and concentrated. The crude material was purified by automated flash column chromatography (0-10% MeOH in DCM) to afford the desired product (0.698 g, 79% yield). LCMS calculated for C34H42FIN5O4 (M+H) + : m/z = 730.2; found 730.2 Intermediate 20: 3-ethynyl-2,3-dihydroindolizin-5(1H)-one

Step 1 : N-methoxy-N-methyl-5-oxo- 1, 2, 3, 5-tetrahydroindolizine-3-carboxamide

A vial was charged with commercially available 5-oxo-1 ,2,3,5-tetrahydroindolizine-3- carboxylic acid (250 mg, 1.395 mmol), /V,O-dimethylhydroxylamine hydrochloride (204 mg, 2.093 mmol), DMF (7 ml), and DIEA (0.675 ml, 3.86 mmol). The reaction mixture was stirred at room temperature for 5 minutes, then HATLI (562 mg, 1.479 mmol) was added. The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was quenched with water and extracted into DCM. The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated. The crude residue was purified by automated flash column chromatography (0-20% MeOH in DCM) to afford the product as a white crystalline solid (0.249 g, 80% yield). LCMS calculated for C11H15N2O3 (M+H) + : m/z = 223.1 ; found 223.1.

Step 2: 5-oxo-1,2,3,5-tetrahydroindolizine-3-carbaldehyde

To a cooled (-45 °C) tetrahydrofuran (11.2 mL) solution of /V-methoxy-/V-methyl-5- oxo-1 ,2,3,5-tetrahydroindolizine-3-carboxamide (0.25 g) was added a 1 M solution of lithium aluminum hydride in tetrahydrofuran (1.34 mL) over 10 minutes. The reaction mixture was stirred at -45 °C for 30 minutes, 0 °C for 90 minutes then cooled to -45 °C, and a solution of potassium hydrogen sulfate (0.305 g) in water (1.0 mL) was added. The mixture was warmed to room temperature, filtered, and concentrated. The crude material was taken to the next step without further purification (0.215 g, 118% yield). LCMS calculated for C9H10NO2 (M+H) + : m/z = 164.1 ; found 164.0.

Step 3: 3-ethynyl-2,3-dihydroindolizin-5(1H)-one

To a cooled (0 °C) solution of 5-oxo-1 ,2,3,5-tetrahydroindolizine-3-carbaldehyde (0.215 g, 1.32 mmol) in methanol (6.6 ml) was added potassium carbonate (0.364 g, 2.64 mmol) followed directly by dimethyl (1-diazo-2-oxopropyl)phosphonate (0.217 ml, 1.45 mmol). The reaction solution was stirred at 0 °C for 2 hours. The reaction was quenched with water and extracted with DCM. The combined organic fractions were dried over magnesium sulfate then concentrated. The crude material was purified by automated flash column chromatography (0-20% MeOH in DCM) to afford the desired product (0.114 g, 54% yield). LCMS calculated for C10H10NO (M+H) + : m/z = 160.1 ; found 160.0 Intermediate 21 : (/?)-1-(but-3-yn-2-yl)pyridin-2(1H)-one

Step 1. (S)-but-3-yn-2-yl methanesulfonate

To a stirred DCM solution (100 mL) containing (S)-but-3-yn-2-ol (3.79 g, 54.1 mmol) cooled to 0 °C was added /V,/V-diisopropylethylamine (18.9 mL, 108 mmol) and methanesulfonyl chloride (4.2 mL, 54.1 mmol) slowly. The reaction was allowed to warm up to ambient temperature. After stirring for 1 h, the reaction was quenched with water and extracted with ethyl acetate. The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was used directly for next step. LCMS calculated for C5H9O3S (M+H) + : m/z = 149.0; found 149.0 Step 2. (R)-1-(but-3-yn-2-yl)pyridin-2(1H)-one

To a stirred THF solution (180 mL) containing pyridin-2(1 H)-one (5.20 g, 54.1 mmol) was added potassium tert-butoxide (6.1 g, 54.1 mmol) slowly. After stirring for 0.5 h, a THF solution containing (S)-but-3-yn-2-yl methanesulfonate was added. The slurry was stirred at 60 °C for 24 hours and then quenched with water. The mixture was extracted with ethyl acetate. The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by flash column chromatography (0-80% ethyl acetate in hexanes) to provide the desired product. LC-MS calculated for C9H10NO (M+H) + : m/z = 148.1 ; found 148.1.

Intermediate 22. tert-Butyl (7/?,4/?,5S)-5-(7-chloro-6-fluoro-2-((/?)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylthio)-1 H-pyrrolo[3,2-c][1,6]naphthyridin-1- yl)-2-azabicyclo[2.1.1 ]hexane-2-carboxylate

To the reaction mixture of (F?)-2-ethynylpyrrolidine hydrochloride (2.63 g, 20 mmol) in

DCM (50.0 mL) at 0 °C was added DIPEA (10.48 mL, 60.0 mmol) followed by the addition of methyl carbonochloridate (2.318 ml, 30.0 mmol). The reaction mixture was then stirred at room temperature for 1 hour. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N NaOH aqueous solution, 1 N HCI aqueous solution, water and brine, dried over MgSC and concentrated. The product was used in the next step directly without further purification. LC-MS calculated for C8H12NO2 (M+H) + : m/z = 154.1 ; found 154.1.

Step 2. tert-butyl (1R,4R,5S)-5-((7-chloro-8-fluoro-3-(((R)-1-(methoxycarbonyl) pyrrolidin-2- yl)ethynyl)-2-(methylthio)- 1, 6-naphthyridin-4-yl)amino)-2-azabicyclo[2. 1. 1]hexane-2- carboxylate

Under the atmosphere of nitrogen, the reaction mixture of ferf-butyl (7F?,4F?,5S)-5-((7- chloro-8-fluoro-3-iodo-2-(methylthio)-1 ,6-naphthyridin-4-yl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (Intermediate 15, 1050 mg, 1.906 mmol), methyl (F?)-2-ethynylpyrrolidine-1-carboxylate (664 pL, 4.77 mmol), copper(l) iodide (145 mg, 0.763 mmol), tetrakis(triphenylphosphine)palladium (441 mg, 0.381 mmol) and DI PEA (999 pL, 5.72 mmol) was stirred at 75 °C in DMF (9.53 mL) for 2 hours. The reaction mixture was poured into water, extracted with DCM. The organic layers was dried over anhydrous MgSC , concentrated and purified by flash chromatography (0-100% EtOAc in Hexanes) to afford the product (600 mg, 74% yield). LCMS calculated for C27H32CIFN5O4S (M+H) + : m/z = 576.2; found 576.2.

Step 3. tert-butyl ( 1R,4R,5S)-5-(7-chloro-6-fluoro-2-((R)-1-(methoxycarbonyl)pyr rolidin-2-yl)- 4-(methylthio)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-az abicyclo[2. 1. 1]hexane-2- carboxylate

To a solution of terf-butyl 5-((7-chloro-8-fluoro-3-(((F?)-1-(methoxycarbonyl)pyrrolidin - 2-yl)ethynyl)-2-(methylthio)-1 ,6-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2- carboxylate (600 mg, 1.042 mmol) in DMF (5.21 mL) was added CS2CO3 (1018 mg, 3.12 mmol). The reaction mixture was stirred at 100 °C for 1 hour. The reaction mixture was then cooled down, poured into ice water. The precipitated product was filtered and dried to afford product (500 mg, 83% yield). LCMS calculated for C27H32CIFN5O4S (M+H) + : m/z = 576.2; found 576.2.

Intermediate 23. tert-Butyl (7/?,4/?,5S)-5-(7-(2,3-dichlorophenyl)-6-fluoro-2-((/?)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylsulfinyl)-1H-pyrr olo[3,2- c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carbox ylate

Step 1. tert-butyl (1R,4R,5S)-5-(7-(2,3-dichlorophenyl)-6-fluoro-2-((R)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylthio)-1H-pyrrolo[ 3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

Under the atmosphere of nitrogen, the reaction mixture of tert-butyl (1R,4R,5S)-5-(7- chloro-6-fluoro-2-((R)-1-(methoxycarbonyl)pyrrolidin-2-yl)-4 -(methylthio)-1H-pyrrolo[3,2- c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carbox ylate (Intermediate 22, 470 mg, 0.816 mmol), tetrakis(triphenylphosphine)palladium (471 mg, 0.408 mmol), (2,3- dichlorophenyl)boronic acid (1245 mg, 6.53 mmol) and K2CO3 (677 mg, 4.90 mmol) in Dioxane (4.53 mL) and Water (0.9 mL) was stirred at 110 °C for 2 hours. Upon completion, the reaction mixture was poured into water, extracted with DCM. The combined organic layers were dried over anhydrous MgSC , concentrated and purified by flash chromatography (0-100% EtOAc in Hexanes) to afford the product (280 mg, 50% yield). LCMS calculated for C33H35CI2FN5O4S (M+H) + : m/z = 686.2; found 686.2. Step 2. tert-butyl (1R,4R,5S)-5-(7-(2,3-dichlorophenyl)-6-fluoro-2-((R)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylsulfinyl)-1H-pyrr olo[3,2-c][1,6]naphthyridin-1-yl)- 2-azabicyclo[2. 1. 1]hexane-2-carboxylate

To a solution of tert-butyl (7R,4/?,5S)-5-(7-(2,3-dichlorophenyl)-6-fluoro-2-((/?)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylthio)-1H-pyrrolo[ 3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (180 mg, 0.262 mmol) in EtOAc (2.62 mL) at 0 °C was added mCPBA (88 mg, 0.393 mmol). The reaction mixture was stirred at 0 °C for 30 minutes. Upon completion, the reaction was quenched by saturated NaHCOs solution. The organic layer was dried over anhydrous MgSO4, filtered and concentrated to afford light yellow solid (170 mg, 92% yield). LCMS calculated for C33H35CI2FN5O5S (M+H) + : m/z = 702.2; found 702.2.

Intermediate 24. (7aS)-5-ethynyltetrahydro-1H,3H-pyrrolo[1,2-c]oxazol-3-one

Step 1. te rt- butyl (5S)-5-(( (tert-butyldiphenylsilyl) oxy) methyl) -2-ethynyl-2-hydroxypyrrolidine- 1-carboxylate

To a solution of TMS acetylene (1.35g, 13.8 mmol) in THF (50 mL), n-BuLi (1.6 M in hexane, 7.9 mL, 12.6 mmol) was added dropwise and the mixture was stirred for 30 min. at - 78 °C. Then a solution of terf-butyl (S)-2-(((terf-butyldiphenylsilyl)oxy)methyl)-5- oxopyrrolidine-1 -carboxylate (5.2 g, 11.5 mmol) in THF (10 mL) was added slowly. The mixture was stirred and warmed up to r.t. slowly with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was used directly for next step without purification. LCMS calculated for C28H 38 NO 4 Si (M+H) + : m/z = 480.2; found 480.2.

Step 2. tert-butyl ((2S)-1-((tert-butyldiphenylsilyl)oxy)-5-hydroxyhept-6-yn-2- yl)carbamate

To a solution of above crude terf-butyl (5S)-5-(((terf-butyldiphenylsilyl)oxy)methyl)-2- ethynyl-2-hydroxypyrrolidine-1-carboxylate (11.5 mmol) in MeOH (50 mL), CeCI 3 »7H 2 O (4.7g, 12.65 mmol) was added at r.t. The slurry was cooled to -45 °C and NaBH 4 (174 mg, 4.6 mmol) was added, additional NaBH 4 (174 mg, 4.6 mmol) was added with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was used directly for next step without purification. LCMS calculated for C2sH 4 oN0 4 Si (M+H) + : m/z = 482.2; found 482.2.

Step 3. tert-butyl (2S)-2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-ethynylpyrro lidine-1- carboxylate To a solution of above crude ferf-butyl ((2S)-1-((ferf-butyldiphenylsilyl)oxy)-5- hydroxyhept-6-yn-2-yl)carbamate (11.5 mmol) in DCM (50 mL), diethylaminosulfur trifluoride (1.8 mL, 13.8 mmol) was added slowly at -78 °C. K2CO3 (2.4g, 17.2 mmol) was added in one portion after 15 mins and the cold bath was removed. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography to give the desired product (2.7 g, 50% over 3 steps). LCMS calculated for C28H3sNO3Si (M+H) + : m/z = 464.2; found 364.2 (-Boc).

Step 4. (7aS)-5-ethynyltetrahydro-1H,3H-pyrrolo[ 1 ,2-c]oxazol-3-one

To a solution of terf-butyl (2S)-2-(((ferf-butyldiphenylsilyl)oxy)methyl)-5- ethynylpyrrolidine-1-carboxylate (930 mg, 2.0 mmol) in 5 mL THF, TBAF(4 mL, 1 M in THF, 4.0 mmol) was added at 0 °C with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was then dissolved in DCM (3 mL) and TFA (3 mL) with LCMS monitoring, upon completion, the mixture was concentrated. The residue was then diluted in DCM (2 mL), TEA (2.8 mL, 20 mmol), DMAP (24 mg, 0.2 mmol), and GDI (480 mg, 3.0 mmol) was added at 0 °C, the reaction mixture was stirred overnight. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography to give the desired product as a mixture of diastereoisomers (60 mg, 20%). LCMS calculated for C 8 HION0 2 (M+H) + : m/z = 152.1 ; found 152.1.

Intermediate 25. 4-ethynyl-/V,/V,1 -trimethyl-1 H-pyrazole-3-carboxamide

To a solution of 4-ethynyl-1-methyl-1/7-pyrazole-3-carboxylic acid (100 mg, 0.666 mmol) in DMF (3.33 mL) were added dimethylamine (866 pL, 0.866 mmol), HATU (279 mg, 0.733 mmol) and Hunig's base (349 pL, 1.998 mmol). The reaction mixture was stirred at room temperature for 1 hour, then quenched with water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated. The crude product was used in following steps without further purification. LCMS calculated for C 9 Hi 2 N 3 O (M+H) + : m/z = 178.1 ; found 178.2.

Example 1a/1 b: methyl 3-(1-((enofo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3- hydroxynaphthalen-1-yl)-4-((S)-1-((S)-1-methylpyrrolidin-2-y l)ethoxy)-1H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)propanoate

Step 1. tert-butyl (endo)-5-(6-fluoro-2-(3-methoxy-3-oxopropyl)-7-(3-

(methoxymethoxy)naphthalen- 1-yl)-4-(methylthio)- 1H-pyrrolo[3, 2-c][ 1, 6]naphthyridin- 1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

A mixture of tert-butyl (encfo)-5-(7-chloro-6-fluoro-2-(3-methoxy-3-oxopropyl)-4- (methylthio)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2-azab icyclo[2.1.1]hexane-2-carboxylate (intermediate 7, 46 mg, 0.086 mmol), 2-(3-(methoxymethoxy)naphthalen-1-yl)-4, 4,5,5- tetramethyl-1,3,2-dioxaborolane (intermediate 6, 54.0 mg, 0.172 mmol), XPhos Pd G2 (6.76 mg, 8.60 pmol), and K3PO4 (54.7 mg, 0.258 mmol) in Dioxane (0.8 ml) and Water (0.160 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by flash chromatography to give the product (46 mg, 78%). LCMS calculated for C37H40FN4O6S (M+H) + : m/z = 687.3; found 687.2. Step 2. tert-butyl (endo)-5-(6-fluoro-2-(3-methoxy-3-oxopropyl)-7-(3- (methoxymethoxy)naphthalen- 1-yl)-4-(methylsulfinyl)-1H-pyrrolo[3, 2-c][ 1, 6]naphthyridin- 1- m-CPBA (17.34 mg, 0.100 mmol) was added to a solution of tert-butyl (encfo)-5-(6- fluoro-2-(3-methoxy-3-oxopropyl)-7-(3-(methoxymethoxy)naphth alen-1-yl)-4-(methylthio)-1 H- pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (46 mg, 0.067 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCOs aq. solution, dried over MgSC and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C37H40FN4O7S (M+H) + : m/z = 703.3; found 703.3.

Step 3. methyl 3-(1-((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-6-fluoro-7-(3-hydroxynaphthalen- 1-yl)-4-((S)-1-((S)-1 -methyl pyrrolidin-2-yl) ethoxy) - 1 H-pyrrolo[ 3, 2-c][ 1, 6]naphthyridin-2- yl)propanoate

□HMDS (1M in THF, 60 pl, 0.06 mmol) was added dropwise to a solution of tert-butyl (encfo)-5-(6-fluoro-2-(3-methoxy-3-oxopropyl)-7-(3-(methoxym ethoxy)naphthalen-1-yl)-4- (methylsulfinyl)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (15 mg, 0.021 mmol) and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (8.1 mg, 0.063 mmol) in THF (1 mL) at 0 °C, and the reaction was stirred at 0 °C for 30 minutes with LCMS monitoring. The reaction mixture was concentrated in vacuo, and the residue was dissolved in TFA (1 mL). The solution was stirred at room temperature for 10 minutes to remove the Boc and MOM protecting groups. The reaction mixture was diluted with methanol, which was purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 1a: Diastereomer 1 , peak 1. LCMS calculated for C36H39FN5O4 (M+H) + : m/z = 624.3; found 624.3.

Example 1b: Diastereomer 2, peak 2. LCMS calculated for C36H39FN5O4 (M+H) + : m/z = 624.3; found 624.3. *Potent peak.

Example 2a/2b. methyl 3-(1-((enc/o)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-4-

(((2/?,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)me thoxy)-7-(3- hydroxynaphthalen-1-yl)-1 H-pyrrolo[3,2-c][1,6]naphthyridin-2-yl)propanoate

Example 2a/2b were prepared according to procedure described in Example 1a/1b, step 3, replacing (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol with ((2R,7aS)-2- fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methanol. Example 2a: Diastereomer 1, peak 1. LCMS calculated for C37H38F2N5O4 (M+H) + : m/z = 654.3; found 654.3.

Example 2b: Diastereomer 2, peak 2. LCMS calculated for C37H38F2N5O4 (M+H) + : m/z = 654.3; found 654.3. * Potent peak. Example 3a/3b. 4-(1-((enofo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-meth yl-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrrolo[3,2-c][1,6]n aphthyridin-7- yl)naphthalen-2-ol

A mixture of tert-butyl (endo)-5-(7-chloro-6-fluoro-2-methyl-4-(methylthio)-1/7- pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (intermediate 10, 43 mg, 0.093 mmol), 2-(3-(methoxymethoxy)naphthalen-1-yl)-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (58.4 mg, 0.186 mmol), XPhos Pd G2 (7.31 mg, 9.29 pmol), and K3PO4 (59.1 mg, 0.279 mmol) in Dioxane (1.5) and Water (0.3 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by flash chromatography (0-60% EtOAc in hexanes) to give the product (38 mg, 66.6% yield). LCMS calculated for C34H36FN4O4S (M+H) + : m/z = 615.2; found 615.2.

Step 2. tert-butyl (endo)-5-(6-fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-2-m ethyl-4- (methylsulfinyl)-1H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2. 1. 1]hexane-2- carboxylate m-CPBA (16.0 mg, 0.093 mmol) was added to a solution of tert-butyl (encfo)-5-(6- fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-2-methyl-4-(met hylthio)-1 H-pyrrolo[3,2- c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (38 mg, 0.067 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCOs solution, dried over MgSO4 and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C34H36FN4O5S (M+H) + : m/z = 631.2; found 631.2.

Step 3. 4-(1-((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-6-fluoro-2-methyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl) ethoxy) - 1 H-pyrrolo[ 3, 2-c][ 1, 6]naphthyridin- 7-yl) naphthalen-2-ol

□HMDS (1M in THF, 60 pl, 0.06 mmol) was added dropwise to a solution of tert-butyl (endo)-5-(6-fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-2-m ethyl-4-(methylsulfinyl)-1 H- pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (intermediate 11 , 13 mg, 0.021 mmol) and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (8.1 mg, 0.063 mmol) in THF (1 mL) at 0 °C, and the reaction was stirred at 0 °C for 30 minutes with LCMS monitoring. The reaction mixture was concentrated in vacuo and the residue was dissolved in TFA (1 mL). The solution was stirred at room temperature for 10 minutes to remove the Boc and MOM protecting groups. The reaction mixture was diluted with methanol, which was purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 3a: Diastereomer 1 , peak 1. LCMS calculated for C33H35FN5O2 (M+H) + : m/z = 552.3; found 552.3. * Potent peak.

Example 3b: Diastereomer 2, peak 2. LCMS calculated for C33H35FN5O2 (M+H) + : m/z = 552.3; found 552.3.

Example 4. 4-(1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2-methy l-4- ((tetrahydro-1 H-pyrrolizin-7a(5H)-yl)methoxy)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-7- yl)naphthalen-2-ol

Example 4 was prepared according to procedure described in Example 3a/3b Step 3, replacing (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol with (tetrahydro-1 H-pyrrolizin- 7a(5H)-yl)methanol. LCMS calculated for C34H35FN5O2 (M+H) + : m/z = 564.3; found 564.3.

Example 5: 3-((1-((enofo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3- hydroxynaphthalen-1-yl)-4-methyl-1H-pyrrolo[3,2-c][1,6]napht hyridin-2- yl)methyl)oxazolidin-2-one

Step 1. tert-butyl (endo)-5-(6-fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-4-( methylthio)-2-

((2-oxooxazolidin-3-yl)methyl)-1H-pyrrolo[3,2-c][1,6]naph thyridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

A mixture of tert-butyl (endo)-5-(7-chloro-6-fluoro-4-(methylthio)-2-((2-oxooxazolid in- 3-yl)methyl)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (intermediate 8, 24 mg, 0.044 mmol), 2-(3-(methoxymethoxy)naphthalen-1-yl)-4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolane (27.5 mg, 0.088 mmol), XPhos Pd G2 (3.45 mg, 4.38 pmol), and K3PO4 (27.9 mg, 0.13 mmol) in Dioxane (0.8 ml) and Water (0.160 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by flash chromatography (0-10% MeOH in DCM) to give the product (20 mg, 65% yield). LCMS calculated for C37H39FN5O6S (M+H) + : m/z = 700.3; found 700.3. Step 2. tert-butyl (endo)-5-(6-fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-4- (methylsulfinyl)-2-((2-oxooxazolidin-3-yl)methyl)-1H-pyrrolo [3,2-c][1,6]naphthyridin-1-yl)-2- m-CPBA (7.43 mg, 0.043 mmol) was added to a solution of tert-butyl (endo)-5-(6- fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-4-(methylthio)- 2-((2-oxooxazolidin-3- yl)methyl)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (20 mg, 0.028 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCOs aq. solution, dried over MgSO4 and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C37H39FN5O7S (M+H) + : m/z = 716.3; found 716.3.

Step 3. 3-((1-((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-4- methyl- 1 H-pyrrolo[ 3, 2-c][1, 6]naphthyridin-2-yl) methyl) oxazolidin-2-one

Methylmagnesium bromide (3M in THF, 13.97 pl, 0.042 mmol) was added to a solution of tert-butyl (endo)-5-(6-fluoro-7-(3-(methoxymethoxy)naphthalen-1-yl)-4- (methylsulfinyl)-2-((2-oxooxazolidin-3-yl)methyl)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (15 mg, 0.021 mmol) in THF at 0 °C, and the resulting mixture was stirred at 0 °C for 1 h with LCMS monitoring. The reaction was quenched with water, extracted with EtOAc, washed with saturated NaHCCh aq. solution, dried over Na2SO4 and then concentrated. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc and MOM protecting groups. The reaction was then diluted with methanol, which was purified prep- LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford example 5 as a TFA salt in the form of a white amorphous powder. LCMS calculated for C30H27FN5O3 (M+H) + : m/z = 524.2; found 524.2.

Example 6a/6b: 3-((1-((enc/o)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(5- fluoro-1 H- indol-3-yl)-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)methyl)oxazolidin-2-one

A mixture of tert-butyl (endo)-5-(7-chloro-6-fluoro-4-((S)-1-((S)-1-methylpyrrolidin -2- yl)ethoxy)-2-((2-oxooxazolidin-3-yl)methyl)-1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (intermediate 9, 23 mg, 0.037 mmol), tert-butyl 5- fluoro-3-(4,4,5,5-tetramethyl-1 ,3-dioxolan-2-yl)-1 H-indole-1-carboxylate (26.9 mg, 0.074 mmol), XPhos Pd G2 (5.82 mg, 7.4 pmol), and K3PO4 (23.3 mg, 0.11 mmol) in Dioxane (0.8 ml) and Water (0.160 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction was concentrated in vacuo. The residue was dissolved in TFA (1 mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting groups. The reaction was then diluted with methanol, which was purified prep- LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 6a: Diastereomer 1 , peak 1. LCMS calculated for C34H36F2N7O3 (M+H) + : m/z = 628.3; found 628.3. * Potent peak. Example 6b: Diastereomer 2, peak 2. LCMS calculated for C34H36F2N7O3 (M+H) + : m/z = 628.3; found 628.3.

Example 7a/7b: 3-((1-((enc/o)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(3,4-dihydro quinolin-

1(2H)-yl)-6-fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl) ethoxy)-1H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)methyl)oxazolidin-2-one

A solution of tert-butyl (encfo)-5-(7-chloro-6-fluoro-4-((S)-1-((S)-1-methylpyrrolidi n-2- yl)ethoxy)-2-((2-oxooxazolidin-3-yl)methyl)-1H-pyrrolo[3,2-c ][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (intermediate 9, 15 mg, 0.024 mmol), 1, 2,3,4- tetrahydroquinoline (4.76 mg, 0.036 mmol), RuPhos Pd G2 (5.56 mg, 7.15 pmol), RuPhos (3.34 mg, 7.15 pmol), and sodium tert-butoxide (6.87 mg, 0.072 mmol) in Dioxane (1 ml) was bubbled with nitrogen gas for 10 minutes, then the reaction was stirred at 105 °C under inert atmosphere for 3h. Upon completion, the reaction was concentrated in vacuo. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting group. The reaction was then diluted with methanol, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 7a: Diastereomer 1, peak 1. LCMS calculated for C35H41FN7O3 (M+H) + : m/z = 626.3; found 626.3. * Potent peak.

Example 7b: Diastereomer 2, peak 2. LCMS calculated for C34H36F2N7O3 (M+H) + : m/z = 62.3; found 626.3.

Example 8a/8b: 1-((enofo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(indoli n-1-yl)-2- methyl-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrr olo[3,2-c][1,6]naphthyridine

A solution of tert-butyl (encfo)-5-(7-chloro-6-fluoro-2-methyl-4-((S)-1-((S)-1- methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (intermediate 11 , 15 mg, 0.028 mmol), indoline (4.93 mg, 0.036 mmol), RuPhos Pd G2 (6.42 mg, 8.27 pmol), RuPhos (3.86 mg, 8.27 pmol), and sodium tert-butoxide (7.95 mg, 0.083 mmol) in Dioxane (1 ml) was bubbled with nitrogen gas for 10 minutes, then the reaction was stirred at 105 °C under inert atmosphere for 3h. Upon completion, the reaction was concentrated in vacuo. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting group. The reaction was then diluted with methanol, which was purified prep- LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 8a: Diastereomer 1, peak 1. LCMS calculated for CsiHseFNeO (M+H) + : m/z = 527.3; found 527.3. * Potent peak.

Example 8b: Diastereomer 2, peak 2. LCMS calculated for CsiHseFNeO (M+H) + : m/z = 527.3; found 527.3.

Example 9a/9b: 3-(1-((enofo)-2-azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-7-(3- hydroxynaphthalen-1-yl)-4-((S)-1-((S)-1-methylpyrrolidin-2-y l)ethoxy)-1H-imidazo[4,5- c][1,6]naphthyridin-2-yl)-N,N-dimethylpropanamide

Step 1. tert-butyl (endo)-5-(2-(3-(dimethylamino)-3-oxopropyl)-6-fluoro-7-(3- (methoxymethoxy)naphthalen- 1-yl)-4-(methylthio)- 1H-imidazo[4, 5-c][ 1, 6]naphthyridin- 1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

A mixture of tert-butyl (endo)-5-(7-chloro-2-(3-(dimethylamino)-3-oxopropyl)-6-fluor o- 4-(methylthio)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (intermediate 12, 50 mg, 0.091 mmol), 2-(3-(methoxymethoxy)naphthalen-1-yl)- 4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (57.2 mg, 0.182 mmol), XPhos Pd G2 (7.16 mg, 9.11 pmol), and K3PO4 (57.9 mg, 0.273 mmol) in Dioxane (1 ml) and Water (0.2 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by flash chromatography (0-10% MeOH in DCM) to give the product (60 mg, 94%). LCMS calculated for C37H42FN6O5S (M+H) + : m/z = 701.3; found 701.3.

Step 2. tert-butyl (endo)-5-(2-(3-(dimethylamino)-3-oxopropyl)-6-fluoro-7-(3- (methoxymethoxy)naphthalen- 1-yl)-4-(methylsulfinyl)-1H-imidazo[4, 5-c][ 1, 6]naphthyridin- 1- yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate m-CPBA (22.1 mg, 0.128 mmol) was added to a solution of tert-butyl (encfo)-5-(2-(3- (dimethylamino)-3-oxopropyl)-6-fluoro-7-(3-(methoxymethoxy)n aphthalen-1-yl)-4- (methylthio)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (60 mg, 0.086 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCOs aq. solution, dried over MgSO4 and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C37H42FN6O6S (M+H) + : m/z = 717.3; found 717.4. Step 3. 3-(1 -((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-4- ((S)-1-((S)-1 -methyl pyrrolidin-2-yl) ethoxy) - 1H-imidazo[ 4, 5-c][ 1, 6]naphthyridin-2-yl) -N, N- dimethylpropanamide

□HMDS (1M in THF, 90 l, 0.09 mmol) was added dropwise to a solution of tert-butyl (endo)-5-(2-(3-(dimethylamino)-3-oxopropyl)-6-fluoro-7-(3-(m ethoxymethoxy)naphthalen-1- yl)-4-(methylsulfinyl)-1H-imidazo[4,5-c][1,6]naphthyridin-1- yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (22 mg, 0.03 mmol) and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (12 mg, 0.09 mmol) in THF (1 mL) at 0 °C, and the reaction was stirred at 0 °C for 30 minutes with LCMS monitoring. The reaction mixture was concentrated in vacuo. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc and MOM protecting groups. The reaction was then diluted with methanol, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 9a: Diastereomer 1, peak 1. LCMS calculated for C36H41FN7O3 (M+H) + : m/z = 638.3; found 638.2. * Potent peak.

Example 9b: Diastereomer 2, peak 2. LCMS calculated for C36H41FN7O3 (M+H) + : m/z = 638.3; found 638.2.

Example 10a/10b: 3-(1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(3-cyano-2,3-d ihydro- 1H-inden-4-yl)-4-(3-(dimethylamino)azetidin-1-yl)-6-fluoro-1 H-imidazo[4,5- c][1,6]naphthyridin-2-yl)-N,N-dimethylpropanamide

Step 1. tert-butyl (endo)-5-(7-(3-cyano-2,3-dihydro-1H-inden-4-yl)-2-(3-(dimeth ylamino)-3- oxopropyl)-6-fluoro-4-(methylthio)-1H-imidazo[4,5-c][1,6]nap hthyridin-1-yl)-2- A mixture of tert-butyl (endo)-5-(7-chloro-2-(3-(dimethylamino)-3-oxopropyl)-6-fluor o- 4-(methylthio)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (intermediate 12, 20 mg, 0.036 mmol), 7-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-2,3-dihydro-1 H-indene-1-carbonitrile (19.3 mg, 0.072 mmol), XPhos Pd G2 (2.83 mg, 3.6 pmol), and K3PO4 (23.2 mg, 0.108 mmol) in Dioxane (0.8 ml) and Water (0.16 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by flash chromatography (0-10% MeOH in DCM) to give the product (21 mg, 90% yield). LCMS calculated for C35H39FN7O3S (M+H) + : m/z = 656.3; found 656.3.

Step 2. tert-butyl (endo)-5-(7-(3-cyano-2,3-dihydro-1H-inden-4-yl)-2-(3-(dimeth ylamino)-3- oxopropyl)-6-fluoro-4-(methylsulfinyl)- 1H-imidazo[4, 5-c][ 1, 6]naphthyridin- 1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate m-CPBA (8.3 mg, 0.48 mmol) was added to a solution of tert-butyl (encfo)-5-(7-(3- cyano-2,3-dihydro-1 H-inden-4-yl)-2-(3-(dimethylamino)-3-oxopropyl)-6-fluoro-4-( methylthio)- 1 H-imidazo[4,5-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (21 mg, 0.032 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 minutes. The reaction was diluted with EtOAc, washed with saturated NaHCOs aq. solution, dried over MgSO4 and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C35H39FN7O4S (M+H) + : m/z = 672.3; found 672.3.

Step 3. 3-(1 -((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-7-(3-cyano-2,3-dihydro-1H-inden-4-yl)-4- (3-(dimethylamino)azetidin- 1-yl) -6- fluoro- 1H-imidazo[4, 5-c][ 1, 6]naphthyridin-2-yl)-N, N- dimethylpropanamide

A mixture of tert-butyl (endo)-5-(7-(3-cyano-2,3-dihydro-1 H-inden-4-yl)-2-(3- (dimethylamino)-3-oxopropyl)-6-fluoro-4-(methylsulfinyl)-1 H-imidazo[4,5-c][1 ,6]naphthyridin- 1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (20 mg, 0.03 mmol), N,N-dimethylazetidin-3- amine dihydrochloride (10.4 mg, 0.06 mmol) and DIPEA (31.4 pl, 0.18 mmol) in CH3CN (1 ml) was stired at 50 °C for 2h with LCMS monitoring. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting group. The reaction was then diluted with methanol, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 10a: Diastereomer 1 , peak 1. LCMS calculated for C34H39FN9O (M+H) + : m/z = 608.3; found 608.3.

Example 10b: Diastereomer 2, peak 2. LCMS calculated for C34H39FN9O (M+H) + : m/z = 608.3; found 608.3. * Potent peak.

Example 11 : methyl 3-(1-((endo)-2-azabicyclo[2.1.1]hexan-5-yl)-4-(3- (dimethylamino)azetidin-1-yl)-6-fluoro-7-(3-hydroxynaphthale n-1-yl)-1H-imidazo[4,5- c][1,6]naphthyridin-2-yl)propanoate

DIPEA (0.87 ml, 5.0 mmol) was added dropwise to a suspension of 2,4,7-trichloro-8- fluoro-3-nitro-1 ,6-naphthyridine (intermediate 1 , 296 mg, 1.0 mmol) and tert-butyl (encfo)-5- amino-2-azabicyclo[2.1.1]hexane-2-carboxylate (200 mg, 1.0 mmol) in DCM (5 ml) at room temperature. After 30 minutes, N,N-dimethylazetidin-3-amine hydrochloride (260 mg, 1.5 mmol) was added to the reaction, and the resulting mixture was stirred for another 1h monitoring with LCMS. The reaction mixture was concentrated in vacuo, and the residue was purified by flash chromatography (0-10% MeOH in DCM) to give the product (392 mg, 75% yield). LCMS calculated for C23H30CIFN7O4 (M+H) + : m/z = 522.2; found 522.2.

Step 2. tert-butyl (endo)-5-((3-amino-7-chloro-2-(3-(dimethylamino)azetidin- 1-yl)-8-fluoro- 1, 6- naphthyridin-4-yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

Sodium hydrosulfite (463 mg, 2.25 mmol) in water (1.5 mL) was added to a solution of tert-butyl (endo)-5-((7-chloro-2-(3-(dimethylamino)azetidin-1-yl)-8-flu oro-3-nitro-1 ,6- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (392 mg, 0.75 mmol) and 30% aq. ammonium hydroxide (0.54 mL, 7.5 mmol) in MeOH (5 mL) at room temperature. After 10 min, the reaction was diluted with water and extracted with DCM for 3 times. The combined organic layers were dried Na2SO4, filtered and concentrated to give the crude product (350 mg, 95% yield), which was used in the next step without further purification. LCMS calculated for C23H32CIFN7O2 (M+H) + : m/z = 492.2; found 492.3.

Step 3. tert-butyl (endo)-5-(7-chloro-4-(3-(dimethylamino)azetidin-1-yl)-6-fluo ro-2-(3- methoxy-3-oxopropyl)- 1H-imidazo[4, 5-c][1, 6]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2- carboxylate

A solution of tert-butyl (endo)-5-((3-amino-7-chloro-2-(3-(dimethylamino)azetidin-1- yl)-8-fluoro-1 ,6-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate (350 mg, 0.71 mmol) and methyl 3-oxopropanoate (217 mg, 2.13 mmol) in i-PrOH (5 mL) was stirred at 80 °C overnight. The reaction mixture was concentrated and the residue was purified by flash chromatography (0-10% MeOH in DCM) to give the product (334 mg, 80% yield). LCMS calculated for C28H36CIFN7O4 (M+H) + : m/z = 588.2; found 588.2.

Step 4. methyl 3-(1-((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-4-(3-(dimethylamino)azetidin-1- yl)-6-fluoro-7-(3-hydroxynaphthalen-1-yl)-1H-imidazo[4,5-c][ 1,6]naphthyridin-2-yl)propanoate

A mixture of tert-butyl (endo)-5-(7-chloro-4-(3-(dimethylamino)azetidin-1-yl)-6-fluo ro- 2-(3-methoxy-3-oxopropyl)-1 H-imidazo[4,5-c][1 ,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (20 mg, 0.034 mmol), 2-(3- (methoxymethoxy)naphthalen-1-yl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (27.6 mg, 0.102 mmol), XPhos Pd G2 (2.68 mg, 3.4 pmol), and K3PO4 (21.6 mg, 0.102 mmol) in Dioxane (0.8 ml) and Water (0.16 ml) was stirred at 105 °C for 2h with LCMS monitoring. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc and MOM protecting groups. The reaction was then diluted with methanol, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford Example 11 as a TFA salt in the form of a white amorphous powder. LCMS calculated for C33H35FN7O3 (M+H) + : m/z = 596.3; found 596.2.

Example 12. 2-((2S,4S)-4-(4-(3-(ethyl(methyl)amino)azetidin-1-yl)-6-fluo ro-7-(5- fluoroquinolin-8-yl)-1 H-[1,2,3]triazolo[4,5-c][1,6]naphthyridin-1-yl)-1-((E)-4-flu orobut-2- enoyl)piperidin-2-yl)acetonitrile

To a solution of tert-butyl (2S,4S)-4-(7-chloro-6-fluoro-4-(methylthio)-1 H- [1 , 2, 3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)-2-(cyanomethyl)piperidine-1 -carboxylate (intermediate 14, 50 mg, 0.102 mmol) in DCM (2 ml) at 0 °C was added m-CPBA (29.6 mg, 0.13 mmol). The reaction mixture was stirred at 0 °C for 20 min. The reaction mixture was dried and concentrated. The crude was dissolve in THF, then DIPEA (89 pl, 0.508 mmol) and N-ethyl-N-methylazetidin-3-amine, 2 HCI (38.0 mg, 0.203 mmol) was added to reaction vial and the resulting mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated and the residue was purified by silica gel column (eluting with a gradient of 0-20% DCM in MeOH) to give the desired product as light brown oil. LC-MS calculated for C26H34CIFN9O2 (M+H) + : m/z = 558.2; found 558.2. Step 2. 2-((2S,4S)-4-(4-(3-(ethyl(methyl)amino)azetidin-1-yl)-6-fluo ro-7-(5-fluoroquinolin-8-

The vial charged with tert-butyl (2S,4S)-4-(7-chloro-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)-2- (cyanomethyl)piperidine-l-carboxylate (20 mg, 0.036 mmol), 5-fluoro-8-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)quinoline (9.79 mg, 0.036 mmol), K3PO4 (15.20 mg, 0.072 mmol), Pd-XphosG2 (2.87 mg, 3.58 pmol) and 5 : 1 dioxane/water (1 ml) were heated at 100 °C for 1 h. The mixture was diluted with brine and EtOAc, the organic layer was separated, dried over MgSC , filtered and concentrated. The residue was dissolved in DCM/TFA 1 :1 (1 mL) solution, stirred for 10 min, then the solvent was removed in vacuo, and the residue was dissolved in DCM, washed with sat. NaHCCU, dried over MgSC , and concentrated. The crude product was used directly in next step. LC-MS calculated for C30H31 F2N10 (M+H) + : m/z = 569.3; found 569.3.

Step 3. 2-((2S,4S)-4-(4-(ethyl(methyl)amino)-6-fluoro-7-(5-fluoroqui nolin-8-yl)- 1H- [1,2,3]triazolo[4,5-c][1,6]naphthyridin-1-yl)-1-((E)-4-fluor obut-2-enoyl)piperidin-2- yl)acetonitrile

To a solution of 2-((2S,4S)-4-(4-(3-(ethyl(methyl)amino)azetidin-1-yl)-6-fluo ro-7-(5- fluoroquinolin-8-yl)-1 H-[1 ,2,3]triazolo[4,5-c][1 ,6]naphthyridin-1-yl)piperidin-2-yl)acetonitrile (20 mg, 0.035 mmol) and (E)-4-fluorobut-2-enoic acid (7.32 mg, 0.070 mmol) in MeCN (0.5 mL) at 0 °C was added DI PEA (18.43 pl, 0.106 mmol) and propanephosphonic acid anhydride (50% solution in 2-methyltetrahydrofuran) (33.6 mg, 0.053 mmol). The reaction mixture was warmed to rt and stirred at this temperature for 20 min. The reaction mixture was dried and concentrated. The mixture was diluted with acetonitrile/water and purified using prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired products as TFA salt. LC-MS calculated for C31H29F3N9O (M+H)+: m/z = 600.3; found 600.3.

Example 13. 2-((2S,4S)-4-(7-(5,6-dimethyl-1H-indazol-3-yl)-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1,2,3]triazolo[4,5-c][1,6]naphthyridin-1- yl)-1-(2-fluoroacryloyl)piperidin-2-yl)acetonitrile

This compound was prepared according to the procedure described in Example 12 step 2, replacing 5-fluoro-8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)quinoline with 5,6- dimethyl-1-(tetrahydro-2H-pyran-2-yl)-3-(4,4,5,5-tetramethyl -1 ,3,2-dioxaborolan-2-yl)-1 H- indazole, and step 3 replacing (E)-4-fluorobut-2-enoic acid with 2-fluoroacrylic acid. LC-MS calculated for C33H36F2N11O (M+H) + : m/z = 640.3; found 640.3.

Example 14. 8-(1-((2S,4S)-2-(cyanomethyl)-1-(2-fluoroacryloyl)piperidin- 4-yl)-4-(3- (ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1 H-[1,2,3]triazolo[4,5-c][1,6]naphthyridin-7- yl)-1-naphthonitrile

This compound was prepared according to the procedure described in Example 12 step 2, replacing 5-fluoro-8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)quinoline with 8- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1-naphthonitrile, and step 3 replacing (E)-4- fluorobut-2-enoic acid with 2-fluoroacrylic acid. LC-MS calculated for C35H33F2N10O (M+H)+: m/z = 647.3; found 647.3.

Example 15. 8-(1-((2S,4S)-2-(cyanomethyl)-1-((E)-4-fluorobut-2-enoyl)pip eridin-4-yl)-4- (3-(ethyl(methyl)amino)azetidin-1-yl)-6-fluoro-1H-[1,2,3]tri azolo[4,5-c][1,6]naphthyridin- 7 -yl)-1 -naphthonitrile

This compound was prepared according to the procedure described in Example 12 step 2, replacing 5-fluoro-8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)quinoline with 8- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1-naphthonitrile. LC-MS calculated for C36H35F2N10O (M+H) + : m/z = 661.3; found 661 .3.

Example 16. 3-(1-((encfo)-2-azabicyclo[2.1.1]hexan-5-yl)-4-(3-(dimethyla mino)azetidin- 1-yl)-7-(3-hydroxynaphthalen-1-yl)-6,8-dimethyl-1H-imidazo[4 ,5-c][1,5]naphthyridin-2- yl)-N,N-dimethylpropanamide

Step 7. tert-butyl (endo)-5-((2,6,7,8-tetrachloro-3-nitro-1,5-naphthyridin-4-yl )amino)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

/V,/V-diisopropylethylamine (3.8 mL, 21.6 mmol) was added to a solution of 2, 3, 4,6,8- pentachloro-7-nitro-1 ,5-naphthyridine (intermediate 2, 2.50 g, 7.20 mmol) and tert-butyl (encfo)-5-amino-2-azabicyclo[2.1.1]hexane-2-carboxylate (1.00 g, 5.04 mmol) in MeCN (100 mL) at room temperature. After stirring for 1 h, the volatiles were removed under reduced pressure. The residue was dissolved in DCM, washed with water, dried over Na2SO4 and concentrated. The crude residue was purified by flash column chromatography (0-20% MeOH/DCM) to afford the desired material. LC-MS calculated for C18H18CI4N5O4 (M+H) + : m/z = 508.0/510.0/512.0; found 508.0/510.0/512.0.

Step 2. tert-butyl (endo)-5-((6, 7,8-trichloro-2-(3-(dimethylamino)azetidin-1-yl)-3-nitro-1,5 - naphthyridin-4-yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

/V,/V-diisopropylethylamine (1.4 mL, 7.66 mmol) was added to a solution of fert-butyl (endo)-5-((2,6,7,8-tetrachloro-3-nitro-1 ,5-naphthyridin-4-yl)amino)-2- azabicyclo[2.1.1]hexane-2-carboxylate (1.30 g, 2.55 mmol) and /V,/V-dimethylazetidin-3- amine dihydrochloride (0.663 g, 3.83 mmol) in MeCN (10 mL) at room temperature. After stirring for 0.5 h, the volatiles were removed under reduced pressure. The residue was dissolved in DCM, washed with water, dried over Na2SO4 and concentrated. The crude residue was purified by flash column chromatography (0-20% MeOH/DCM) to afford the desired material. LCMS calculated for C23H29CI3N7O4 (M+H) + : m/z = 572.1/574.1 ; found 572.1/574.1.

Step 3. tert-butyl (endo)-5-((3-amino-6, 7,8-trichloro-2-(3-(dimethylamino)azetidin-1-yl)-1,5- naphthyridin-4-yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate fert-butyl (endo)-5-((6,7,8-trichloro-2-(3-(dimethylamino)azetidin-1-yl )-3-nitro-1 ,5- naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxy late (2.10 g, 3.67 mmol) was dissolved in glacial acetic acid (30 mL). Iron powder (1.03 g, 18.4 mmol) was added in one portion. The suspension was stirred under nitrogen and heated to 80 °C for 30 minutes. The reaction mixture was cooled to room temperature, and the solvent was removed under reduced pressure. The crude residue was neutralized with sat. aq. NaHCCh and diluted with DCM. The layers were separated and the aqueous layer extracted with DCM. The combined organic fractions were dried over Na2SO4, filtered, and concentrated. The crude residue was purified by flash column chromatography (0-20% MeOH/DCM) to afford the desired material. LCMS calculated for C23H31 CI3N7O2 (M+H) + : m/z = 542.2/544.2; found 542.1/544.1. Step 4. tert-butyl (endo)-5-(6, 7,8-trichloro-2-(3-(dimethylamino)-3-oxopropyl)-4-(3- (dimethylamino)azetidin- 1-yl)-1H-imidazo[4, 5-c][1, 5]naphthyridin- 1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

/V,/V-dimethyl-4-oxobutanamide (0.357 g, 2.76 mmol) was added to a ethanol (10 mL) containing tert-butyl (endo)-5-((3-amino-6,7,8-trichloro-2-(3-(dimethylamino)azeti din-1-yl)- 1 ,5-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate (0.500 g, 0.921 mmol) and pyridium p-toluenesulfonate (0.231 g, 0.921 mmol). After stirring for 16 hours, the volatiles were removed under reduced pressure. The residue was dissolved in DCM, washed with aqueous NaHCCh solution, dried over Na2SO4, filtered and concentrated. The crude residue was purified by flash column chromatography (0-20% MeOH/DCM) to afford the desired material. LCMS calculated for C29H38CI3N8O3 (M+H) + : m/z = 651.2/653.2; found 651.1/653.2.

Step 5. tert-butyl (endo)-5-(6, 7-dichloro-2-(3-(dimethylamino)-3-oxopropyl)-4-(3- (dimethylamino)azetidin- 1-yl)-8-methyl- 1H-imidazo[4, 5-c][ 1, 5]naphthyridin- 1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

A reaction vial was charged with tert-butyl (endo)-5-(6,7,8-trichloro-2-(3- (dimethylamino)-3-oxopropyl)-4-(3-(dimethylamino)azetidin-1- yl)-1/7-imidazo[4,5- c][1 ,5]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (0.166 g, 0.255 mmol) and iron(lll) acetylacetonate (0.045 g, 0.127 mmol). The vial was evacuated and refilled with N2 three times, followed by the addition of 1.2 mL anhydrous THF and MeMgBr (3 M THF solution, 255 pL, 0.764 mmol). The reaction was stirred for 2 h and quenched with aqueous solution of NH4CI. The mixture was extracted with ethyl acetate and the organic layer was dried over Na2SO4, filtered and concentrate. The crude residue was purified by flash column chromatography (0-30% MeOH/DCM) to afford the desired material. LCMS calculated for C30H41CI2N8O3 (M+H) + : m/z = 631.3; found 631 .5.

Step 6. tert-butyl (endo)-5-(7-chloro-2-(3-(dimethylamino)-3-oxopropyl)-4-(3- (dimethylamino)azetidin- 1-yl)-6, 8-dimethyl- 1H-imidazo[4, 5-c][ 1, 5]naphthyridin- 1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate

A reaction vial was charged with tert-butyl (endo)-5-(6,7-dichloro-2-(3- (dimethylamino)-3-oxopropyl)-4-(3-(dimethylamino)azetidin-1- yl)-8-methyl-1/7-imidazo[4,5- c][1 ,5]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (10 mg, 0.016 mmol), tetrakis(triphenylphosphine)palladium(0) (2 mg, 1.6 pmol), potassium methyltrifluoroborate (2,9 mg, 0.024 mmol), K3PO4 (10 mg, 0.047 mmol) and 10:1 dioxane/water solution (0.2 mL). The mixture was sparged with N2 for 1 min before it was heated at 110 °C for 2 hours. Upon completion, the reaction was cooled to room temperature, diluted with water and DCM, and the layers were separated. The aqueous layer was extracted with DCM. The combined organic fractions were dried over Na2SO4, filtered and concentrated. The crude residue was purified by preparative LCMS to afford the desired product. LCMS calculated for C31 H44CIN8O3 (M+H) + : m/z = 611 .3; found 611.3.

Step 7. 3-(1-((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-4-(3-(dimethylamino)azetidin-1-yl)-7-(3- hydroxynaphthalen- 1-yl)-6, 8-dimethyl- 1H-imidazo[4, 5-c][ 1, 5]naphthyridin-2-yl)-N, N- dimethylpropanamide

A reaction vial was charged with tert-butyl (endo)-5-(7-chloro-2-(3-(dimethylamino)-3- oxopropyl)-4-(3-(dimethylamino)azetidin-1-yl)-6,8-dimethyl-1 H-imidazo[4,5- c][1 ,5]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (5 mg, 8.2 pmol), XPhos Pd G2 (1 mg, 1 pmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)naphthalene-2-ol (3 mg, 0.012 mmol), K3PO4 (7 mg, 0.033 mmol) and 10:1 dioxane/water solution (0.2 mL). The mixture was sparged with N2 for 1 min before it was heated at 100 °C for 2 hours. Upon completion, the reaction was filtered through a pad of Celite packed on a medium porosity fritted filter. The Celite was washed with MeCN. The filtrates were combined and concentrated.

The crude residue was dissolved into 1 :1 DCM/TFA (1 ml) and stirred at room temperature for 30 mins. Upon deprotection, the reaction mixture was concentrated, diluted with MeCN (5 mL) and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the desired product as a TFA salt. The products were isolated as diastereomers. LCMS calculated for C36H43N8O2 (M+H) + : m/z = 619.3; found 619.3.

Example 17a/17b. 3-(1-((enofo)-2-azabicyclo[2.1.1]hexan-5-yl)-8-(2-cyanoethyl )-4-(3- (dimethylamino)azetidin-1-yl)-7-(3-hydroxynaphthalen-1-yl)-1 H-imidazo[4,5- c][1,5]naphthyridin-2-yl)-N,N-dimethylpropanamide

Step 1. tert-butyl (endo)-5-((7-bromo-2,6-dichloro-3-nitro-1,5-naphthyridin-4-y l)amino)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

This compound was prepared according to the procedure described in Example 16, Step 1 , replacing intermediate 2 with 7-bromo-2,4,6-trichloro-3-nitro-1,5-naphthyridine (intermediate 3). LCMS calculated for C H BrC^NsC (M+H) + : m/z = 518.0/520.0/522.0; found 518.0/520.0/522.0.

Step 2. tert-butyl (endo)-5-((7-bromo-6-chloro-2-(3-(dimethylamino)azetidin- 1-yl)-3-nitro- 1, 5- naphthyridin-4-yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

This compound was prepared according to the procedure described in Example 16,

Step 2, replacing tert-butyl (er?cfo)-5-((2,6,7,8-tetrachloro-3-nitro-1,5-naphthyridin-4- yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxylate with tert-butyl (endo)-5-((7-bromo-2,6- dichloro-3-nitro-1 ,5-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate.

LCMS calculated for C 2 3H3oBrCIN 7 0 4 (M+H) + : m/z = 582.1/584.1 ; found 582.1/584.1.

Step 3. tert-butyl (endo)-5-((3-amino-7-bromo-6-chloro-2-(3-(dimethylamino)azet idin-1-yl)-

1,5-naphthyridin-4-yl)amino)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

This compound was prepared according to the procedure described in Example 16, Step 3, replacing tert-butyl (endo)-5-((6,7,8-trichloro-2-(3-(dimethylamino)azetidin-1-yl )-3- nitro-1 ,5-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate with tert-butyl (endo)-5-((7-bromo-6-chloro-2-(3-(dimethylamino)azetidin-1-y l)-3-nitro-1 ,5-naphthyridin-4- yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxylate. LCMS calculated for C23H32BrCIN 7 O2 (M+H) + : m/z = 552.1/554.1 ; found 552.1/554.1.

Step 4. tert-butyl (endo)-5-(7-bromo-8-chloro-2-(3-(dimethylamino)-3-oxopropyl) -4-(3- (dimethylamino)azetidin- 1-yl)-1H-imidazo[4, 5-c][1, 5]naphthyridin- 1-yl)-2-

This compound was prepared according to the procedure described in Example 16, Step 4, replacing tert-butyl (endo)-5-((3-amino-6,7,8-trichloro-2-(3-(dimethylamino)azeti din- 1-yl)-1 ,5-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane-2-carb oxylate with tert-butyl (endo)-5-((3-amino-7-bromo-6-chloro-2-(3-(dimethylamino)azet idin-1-yl)-1 ,5-naphthyridin-4- yl)amino)-2-azabicyclo[2.1.1]hexane-2-carboxylate. LCMS calculated for C29H39BrCINsO3 (M+H) + : m/z = 661.2/663.2; found 661.1/663.1.

Step 5. tert-butyl (endo)-5-(8-chloro-2-(3-(dimethylamino)-3-oxopropyl)-4-(3- (dimethylamino)azetidin- 1-yl)-7-(3-(methoxymethoxy)naphthalen- 1-yl)- 1H-imidazo[4, 5- c][ 1, 5]naphthyridin- 1-yl)-2-azabicyclo[2. 1. 1]hexane-2-carboxylate

A reaction vial was charged with tert-butyl (endo)-5-(7-bromo-8-chloro-2-(3- (dimethylamino)-3-oxopropyl)-4-(3-(dimethylamino)azetidin-1- yl)-1/7-imidazo[4,5- c][1 ,5]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (111 mg, 0.168 mmol), tetrakis(triphenylphosphine)palladium(0) (29 mg, 0.025 mmol), 2-(3- (methoxymethoxy)naphthalen-1-yl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (53 mg, 0.168 mmol), K3PO4 (142 mg, 0.671 mmol) and 10:1 dioxane/water solution (0.8 mL). The mixture was sparged with N2 for 1 min before it was heated at 100 °C for 2 hours. Upon completion, the reaction was filtered through a pad of Celite packed on a medium porosity fritted filter. The Celite was washed with MeCN. The filtrates were combined and concentrated. The crude residue was purified by flash column chromatography (0-30% MeOH/DCM) to afford the desired material. LCMS calculated for C41H50CIN8O5 (M+H) + : m/z = 769.4; found 769.3. Step 6. 3-(1 -((endo)-2-azabicyclo[2. 1. 1]hexan-5-yl)-8-(2-cyanoethyl)-4-(3- (dimethylamino)azetidin- 1-yl)- 7-(3-hydroxynaphthalen- 1-yl)-1 H-imidazo[ 4, 5- c][ 1, 5]naphthyridin-2-yl)-N, N-dimethylpropanamide

A reaction vial was charged with tert-butyl (endo)-5-(8-chloro-2-(3-(dimethylamino)-3- oxopropyl)-4-(3-(dimethylamino)azetidin-1-yl)-7-(3-(methoxym ethoxy)naphthalen-1-yl)-1/7- imidazo[4,5-c][1 ,5]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (50 mg, 0.065 mmol) and XPhosPd G4 (17 mg, 0.032 mmol). After evacuation and refilling with N2 three times, 0.5 M THF solution of (2-cyanoethyl)zinc(ll) bromide (0.65 mL, 0.325 mmol) was added and the reaction heated at 90 °C for 16 hours. Upon completion, the reaction was filtered through a pad of Celite packed on a medium porosity fritted filter. The Celite was washed with MeCN. The filtrates were combined and concentrated.

The crude residue was dissolved into 1 :1 DCM/TFA (1 ml). After stirring at room temperature for 10 mins, a drop of water was added and the mixture was stirred for another 30 mins. Upon deprotection, the reaction mixture was concentrated, diluted with MeCN (5 mL) and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the desired product as a TFA salt. The products were isolated as pairs of enantiomers. Example 17a: Diastereomer 1 , peak 1. LCMS calculated for C37H42N9O2 (M+H) + m/z = 644.3; found 644.3.

Example 17b: Diastereomer 2, peak 2. LCMS calculated for C37H42N9O2 (M+H) + m/z = 644.3; found 644.3

Example 18. 4-(1-((7/?,4/?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-2 -methyl-4-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrrolo[3,2-c][1,6]n aphthyridin-7-yl)-6- fluoronaphthalen-2-ol

This compound was prepared according to the procedure described in Example 3a/3b, by using terf-butyl (7F?,4F?,5S)-5-(7-chloro-6-fluoro-2-methyl-4-(methylthio)-1/ 7- pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (intermediate 16) instead of intermediate 10, and replacing 2-(3-(methoxymethoxy)naphthalen-1-yl)- 4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane with 2-(7-fluoro-3-(methoxymethoxy)naphthalen-1- yl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane.

Example 18: isolated as TFA salt, LCMS calculated for C33H34F2N5O2 (M+H) + m/z = 570.3; found 570.3. 1 H NMR (600 MHz, DMSO-d6) 5 10.11 (s, 1 H), 9.60 (s, 1 H), 9.36 (s, 1 H), 8.32 (s, 1 H), 7.92 (dd, J = 9.1 , 6.0 Hz, 1 H), 7.42 - 7.28 (m, 4H), 6.76 (s, 1 H), 5.68 (dt, J = 9.2, 6.2 Hz, 1 H), 5.40 (d, J = 2.9 Hz, 1 H), 4.98 (s, 1 H), 3.96 - 3.79 (m, 2H), 3.59 (dq, J = 12.3, 6.2 Hz, 1 H), 3.46 - 3.36 (m, 1 H), 3.18 (dq, J = 12.0, 6.3 Hz, 1 H), 3.07 (d, J = 4.6 Hz, 3H), 3.04 - 2.96 (m, 1 H), 2.55 (s, 3H), 2.35 - 2.25 (m, 2H), 2.15 - 2.02 (m, 1 H), 1.98 - 1.86 (m, 2H), 1.60 (d, J = 9.1 Hz, 1 H), 1.53 (d, J = 6.0 Hz, 3H).

Example 19. 1-((/?)-2-(7-(Benzo[oQthiazol-4-yl)-1-((7/?,4/?,5S)-2-azabic yclo[2.1.1]hexan- 5-yl)-6-fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy) -1H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)pyrrolidin-1-yl)ethan-1-one

A mixture of tert-butyl (7 ?,4 ?,5S)-5-(2-(( ?)-1-acetylpyrrolidin-2-yl)-7-chloro-6-fluoro- 4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1/7-pyrrolo[3, 2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (intermediate 18, 50 mg, 0.078 mmol), 4-(4, 4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]thiazole (61.1 mg, 0.234 mmol), Xphos Pd G2 (18.41 mg, 0.023 mmol), and K3PO4 (49.6 mg, 0.234 mmol) in dioxane (5.0 ml) and Water (1.0 ml) was stirred at 90 °C for 2h. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in TFA (2 ml) and DCM (2 ml), and the solution was stirred at room temperature for 30 minutes to remove the Boc protecting group. The reaction was then diluted with CH3CN, which was purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 19: LCMS calculated for C35H39FN7O2S (M+H) + m/z = 640.3; found 640.3. 1 H NMR (600 MHz, DMSO-d6) 59.99 (s, 1H), 9.53 (s, 1H), 9.42 (s, 1H), 9.29 (s, 1H), 8.35 (dd, J = 8.1, 1.2 Hz, 1H), 7.80 (dd, J = 7.4, 1.2 Hz, 1H), 7.70 (td, J =7.7, 2.5 Hz, 1H), 6.47 (s, 1H), 5.72 (dq, J= 9.1, 6.2 Hz, 1H), 5.52 (d, J= 2.9 Hz, 1H), 5.20 (d, J= 8.2 Hz, 1H), 4.88 (d, J = 6.1 Hz, 1H), 3.96-3.90 (m, 1H), 3.90-3.80 (m, 2H), 3.77 (t, J= 8.7 Hz, 1H), 3.63-3.52 (m, 2H), 3.45-3.40 (m, 1H), 3.22-3.12 (m, 1H), 3.04 (d, J= 4.7 Hz, 3H), 2.35-2.23 (m, 3H), 2.17 (s, 3H), 2.13-2.04 (m, 1H), 2.00-1.87 (m, 3H), 1.82 (dd, J= 12.6, 6.5 Hz, 1H), 1.71 - 1.61 (m, 1H), 1.59 (d, J= 9.2 Hz, 1H), 1.54 (d, J= 6.3 Hz, 3H).

Example 20a/20b: 3-(1-((7/?,4/?,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-7-(chroman -8-yl)-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrr olo[3,2-c][1,6]naphthyridin-

2-yl)-2,3-dihydroindolizin-5 To a solution of terf-butyl (1F?,4F?,5S)-5-((7-(chroman-8-yl)-8-fluoro-3-iodo-2-((S)-1- ((S)-1-methylpyrrolidin-2-yl)ethoxy)-1 ,6-naphthyridin-4-yl)amino)-2-azabicyclo[2.1.1]hexane- 2-carboxylate (Intermediate 19, 18 mg, 0.025 mmol) and 3-ethynyl-2,3-dihydroindolizin- 5(1/-/)-one (Intermediate 20, 12 mg, 0.074 mmol) in DMF (0.3 ml) was added DIPEA (0.043 ml, 0.247 mmol), Pd(Phs )4 (5.70 mg, 4.93 pmol) and copper(l) iodide (1.879 mg, 9.87 pmol). The reaction mixture was heated to 50 °C for 1 hour. Then CS2CO3 (0.080 g, 0.247 mmol) was added to the reaction mixture. The reaction mixture was heated at 90 °C for 1 hour. The reaction mixture was quenched with NH4OH solution and extracted with DCM. The combined organic fractions were dried over MgSC , filtered, and concentrated. To the crude residue was added DCM (1 ml) and TFA (1 ml) the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated then diluted with acetonitrile (5 ml) and filtered. The crude solution was purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder.

Example 20a: Diastereomer 1 , peak 1. LCMS calculated for C39H42FN6O3 (M+H) + : m/z =661.3; found 661.3.

Example 20b: Diastereomer 2, peak 2. LCMS calculated for C39H42FN6O3 (M+H) + : m/z =661.3; found 661.3.

Example 21. 1-((R)-1-(1-((1R,4R,5S)-2-Azabicyclo[2.1.1]hexan-5-yl)-7-(ch roman-8-yl)-6- fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrr olo[3,2-c][1,6]naphthyridin- 2-yl)ethyl)pyridin-2(1H)-one

Example 21 was prepared according to procedure described in Example 20a/20b, replacing 3-ethynyl-2,3-dihydroindolizin-5(1/7)-one (Intermediate 20) with (F?)-1-(but-3-yn-2- yl)pyridin-2(1/7)-one (Intermediate 21). LCMS calculated for C38H42FN6O3 (M+H) + : m/z =649.3; found 649.5.

Example 22: Methyl (R)-2-(1-((1R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2,3- dichlorophenyl)-4-(3-(dimethylamino)-3-methylazetidin-1-yl)- 6-fluoro-1H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)pyrrolidine-1 -carboxylate

To the reaction mixture of terf-butyl (7F?,4F?,5S)-5-(7-(2,3-dichlorophenyl)-6-fluoro-2- ((F?)-1-(methoxycarbonyl)pyrrolidin-2-yl)-4-(methylsulfinyl) -1 H-pyrrolo[3,2-c][1 ,6]naphthyridin- 1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (Intermediate 23, 50 mg, 0.071 mmol) and /V,/V,3-trimethylazetidin-3-amine dihydrochloride (39.7 mg, 0.213 mmol) in acetonitrile (1.423 mL) was added DIPEA (62.1 pL, 0.356 mmol). The reaction mixture was stirred at 70 °C for 30 minutes. Upon completion, the reaction was concentrated in vacuo. The residue was dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting group. The reaction was then diluted with acetonitrile, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder. LC-MS calculated for C33H37CI2FN7O2 (M+H) + : m/z = 652.2; found 652.2. 1 H NMR (600 MHz, DMSO-cfe, 4/6 ratio rotamer in solution) 10.88 (s, 1 H), 9.31 (m, 1 H), 8.33 (s, 0.4H), 8.24 (s, 0.6H), 7.80 (m, 1 H), 7.56 - 7.52 (m, 2H), 6.41 (m, 1 H), 5.45 (m, 1 H), 5.10 (m, 1 H), 4.81 (m, 1 H), 4.69 (m, 1 H), 4.60 (m, 1 H), 4.32 (m, 2H), 3.90 (m, 1 H), 3.74 - 3.61 (m, 5H), 3.52 - 3.38 (m, 2H), 2.84 (s, 6H), 2.36 - 2.21 (m, 2H), 1,92 - 1.72 (m, 2H), 1.67 (s, 3H), 1.58 (m, 1 H).

Example 23: Methyl (R)-2-(1-((7R,4R,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2,3- dichlorophenyl)-6-fluoro-4-((S)-1-((S)-1-methylpyrrolidin-2- yl)ethoxy)-1H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)pyrrolidine-1 -carboxylate

At 0 °C, to a solution of terf-butyl (7F?,4F?,5S)-5-(7-(2,3-dichlorophenyl)-6-fluoro-2- ((R)-1-(methoxycarbonyl)pyrrolidin-2-yl)-4-(methylsulfinyl)- 1H-pyrrolo[3,2-c][1,6]naphthyridin- 1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylate (Intermediate 23, 100 mg, 0.142 mmol) and (S)-1-((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (55.2 mg, 0.427 mmol) in THF (1.423 mL) was added 1.0M potassium terf-butoxide solution in THF (427 pL, 0.427 mmol). The reaction mixture was stirred at 0 °C for 30minutes. Upon completion, the reaction was filtered through celite and washed with EtOAc. The filtrate was concentrated in vacuo. The resulting residue was then dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting group. The reaction was then diluted with acetonitrile, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder. LC-MS calculated for C34H38CI2FN6O3 (M+H) + : m/z = 667.2; found 667.2.

Example 24. Methyl (/?)-2-(1-((7/?,4/?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(2- chloro-3- methylphenyl)-6-fluoro-4-(6-(methylcarbamoyl)pyridin-2-yl)-1 H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)pyrrolidine-1 -carboxylate

Step 1. tert-Butyl (1R,4R,5S)-5-(7-(2-chloro-3-methylphenyl)-6-fluoro-2-((R)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylthio)-1H-pyrrolo[ 3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

Under the atmosphere of nitrogen, the reaction mixture of terf-butyl (1 R,4R,5S)-5-(7- chloro-6-fluoro-2-((F?)-1-(methoxycarbonyl)pyrrolidin-2-yl)- 4-(methylthio)-1H-pyrrolo[3,2- c][1,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carbox ylate (Intermediate 22, 450 mg, 0.781 mmol), potassium phosphate tribasic (497 mg, 2.343 mmol), (2-chloro-3- methylphenyl)boronic acid (666 mg, 3.91 mmol) and tetrakis(triphenylphosphine)palladium (271 mg, 0.234 mmol) in Dioxane (6.51 mL) and Water (1.302 mL) was stirred at 110 °C for 3 hours. Upon completion, the reaction was cooled down and poured into water, extracted with EtOAc. The combined organic layers were dried over anhydrous MgSC , concentrated and purified by flash chromatography (0-100% EtOAc in Hexanes) to afford the product as light yellow solid (400 mg, 77% yield). LC-MS calculated for C34H38CIFN5O4S (M+H) + : m/z = 666.2; found 666.2.

Step 2. N-methyl-6-(trimethylstannyl)picolinamide

Under the atmosphere of nitrogen, the reaction mixture of 6-bromo-N- methylpicolinamide (40 mg, 0.186 mmol), hexamethylditin (57.9 pL, 0.279 mmol) and tetrakis(triphenylphosphine)palladium (21.49 mg, 0.019 mmol) in Dioxane (0.620 mL) were stirred at 100 °C for overnight. The mixture was cooled to room temperature and then filtered over a pad of celite. The filtrate was concentrated to give the crude product, which was used in the next step without further purification.

Step 3. methyl (R)-2-(1-((1R,4R,5S)-2-azabicyclo[2. 1. 1]hexan-5-yl)-7-(2-chloro-3- methylphenyl)-6-fluoro-4-(6-(methylcarbamoyl)pyridin-2-yl)-1 H-pyrrolo[3,2- c][ 1, 6]naphthyridin-2-yl)pyrrolidine- 1 -carboxylate

Under the atmosphere of nitrogen, the reaction mixture of terf-butyl (1R,4R,5S)-5-(7- (2-chloro-3-methylphenyl)-6-fluoro-2-((F?)-1-(methoxycarbony l)pyrrolidin-2-yl)-4-(methylthio)- 1 H-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (20 mg, 0.030 mmol), tetrakis(triphenylphosphine)palladium (6.94 mg, 6.00 pmol), copper(l) 3- methylsalicylate (23.20 mg, 0.108 mmol), and /V-methyl-6-(trimethylstannyl)picolinamide (22.44 mg, 0.075 mmol) in Dioxane (0.300 mL) was stirred at 100 °C for 2 hours. Upon completion, the reaction was cooled down to room temperature, diluted with EtOAc and filtered through Slica Prep Thiol. The filtrate was concentrated and the resulting residue was then dissolved in TFA (1mL), and the solution was stirred at room temperature for 10 minutes to remove the Boc protecting group. The reaction was then diluted with acetonitrile, which was purified prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1 % TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder. LC-MS calculated for C35H34CIFN7O3 (M+H) + : m/z = 654.2; found 654.2 Example 25: methyl (R)-2-(7-(benzo[c/]thiazol-4-yl)-1-((1R,4R,5S)-2- azabicyclo[2.1.1]hexan-5-yl)-6-fluoro-4-(((S)-1-(pyrrolidin- 1-yl)propan-2-yl)oxy)-1H- pyrrolo[3,2-c][1,6]naphthyridin-2-yl)pyrrolidine-1 -carboxylate

Step 1. tert-butyl (1R,4R,5S)-5-(7-(benzo[d]thiazol-4-yl)-6-fluoro-2-((R)-1-

(methoxycarbonyl)pyrrolidin-2-yl)-4-(methylthio)-1H-pyrro lo[3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

A mixture of tert-butyl (1R,4R,5S)-5-(7-chloro-6-fluoro-2-((R)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylthio)-1/7-pyrrolo [3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (Intermediate 22, 250 mg, 0.43 mmol), 4-(4, 4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]thiazole (170 mg, 0.65 mmol), XPhos Pd G2 (34 mg, 0.043 mmol), and K3PO4 (274 mg, 1.3 mmol) in Dioxane (4 ml) and Water (1 ml) was stirred at 100 °C for 1 h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography to give the product (203 mg, 70%). LCMS calculated for C34H36FN6O4S2 (M+H) + : m/z = 675.2; found 675.3.

Step 2. tert-butyl (1R,4R,5S)-5-(7-(benzo[d]thiazol-4-yl)-6-fluoro-2-((R)-1- (methoxycarbonyl)pyrrolidin-2-yl)-4-(methylsulfinyl)- 1H- pyrrole [3, 2-c][ 1, 6]naphthyridin- 1-yl)- 2-azabicyclo[2. 1. 1]hexane-2-carboxylate m-CPBA (18 mg, 77% w/w, 0.075 mmol) was added to a solution of terf-butyl (1R,4R,5S)-5-(7-(benzo[c(]thiazol-4-yl)-6-fluoro-2-((/?)-1-( methoxycarbonyl)pyrrolidin-2-yl)-4- (methylthio)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2-carboxylat e (50 mg, 0.074 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 min. The reaction was diluted with DCM, washed with saturated aq. NaHCOs solution, dried over MgSC and concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C34H36FN6O5S2 (M+H) + : m/z = 691.2; found 691.2.

Step 3. methyl (R)-2-(7-(benzo[d]thiazol-4-yl)-1-((1R,4R,5S)-2-azabicyclo[2 .1.1]hexan-5-yl)- 6-fluoro-4-(((S)- 1-(pyrrolidin- 1-yl)propan-2-yl) oxy)- 1 H- pyrrol o [3, 2-c][ 1, 6]naphthyridin-2- yl)pyrrolidine- 1 -carboxylate

□HMDS (1M in THF, 100 pl, 0.1 mmol) was added dropwise to a solution of (S)-1- (pyrrolidin-1-yl)propan-2-ol (13 mg, 0.1 mmol) in THF (0.5 mL) at 0 °C. After 15 mins, terf- butyl (1R,4R,5S)-5-(7-(benzo[c(]thiazol-4-yl)-6-fluoro-2-((/?)-1-( methoxycarbonyl)pyrrolidin-2- yl)-4-(methylsulfinyl)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane-2- carboxylate (15 mg, 0.022 mmol) in THF (0.3 mL) was added at 0 °C, and the reaction was stirred at 0 °C for 30 min with LCMS monitoring. The reaction mixture was concentrated in vacuo, and the residue was dissolved in TFA (1 mL). The solution was stirred at r.t. for 10 min, then mixture was diluted with methanol and was purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the product as a TFA salt in the form of a white amorphous powder. LCMS calculated for C35H39FN7O3S (M+H) + : m/z = 656.3; found 656.3.

Example 26a/26b: (7aS)-5-(1-((1/?,4/?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(c hroman-8- yl)-6-fluoro-4-((S)-1 -(( S)-1 -methylpyrrolidin-2-yl)ethoxy)-1 H-pyrrolo[3,2- c][1,6]naphthyridin-2-yl)tetrahydro-1H,3H-pyrrolo[1,2-c]oxaz ol-3-one

Step 1. tert-butyl (1R,4R,5S)-5-(7-chloro-6-fluoro-4-(methylthio)-2-((7aS)-3-ox otetrahydro-

1 H, 3H-pyrrolo[ 1 ,2-c]oxazol-5-yl) - 1 H -pyrrolo[3, 2-c][ 1, 6]naphthyridin- 1-yl) -2- azabicyclo[2.1. 1]hexane-2-carboxylate

Boc

This compound was prepared according to the procedure described for Intermediate 7 by replacing methyl pent-4-ynoate with (7aS)-5-ethynyltetrahydro-1/7,3/7-pyrrolo[1 ,2- c]oxazol-3-one (Intermediate 24). LCMS calculated for C27H30CIFN5O4S (M+H) + : m/z = 574.2; found 574.2.

Step 2. tert-butyl (1R,4R,5S)-5-(7-(chroman-8-yl)-6-fluoro-4-(methylthio)-2-((7 aS)-3- oxotetrahydro-1H,3H-pyrrolo[1,2-c]oxazol-5-yl)-1H-pyrrolo[3, 2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1. 1]hexane-2-carboxylate

A mixture of terf-butyl (1R,4R,5S)-5-(7-chloro-6-fluoro-4-(methylthio)-2-((7aS)-3- oxotetrahydro-1/7,3/7-pyrrolo[1,2-c]oxazol-5-yl)-1/7-pyrrolo [3,2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (150 mg, 0.26 mmol), chroman-8-ylboronic acid (70 mg, 0.39 mmol), XPhos Pd G2 (20 mg, 0.026 mmol), and K3PO4 (274 mg, 1.3 mmol) in Dioxane (4 ml) and Water (1 ml) was stirred at 100 °C for 1 h with LCMS monitoring. Upon completion, the reaction was diluted with EtOAc, washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography to give the product (140 mg, 80%). LCMS calculated for C36H39FN5O5S (M+H) + : m/z = 672.3; found 672.3.

Step 3. tert-butyl (1R,4R,5S)-5-(7-(chroman-8-yl)-6-fluoro-4-(methylsulfinyl)-2 -((7aS)-3- oxotetrahydro-1H,3H-pyrrolo[1,2-c]oxazol-5-yl)-1H-pyrrolo[3, 2-c][1,6]naphthyridin-1-yl)-2- azabicyclo[2. 1. 1]hexane-2-carboxylate m-CPBA (18 mg, 77% w/w, 0.075 mmol) was added to a solution of ferf-butyl (1R,4R,5S)-5-(7-(chroman-8-yl)-6-fluoro-4-(methylthio)-2-((7 aS)-3-oxotetrahydro-1/7,3/7- pyrrolo[1 ,2-c]oxazol-5-yl)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2-azabicyclo[2.1.1]hexane- 2-carboxylate (50 mg, 0.074 mmol) in DCM (1 ml) at 0 °C, and the resulting mixture was stirred at 0 °C for 30 min. The reaction was diluted with DCM, washed with saturated aq. NaHCOs solution, dried over MgSC and then concentrated in vacuo to give the crude product, which was used in the next step without further purification. LCMS calculated for C36H39FN5O6S (M+H) + : m/z = 688.2; found 688.2.

Step 4. (7aS)-5-(1-((1R,4R,5S)-2-azabicyclo[2. 1. 1]hexan-5-yl)-7-(chroman-8-yl)-6-fluoro-4- ((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1R-pyrrolo[3,2-c ][1,6]naphthyridin-2-yl)tetrahydro- 1 H, 3H-pyrrolo[ 1 ,2-c]oxazol-3-one

□HMDS (1M in THF, 100 pl, 0.1 mmol) was added dropwise to a solution of (S)-1- ((S)-1-methylpyrrolidin-2-yl)ethan-1-ol (13 mg, 0.1 mmol) in THF (0.5 mL) at 0 °C. After 15 mins, terf-butyl (1R,4R,5S)-5-(7-(chroman-8-yl)-6-fluoro-4-(methylsulfinyl)-2 -((7aS)-3- oxotetrahydro-1/7,3/7-pyrrolo[1 ,2-c]oxazol-5-yl)-1/7-pyrrolo[3,2-c][1 ,6]naphthyridin-1-yl)-2- azabicyclo[2.1.1]hexane-2-carboxylate (14 mg, 0.02 mmol) in THF (0.3 mL) was added at 0 °C, and the reaction was stirred at 0 °C for 30 min with LCMS monitoring. The reaction mixture was concentrated in vacuo, and the residue was dissolved in TFA (1 mL). The solution was stirred at room temperature for 10 minutes, then diluted with methanol and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the products as a TFA salt in the form of a white amorphous powder.

Example 26a peak 1 : LCMS calculated for C37H42FN6O4 (M+H) + : m/z = 653.3; found 653.3.

Example 26b peak 2: LCMS calculated for C37H42FN6O4 (M+H) + : m/z = 653.3; found 653.3. Example 27. 4-(1-((1/?,4/?,5S)-2-azabicyclo[2.1.1]hexan-5-yl)-7-(chroman -8-yl)-6-fluoro- 4-((S)-1-((S)-1-methylpyrrolidin-2-yl)ethoxy)-1H-pyrrolo[3,2 -c][1,6]naphthyridin-2-yl)- A/,/V,1-trimethyl-1H-pyrazole-3-carboxamide

Example 27 was prepared according to the procedure described in Example 20a/20b, replacing 3-ethynyl-2,3-dihydroindolizin-5(1/-/)-one with 4-ethynyl-/V,/V,1-trimethyl- 1/7-pyrazole-3-carboxamide (Intermediate 25). LCMS calculated for C38H44FN8O3 (M+H) + : m/z = 679.3; found 679.4.

Example A. GDP-GTP exchange assay.

The inhibitor potency of the exemplified compounds was determined in a fluorescence based guanine nucleotide exchange assay, which measures the exchange of bodipy-GDP (fluorescently labeled GDP) for GppNHp (Non-hydrolyzable GTP analog) to generate the active state of KRAS in the presence of SOS1 (guanine nucleotide exchange factor). Inhibitors were serially diluted in DMSO and a volume of 0.1 L was transferred to the wells of a black low volume 384-well plate. 5 L/well volume of bodipy-loaded KRAS G12D diluted to 2.5 nM in assay buffer (25 mM Hepes pH 7.5, 50 mM NaCI, 10 mM MgCh and 0.01% Brij-35) was added to the plate and pre-incubated with inhibitor for 4 hours at ambient temperature. Appropriate controls (enzyme with no inhibitor or with a G12D inhibitor) were included on the plate. The exchange was initiated by the addition of a 5 L/well volume containing 1 mM GppNHp and 300 nM SOS1 in assay buffer. The 10 L/well reaction concentration of the bodipy-loaded KRAS G12D, GppNHp, and SOS1 were 2.5 nM, 500 uM, and 150 nM, respectively. The reaction plates were incubated at ambient temperature for 2 hours, a time estimated for complete GDP-GTP exchange in the absence of inhibitor. For the KRAS G12V mutant, similar guanine nucleotide exchange assays were used with 2.5nM as final concentration for the bodipy loaded KRAS proteins and 3 hours incubation after adding GppNHp-SOS1 mixture. A cyclic peptide described to selectively bind G12D mutant (Sakamoto et al., BBRC 484.3 (2017), 605-611) or internal compounds with confirmed binding were used as positive controls in the assay plates. Fluorescence intensities were measured on a PheraStar plate reader instrument (BMG Labtech) with excitation at 485 nm and emission at 520 nm.

The inhibitor potency of the exemplified compounds was determined in a fluorescence based guanine nucleotide exchange assay, which measures the exchange of bodipy-GDP (fluorescently labeled GDP) for GppNHp (Non-hydrolyzable GTP analog) to generate the active state of KRAS in the presence of SOS1 (guanine nucleotide exchange factor). Inhibitors were serially diluted in DMSO and a volume of 0.1 pL was transferred to the wells of a black low volume 384-well plate. 5 pL/well volume of bodipy-loaded KRAS G12C diluted to 5 nM in assay buffer (25 mM Hepes pH 7.5, 50 mM NaCI, 10 mM MgCh and 0.01 % Brij-35) was added to the plate and pre-incubated with inhibitor for 2 hours at ambient temperature. Appropriate controls (enzyme with no inhibitor or with a G12C inhibitor (AMG- 510)) were included on the plate. The exchange was initiated by the addition of a 5 pL/well volume containing 1 mM GppNHp and 300 nM SOS1 in assay buffer. The 10 pL/well reaction concentration of the bodipy-loaded KRAS G12C, GppNHp, and SOS1 were 2.5 nM, 500 uM, and 150 nM, respectively. The reaction plates were incubated at ambient temperature for 2 hours, a time estimated for complete GDP-GTP exchange in the absence of inhibitor. Internal compounds with confirmed binding were used as positive controls in the assay plates. Fluorescence intensities were measured on a PheraStar plate reader instrument (BMG Labtech) with excitation at 485 nm and emission at 520 nm.

Either GraphPad prism or XLfit was used to analyze the data. The IC50 values were derived by fitting the data to a four parameter logistic equation producing a sigmoidal doseresponse curve with a variable Hill coefficient. Prism equation: Y=Bottom + (Top- Bottom)/(1+10 A ((LoglC 5 o-X)*Hill slope)); XLfit equation: Y = (A+((B-A)/(1+((X/C) A D)))) where X is the logarithm of inhibitor concentration and Y is the response.

The KRAS_G12D exchange assay IC50 data and KRAS_G12D pERK assay IC50 data are provided in Table 1 below. The symbol “f” indicates IC50 100 nM, “ft” indicates IC50 > 100 nM but < 1 pM; and “ttt” indicates IC50 is >1 pM but < 5 pM, “tttt” indictes IC50 is >5 pM but < 10 pM. “N/A” indicates IC50 not available.

Table 1

The KRAS_G12V exchange assay IC50 data and KRAS_G12V pERK assay IC50 data are provided in Table 1 below. The symbol “f” indicates IC50 100 nM, “ft” indicates IC50 > 100 nM but < 1 pM; and “ttt” indicates IC50 is >1 pM but < 5 pM, “tttt” indictes IC50 is >5 pM but < 10 pM. “N/A” indicates IC50 not available.

Table 2

The KRAS_G12C exchange assay IC50 data and KRAS_G12C pERK assay IC50 data are provided in Table 1 below. The symbol “f” indicates IC50 100 nM, “ft” indicates IC50 > 100 nM but < 1 pM; and “ttt” indicates IC50 is >1 pM but < 5 pM, “tttt” indictes IC50 is >5 pM but < 10 pM. “N/A” indicates IC50 not available.

Table 3 Example B: Luminescent Viability Assay

MIA PaCa-2 (KRAS G12C; ATCC® CRL-1420), A427 (KRAS G12D; ATCC® HTB53) and NCI-H838 (KRAS WT; ATCC® CRL-5844) cells are cultured in RPMI 1640 media supplemented with 10% FBS (Gibco/Life Technologies). The cells are seeded (5x10 3 cells/well/in 50 uL) into black, clear bottomed 96-well Greiner tissue culture plates and cultured overnight at 37°C, 5% CO2. After overnight culture, 50 uL per well of serially diluted test compounds (2x final concentration) are added to the plates and incubated for 3 days. At the end of the assay, 100ul/well of CellTiter-Glo reagent (Promega) is added. Luminescence is read after 15 minutes with a TopCount (PerkinElmer). IC50 determination is performed by fitting the curve of percent inhibition versus the log of the inhibitor concentration using the GraphPad Prism 7 software.

Example C: Cellular pERK HTRF Assay

MIA PaCa-2 (KRAS G12C; ATCC® CRL-1420), A427 (KRAS G12D; ATCC® HTB53), HPAF-II (KRAS G12D; ATCC® CRL-1997) and NCI-H838 (KRAS WT; ATCC® CRL-5844) cells are purchased from ATCC and maintained in RPMI 1640 media supplemented with 10% FBS (Gibco/Life Technologies). The cells are plated at 5000 cells per well (8 uL) into Greiner 384-well low volume, flat-bottom, tissue culture treated white plates and incubated overnight at 37°C, 5% CO2. The next morning, test compound stock solutions are diluted in media at 3x the final concentration, and 4 uL are added to the cells. The plate is mixed by gentle rotation for 30 seconds (250rpm) at room temperature. The cells are incubated with the KRAS G12C and G12D compounds for 4 hours or 2 hours respectively at 37°C, 5% CO2.

4 uL of 4x lysis buffer with blocking reagent (1 :25) (Cisbio) are added to each well and plates are rotated gently (300 rpm) for 30 minutes at room temperature. 4 uL per well of Cisbio anti Phospho-ERK 1/2 d2 is mixed with anti Phospho-ERK 1/2 Cryptate (1:1) are added to each well, mixed by rotation and incubated overnight in the dark at room temperature. Plates are read on the Pherastar plate reader at 665 nm and 620 nm wavelengths. IC50 determination is performed by fitting the curve of inhibitor percent inhibition versus the log of the inhibitor concentration using the GraphPad Prism 7 software.

Example D: Whole Blood pERK1/2 HTRF Assay

MIA PaCa-2 cells (KRAS G12C; ATCC® CRL-1420) and HPAF-II (KRAS G12D; ATCC® CRL-1997) are maintained in RPMI 1640 with 10% FBS (Gibco/Life Technologies). The cells are seeded into 96 well tissue culture plates (Corning #3596) at 25000 cells per well in 100 uL media and cultured for 2 days at 37 °C, 5% CO2SO that they are approximately 80% confluent at the start of the assay. Whole Blood are added to the 1uL dots of compounds (prepared in DMSO) in 96 well plates and mixed gently by pipetting up and down so that the concentration of the compound in blood is 1x of desired concentration. The media is aspirated from the cells and 50 uL per well of whole blood with G12C or G12D compound is added and incubated for 4 or 2 hours respectively at 37 °C, 5% CO2. After dumping the blood, the plates are gently washed twice by adding PBS to the side of the wells and dumping the PBS from the plate onto a paper towel, tapping the plate to drain well. 50ul/well of 1x lysis buffer #1 (Cisbio) with blocking reagent (1 :25) (Cisbio) is then added and incubated at room temperature for 30 minutes with shaking (250 rpm). Following lysis, 16 uL of lysate is transferred into 384-well Greiner small volume white plate using an Assist Plus (Integra Biosciences, NH). 4uL of 1:1 mixture of anti Phospho-ERK 1/2 d2 and anti Phospho- ERK 1/2 Cryptate (Cisbio) is added to the wells using the Assist Plus and incubated at room temperature overnight in the dark. Plates are read on the Pherastar plate reader at 665 nm and 620 nm wavelengths. IC50 determination is performed by fitting the curve of inhibitor percent inhibition versus the log of the inhibitor concentration using the GraphPad Prism 7 software.

Example E: Ras Activation Elisa

The 96-Well Ras Activation ELISA Kit (Cell Biolabs Inc; #STA441) uses the Rafi RBD (Rho binding domain) bound to a 96-well plate to selectively pull down the active form of Ras from cell lysates. The captured GTP-Ras is then detected by a pan- Ras antibody and HRP-conjugated secondary antibody.

MIA PaCa-2 cells (KRAS G12C; ATCC® CRL-1420) and HPAF-II (KRAS G12D; ATCC® CRL-1997) are maintained in RPMI 1640 with 10% FBS (Gibco/Life Technologies). The cells are seeded into 96 well tissue culture plates (Corning #3596) at 25000 cells per well in 100 uL media and cultured for 2 days at 37 °C, 5% CO2SO that they are approximately 80% confluent at the start of the assay. The cells are treated with compounds for either 2 hours or overnight at 37 °C, 5% CO2. At the time of harvesting, the cells are washed with PBS, drained well and then lysed with 50 uL of the 1x Lysis buffer (provided by the kit) plus added Halt Protease and Phosphatase inhibitors (1:100) for 1 hour on ice.

The Raf-1 RBD is diluted 1:500 in Assay Diluent (provided in kit) and 100 pL of the diluted Raf-1 RBD is added to each well of the Raf-1 RBD Capture Plate. The plate is covered with a plate sealing film and incubated at room temperature for 1 hour on an orbital shaker. The plate is washed 3 times with 250 pL 1X Wash Buffer per well with thorough aspiration between each wash. 50 pL of Ras lysate sample (10-100 pg) is added per well in duplicate. A “no cell lysate” control is added in a couple of wells for background determination. 50 pL of Assay Diluent is added immediately to each well and the plate is incubated at room temperature for 1 hour on an orbital shaker. The plate is washed 5 times with 250 pL 1X Wash Buffer per well with thorough aspiration between each wash. 100 pL of the diluted Anti-pan-Ras Antibody is added to each well and the plate is incubated at room temperature for 1 hour on an orbital shaker. The plate is washed 5 times as previously. 100 pL of the diluted Secondary Antibody, HRP Conjugate is added to each well and the plate is incubated at room temperature for 1 hour on an orbital shaker. The plate is washed 5 times as previously and drained well. 100 pL of Chemiluminescent Reagent (provided in the kit) is added to each well, including the blank wells. The plate is incubated at room temperature for 5 minutes on an orbital shaker before the luminescence of each microwell is read on a plate luminometer. The % inhibition is calculated relative to the DMSO control wells after a background level of the “no lysate control” is subtracted from all the values. IC50 determination is performed by fitting the curve of inhibitor percent inhibition versus the log of the inhibitor concentration using the GraphPad Prism 7 software.

Example F: Inhibition of RAS-RAF and PI3K-AKT Pathways

The cellular potency of compounds is determined by measuring phosphorylation of KRAS downstream effectors extracellular-signal-regulated kinase (ERK), ribosomal S6 kinase (RSK), AKT (also known as protein kinase B, PKB) and downstream substrate S6 ribosomal protein.

To measure phosphorylated extracellular-signal-regulated kinase (ERK), ribosomal S6 kinase (RSK), AKT and S6 ribosomal protein, cells (details regarding the cell lines and types of data produced are further detailed in Table 4) are seeded overnight in Corning 96- well tissue culture treated plates in RPMI medium with 10% FBS at 4x10 4 cells/well. The following day, cells are incubated in the presence or absence of a concentration range of test compounds for 4 hours at 37 °C, 5% CO2. Cells are washed with PBS and lysed with 1x lysis buffer (Cisbio) with protease and phosphatase inhibitors. 10 pg of total protein lysates is subjected to SDS-PAGE and immunoblot analysis using following antibodies: phospho- ERK1/2-Thr202/Tyr204 (#9101 L), total-ERK1/2 (#9102L), phosphor-AKT-Ser473 (#4060L), phospho-p90RSK-Ser380 (#11989S) and phospho-S6 ribosomal protein-Ser235/Ser236 (#2211 S) are from Cell Signaling Technologies (Danvers, MA).

Table 4 Example G: In vivo efficacy studies

Mia-Paca-2 human pancreatic cancer cells are obtained from the American Type Culture Collection and maintained in RPMI media supplemented with 10% FBS. For efficacy studies experiments, 5 x 10 6 Mia-Paca-2 cells are inoculated subcutaneously into the right hind flank of 6- to 8-week-old BALB/c nude mice (Charles River Laboratories, Wilmington, MA, USA). When tumor volumes are approximately 150-250 mm3, mice are randomized by tumor volume and compounds are orally administered. Tumor volume is calculated using the formula (L x W 2 )^, where L and W refer to the length and width dimensions, respectively. Tumor growth inhibition is calculated using the formula (1 - (VT/VC)) X 100, where VT is the tumor volume of the treatment group on the last day of treatment, and Vc is the tumor volume of the control group on the last day of treatment. Two-way analysis of variance with Dunnett’s multiple comparisons test is used to determine statistical differences between treatment groups (GraphPad Prism). Mice are housed at 10-12 animals per cage, and are provided enrichment and exposed to 12-hour light/dark cycles. Mice whose tumor volumes exceeded limits (10% of body weight) are humanely euthanized by CO2 inhalation. Animals are maintained in a barrier facility fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care, International. All of the procedures are conducted in accordance with the US Public Service Policy on Human Care and Use of Laboratory Animals and with Incyte Animal Care and Use Committee Guidelines.

Various modifications of the present disclosure, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including without limitation all patent, patent applications, and publications, cited in the present application is incorporated herein by reference in its entirety.