Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
6-SUBSTITUTED INDOLE COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2022/140325
Kind Code:
A1
Abstract:
The present disclosure relates generally to certain compounds, pharmaceutical compositions comprising said compounds, and methods of making and using said compounds and pharmaceutical compositions. The compounds and compositions provided herein may be used for the treatment or prevention of an autoimmune disease and/or inflammatory condition, including systemic lupus erythematosus and cutaneous lupus erythematosus.

Inventors:
AMMANN STEPHEN E (US)
CANALES EDA Y (US)
CHANG WENG K (US)
LAZERWITH SCOTT E (US)
SHORE DANIEL G (US)
Application Number:
PCT/US2021/064516
Publication Date:
June 30, 2022
Filing Date:
December 21, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GILEAD SCIENCES INC (US)
International Classes:
C07D401/04; A61K31/437; A61P17/06; A61P37/00; C07D401/14; C07D405/14; C07D413/14; C07D417/14; C07D451/04; C07D471/04; C07D487/04; C07D495/04; C07D498/10
Domestic Patent References:
WO2017178844A12017-10-19
WO2017075694A12017-05-11
WO2013052263A22013-04-11
WO2019126253A12019-06-27
WO2019126113A12019-06-27
WO2018026620A12018-02-08
WO2022040293A12022-02-24
Foreign References:
US20150236272A12015-08-20
US6329412B12001-12-11
US20070112048A12007-05-17
US20080194668A12008-08-14
US6486153B12002-11-26
US3845770A1974-11-05
US4326525A1982-04-27
US4902514A1990-02-20
US5616345A1997-04-01
US5023252A1991-06-11
US4992445A1991-02-12
US5001139A1991-03-19
Other References:
ZHOU JIA ET AL.: "Remote C6-Enantioselective C-H Functionalization of 2,3-Disubstituted Indoles through the Dual H-Bonds and [pi]-[pi] Interaction Strategy Enabled by CPAs", ORG. LETT., 22 October 2019 (2019-10-22), pages 8662 - 8666, XP055905040, Retrieved from the Internet [retrieved on 20220324], DOI: 10.1021/acs.orglett.9b03276
DUNCAN ET AL.: "Supporting Information Discovery and optimization of small molecule ligands for the CBP/p300 bromodomains", J. AM. CHEM. SOC. 2014, 136, 26, 9308-9319, 19 June 2014 (2014-06-19), pages S1 - S99, XP055905070, Retrieved from the Internet [retrieved on 20220324]
MARSILJE ET AL.: "Optimization of small molecule agonists of the thrombopoietin (Tpo) receptor derived from a benzo[a]carbazole hit scaffold", BIOORG. MED. CHEM. LETT., vol. 18, no. 19, 2008, pages 5259 - 5262, XP025433913, DOI: 10.1016/J.BMCL.2008.08.077
NGUYEN ET AL.: "Synthesis and antibacterial evaluation of new, unsymmetrical triaryl bisamidine compounds", BIOORG. MED. CHEM. LETT., vol. 24, no. 15, 12 June 2014 (2014-06-12), pages 3366 - 3372, XP028864140, DOI: 10.1016/J.BMCL.2014.05.094
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 19 April 2007 (2007-04-19), KOSEKI JUNICHI ET AL.: "Preparation of indolylquinoxaline compounds as VEGF inhibitors and pharmaceutical compositions containing them", XP055906506, retrieved from STN Database accession no. 2007:431922
"Remington's Pharmaceutical Sciences", 1985, MACE PUBLISHING CO.
GUYATT ET AL., GASTROENTEROLOGY, vol. 96, 1989, pages 804 - 810
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis", 2006, WILEY
Attorney, Agent or Firm:
CHOI, Lydia B. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS: 1. A compound of Formula I, Formula I or a pharmaceutically acceptable salt thereof, wherein R1 is 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-4 Ra groups; R2 is H, -CN, C1-6 alkyl, or C3-7 monocyclic cycloalkyl, wherein the C1-6 alkyl is optionally substituted with 1-4 Rb groups, wherein the C3-7 monocyclic cycloalkyl is optionally substituted with 1-4 Ra groups; X1 and X3 are each independently N or CR3; each R3 independently is H, halogen, C1-6 alkyl, C3-6 monocyclic cycloalkyl, or -O(C1-4 alkyl), wherein the C1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR4R4, and C1-4 alkoxy; X2 is N or CH; Y is C1-10 alkyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, or L1, wherein the C1-10 alkyl and C2-6 alkynyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 Rb groups, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 Ra groups; L1 is -OR5, -C(O)R5, -C(O)N(R5)(R5), -NR5R5, -N(R5)2(R5)+, -N(R5)C(O)R5, -N(R5)C(O)OR5, -N(R5)C(O)N(R5)(R5), -N(R5)S(O)2(R5a), -NR5S(O)2N(R5)(R5), -NR5S(O)2O(R5a), -OC(O)N(R5)(R5), -SR5, -S(O)R5a, -S(O)(NH)R5, -S(O)2R5a, -S(O)2N(R5)(R5), or -N=S(R5a)(R5a)=O; Z is C1-6 alkyl, -NR6R7, -C(O)R13, -C(O)NR6R7,-S(O)2R6, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl is optionally substituted with 1-4 Rb groups; wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R8 groups and are each independently optionally substituted with 1-3 Ra groups; R6 is C1-6 alkyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl is optionally substituted with 1-4 Rb groups, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 Ra groups; R13 is C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6- 10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 Ra groups; each R4 and R7 independently is H or C1-3 alkyl; each R8 independently is halogen, -C(O)R9, -NR10R10, C1-6 alkyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR5, -C(O)OR5, -C(O)N(R5)(R5), -N(R5)2(R5)+, -N(R5)C(O)R5, -N(R5)C(O)OR5, -N(R5)C(O)N(R5)(R5), -N(R5)S(O)2(R5a), -NR5S(O)2N(R5)(R5), -NR5S(O)2O(R5a), -OC(O)R5, -OC(O)OR5, -OC(O)N(R5)(R5), -SR5, -S(O)R5a, -S(O)(NH)R5, -S(O)2R5a, -S(O)2N(R5)(R5), or -N=S(R5a)(R5a)=O, wherein the C1-6 alkyl is optionally substituted with 1-4 Rb groups, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 Ra groups; R9 is C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C2-6 alkenyl and C2-6 alkynyl are each independently optionally substituted with 1-4 Rb groups, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 Ra groups; each R5 and R10 independently is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6- 10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each independently optionally substituted with 1-4 Rb groups, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 Ra groups; each R5a independently is C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each independently optionally substituted with 1-4 Rb groups, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 Ra groups; each Ra independently is oxo, imino, halogen, -NO2, -N3, -CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR11, -C(O)R11, -C(O)OR11, -C(O)N(R11)(R11), -NR11R11, -N(R11)2(R11)+, -N(R11)C(O)R11, -N(R11)C(O)OR11, -N(R11)C(O)N(R11)(R11), -N(R11)S(O)2(R11a), -NR11S(O)2N(R11)(R11), -NR11S(O)2O(R11a), -OC(O)R11, -OC(O)OR11, -OC(O)N(R11)(R11), -SR11, -S(O)R11a, -S(O)(NH)R11, -S(O)2R11a, -S(O)2N(R11)(R11), or -N=S(R11a)(R11a)=O, wherein the C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 Rc groups, each Rb independently is oxo, imino, halogen, -NO2, -N3, -CN, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR11, -C(O)R11, -C(O)OR11, -C(O)N(R11)(R11), -NR11R11, -N(R11)2(R11)+, -N(R11)C(O)R11, -N(R11)C(O)OR11, -N(R11)C(O)N(R11)(R11), -N(R11)S(O)2(R11a), -NR11S(O)2N(R11)(R11), -NR11S(O)2O(R11a), -OC(O)R11, -OC(O)OR11, -OC(O)N(R11)(R11), -SR11, -S(O)R11a, -S(O)(NH)R11, -S(O)2R11a, -S(O)2N(R11)(R11), or -N=S(R11a)(R11a)=O, wherein the C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 Rc groups; each Rc independently is halogen, -CN, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR12, -C(O)R12, -C(O)OR12, -C(O)N(R12)(R12), -NR12R12, -N(R12)2(R12)+, -N(R12)C(O)R12, -N(R12)C(O)OR12, -N(R12)C(O)N(R12)(R12), -N(R12)S(O)2(R12a), -NR12S(O)2N(R12)(R12), -NR12S(O)2O(R12a), -OC(O)R12, -OC(O)OR12, -OC(O)N(R12)(R12), -SR12, -S(O)R12a, -S(O)(NH)R12, -S(O)2R12a, -S(O)2N(R12)(R12), or -N=S(R12a)(R12a)=O; each R11 independently is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 Rc groups; each R11a independently is C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 Rc groups; each R12 independently is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; each R12a independently is C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. 2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 Ra groups; R2 is -CN, C1-6 alkyl, or C3-5 monocyclic cycloalkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 Rb groups, wherein the C3-5 monocyclic cycloalkyl is optionally substituted with 1-3 Ra groups; X1 is N or CR3; X3 is CR3; each R3 independently is H, halogen, or C1-3 alkyl; X2 is CH; Y is C2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8- 10 membered fused bicyclic heteroaryl, or L1, wherein the C2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 Ra groups; L1 is -C(O)N(R5)(R5); each R5 independently is H or 4-7 membered monocyclic heterocyclyl; Z is C1-6 alkyl, -NR6R7, -C(O)R13, -S(O)2R6, 5-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl is optionally substituted with 1-3 Rb groups, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R8 groups and are each independently optionally substituted with 1-3 Ra groups; R6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 Ra groups; R13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 Ra groups; each R8 independently is halogen, C1-6 alkyl, -C(O)R9, -NR10R10, 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C1-6 alkyl is optionally substituted with 1-3 Rb groups, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 Ra groups; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. 3. The compound of any one of claims 1-2, or a pharmaceutically acceptable salt thereof, wherein R1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, wherein the C1-5 alkyl is optionally substituted with 1-3 Rc groups; R2 is -CN, C1-6 alkyl, or C3-5 monocyclic cycloalkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C1-3 alkoxy; X1 is N or CR3; X3 is CR3; each R3 independently is H, halogen, or C1-3 alkyl; X2 is CH; Y is C2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8- 10 membered fused bicyclic heteroaryl, or L1, wherein the C2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, wherein the C1-5 alkyl is optionally substituted with 1-3 Rc groups; L1 is -C(O)N(R5)(R5); each R5 independently is H or 4-7 membered monocyclic heterocyclyl; Z is C1-6 alkyl, -NR6R7, -C(O)R13, -S(O)2R6, 5-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C1-6 alkyl is optionally substituted with one Rb group, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl; R6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl; R13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl; each R8 independently is halogen, C1-6 alkyl, -C(O)R9, -NR10R10, 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R11)(R11), -NR11R11, and C1-4 alkoxy, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl; R9 is 4-7 membered monocyclic heterocyclyl or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl; each R11 independently is H or C1-4 alkyl, wherein the C1-4 alkyl is optionally substituted with one group selected from -OH and -NR12R12; Rb is -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, C3-7 monocyclic cycloalkyl, C7-10 fused bicyclic cycloalkyl, C5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; each Rc independently is -OH, halogen, -CN, -NR12R12, or C1-4 alkoxy; each R12 independently is H or C1-3 alkyl; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. 4. The compound of any one of claims 1-3, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinolinyl, each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, wherein the C1-5 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR12R12, and C1-4 alkoxy. 5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein R1 is substituted with 1-3 groups independently selected from -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl, wherein the C1-3 alkyl is optionally substituted with 1-3 halogen. 6. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt thereof, wherein R1 is substituted with 1-3 groups independently selected from -CN, oxo, -NH2, methoxy, methyl, and -CF3. 7. The compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein R1 is pyridinyl substituted with 1-3 groups independently selected from -CN, methoxy, methyl, and -NH2. 8. The compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein R1 is 9. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, wherein R1 is . 10. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein R2 is -CN, C1-4 alkyl, or cyclopropyl, wherein the C1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C1-3 alkoxy and wherein the cyclopropyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, C1-3 alkoxy, and C1-3 alkyl. 11. The compound of any one of claims 1-10, or a pharmaceutically acceptable salt thereof, wherein R2 is -CN, -C(O)CH3, methyl, ethyl, isopropyl, or cyclopropyl. 12. The compound of any one of claims 1-10, or a pharmaceutically acceptable salt thereof, wherein R2 is C1-3 alkyl. 13. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt thereof, wherein R2 is methyl. 14. The compound of any one of claims 1-13, or a pharmaceutically acceptable salt thereof, wherein X1 is N or CH. 15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt thereof, wherein X3 is CH. 16. The compound of any one of claims 1-15, or a pharmaceutically acceptable salt thereof, wherein X1 and X3 are CH. 17. The compound of any one of claims 1-16, or a pharmaceutically acceptable salt thereof, wherein Y is C2 alkynyl and Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-4 alkoxy, and C1-5 alkyl. 18. The compound of any one of claims 1-17, or a pharmaceutically acceptable salt thereof, wherein Y is C2 alkynyl and Z is piperazinyl. 19. The compound of any one of claims 1-16, or a pharmaceutically acceptable salt thereof, wherein Y is 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, and wherein the C1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR12R12, and C1-4 alkoxy. 20. The compound of any one of claims 1-16 and 19, or a pharmaceutically acceptable salt thereof, wherein Y is pyrrolidinyl, piperidinyl, pyrazolyl, pyridinyl, quinolinyl, triazolyl, pyrimidinyl, thiazolyl, each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl, wherein the C1-3 alkyl is optionally substituted with -NH2. 21. The compound of any one of claims 1-16 and 19-20, or a pharmaceutically acceptable salt thereof, wherein Y is optionally substituted with 1-2 groups independently selected from -CH2CH2NH2, methoxy, -N(CH3)2, -NH2, -N(CH3)CH2CH2NHCH3, and methyl. 22. The compound of any one of claims 1-16 and 19-21, or a pharmaceutically acceptable salt thereof, wherein Y is piperidinyl. 23. The compound of any one of claims 1-16 and 19-21, or a pharmaceutically acceptable salt thereof, wherein Y is ,

24. The compound of any one of claims 1-16, or a pharmaceutically acceptable salt thereof, wherein Y is 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each substituted with one Z group and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, and wherein the C1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR12R12, and C1-4 alkoxy. 25. The compound of any one of claims 1-16 and 24, or a pharmaceutically acceptable salt thereof, wherein Y is phenyl, pyrazolyl, triazolyl, thiazolyl, pyridinyl, pyrimidinyl, or , each of which is substituted with one Z group and independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 26. The compound of any one of claims 1-16 and 24-25, or a pharmaceutically acceptable salt thereof, wherein Y is substituted with one Z group and is optionally substituted with C1-3 alkyl. 27. The compound of any one of claims 1-16 and 24-26, or a pharmaceutically acceptable salt thereof, wherein Y is substituted with one Z group and is optionally substituted with methyl. 28. The compound of any one of claims 1-16 and 24-25, or a pharmaceutically acceptable salt thereof, wherein Y is pyridinyl, wherein the pyridinyl is substituted with one Z group and is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 29. The compound of any one of claims 1-16 and 24-25, or a pharmaceutically acceptable salt thereof, wherein Y is phenyl, wherein the phenyl is substituted with one Z group and is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 30. The compound of any one of claims 1-16 and 24-25, or a pharmaceutically acceptable salt thereof, wherein Y substituted with Z is , each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 31. The compound of any one of claims 1-16 and 24-30, or a pharmaceutically acceptable salt thereof, wherein Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl. 32. The compound of any one of claims 1-16 and 24-31, or a pharmaceutically acceptable salt thereof, wherein Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl each independently have one or two ring heteroatoms that is N. 33. The compound of any one of claims 1-16 and 24-32, or a pharmaceutically acceptable salt thereof, wherein Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-5 alkyl.

34. The compound of any one of claims 1-16 and 24-33, or a pharmaceutically acceptable salt thereof, wherein Z is each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 35. The compound of any one of claims 1-16 and 24-34, or a pharmaceutically acceptable salt thereof, wherein Z is optionally substituted with -NH2. 36. The compound of any one of claims 1-16 and 24-35, or a pharmaceutically acceptable salt thereof, wherein Z is piperazinyl. 37. The compound of any one of claims 1-16 and 24-30, or a pharmaceutically acceptable salt thereof, wherein Z is C1-6 alkyl, -NR6R7, -C(O)R13, or -S(O)2R6, wherein the C1-6 alkyl is optionally substituted with one group selected from 4-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl. 38. The compound of any one of claims 1-16, 24-30, and 37, or a pharmaceutically acceptable salt thereof, wherein Z is -NHR6, -N(CH3)R6, -C(O)R13, -S(O)2R6, or methyl substituted with piperazinyl. 39. The compound of any one of claims 1-16, 24-27, and 29-30, or a pharmaceutically acceptable salt thereof, wherein Y is phenyl and Z is -C(O)R13. 40. The compound of any one of claims 1-16 and 24-30, or a pharmaceutically acceptable salt thereof, wherein Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each substituted with 1-2 R8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl. 41. The compound of any one of claims 1-16, 24-30, and 40, or a pharmaceutically acceptable salt thereof, wherein Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each substituted with 1-2 R8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl each independently have one or two ring heteroatoms that is N. 42. The compound of any one of claims 1-16, 24-30, and 40-41, or a pharmaceutically acceptable salt thereof, wherein Z is 5-7 membered monocyclic heterocyclyl or 8-10 membered fused bicyclic heterocyclyl, wherein the 5-7 membered heterocyclyl and 8-10 membered fused bicyclic heterocyclyl are each substituted with 1-2 R8 groups and are each independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl. 43. The compound of any one of claims 1-16, 24-30, and 40-41, or a pharmaceutically acceptable salt thereof, wherein Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, , each of which is substituted with 1-2 R8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl 44. The compound of any one of claims 1-16, 24-30, and 40-43, or a pharmaceutically acceptable salt thereof, wherein Z is pyrrolidinyl, piperidinyl, or piperazinyl, each of which is substituted with 1-2 R8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 45. The compound of any one of claims 1-16, 24-30, and 40-43, or a pharmaceutically acceptable salt thereof, wherein Z is each of which is substituted with 1-2 R8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 46. The compound of any one of claims 1-16, 24-30, 40-43, and 45, or a pharmaceutically acceptable salt thereof, wherein Z substituted with 1-2 R8 groups is: each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 47. The compound of any one of claims 1-16, 24-30, and 40-46, or a pharmaceutically acceptable salt thereof, wherein Z substituted with 1-2 R8 groups is: . 48. The compound of any one of claims 1-16, 24-30, and 40-47, or a pharmaceutically acceptable salt thereof, wherein R13 and each R8 independently is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl. 49. The compound of any one of claims 1-16, 24-30, and 40-48, or a pharmaceutically acceptable salt thereof, wherein R13 and each R8 independently is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-4 alkoxy, and C1-5 alkyl, wherein the 4-membered monocyclic heterocyclyl has one ring heteroatom that is O, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl each independently have one or two ring heteroatoms that is N. 50. The compound of any one of claims 1-16, 24-30, and 40-49, or a pharmaceutically acceptable salt thereof, wherein R13 and each R8 independently is oxetanyl, pyrrolidinyl, piperidinyl, piperazinyl, , each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 51. The compound of any one of claims 1-16, 24-30, and 40-50, or a pharmaceutically acceptable salt thereof, wherein R13 and each R8 independently is optionally substituted with one -NH2. 52. The compound of any one of claims 1-16, 24-30, and 40-51, or a pharmaceutically acceptable salt thereof, wherein R13 and each R8 independently is . 53. The compound of any one of claims 1-16, 24-30, and 40-47, or a pharmaceutically acceptable salt thereof, wherein each R8 independently is halogen, C1-6 alkyl, -C(O)R9, or -NR10R10, wherein the C1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R11)(R11), -NR11R11, and C1-4 alkoxy. 54. The compound of any one of claims 1-16, 24-30, 40-47, and 53, or a pharmaceutically acceptable salt thereof, wherein each R8 independently is -F, -NH2, methyl, 55. The compound of any one of claims 1-16, 24-30, 40-47, and 53, or a pharmaceutically acceptable salt thereof, wherein one R8 is -C(O)R9. 56. The compound of any one of claims 1-16, 24-30, 40-47, and 55, or a pharmaceutically acceptable salt thereof, wherein one R8 is C1-3 alkyl optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR11R11, and C1-3 alkoxy. 57. The compound of any one of claims 1-16, 24-30, 40-47, and 55, or a pharmaceutically acceptable salt thereof, wherein R9 is 5-7 membered monocyclic heterocyclyl or 6-10 membered bridged bicyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 58. The compound of any one of claims 1-16, 24-30, 40-47, 55, and 57, or a pharmaceutically acceptable salt thereof, wherein R9 is pyrrolidinyl, morpholinyl, or , each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR11R11, C1-3 alkoxy, and C1-3 alkyl. 59. The compound of any one of claims 1-16, 24-30, 40-47, and 55, or a pharmaceutically acceptable salt thereof, wherein R8 is 60. The compound of any one of claims 1-5, 17, 19-20, 24-25, 28-34, 40-41, 43-46, 48-50, 53, and 56-58, or a pharmaceutically acceptable salt thereof, wherein each R11 independently is H or methyl. 61. The compound of any one of claims 1-16, or a pharmaceutically acceptable salt thereof, wherein each R5 independently is H or 6 membered monocyclic heterocyclyl, wherein the 6 membered monocyclic heterocyclyl has 1-2 ring heteroatoms independently selected from N, O and S. 62. The compound of any one of claims 1-16 or 61, or a pharmaceutically acceptable salt thereof, wherein L1 is

or a pharmaceutically acceptable salt thereof. 64. A compound selected from the group consisting of: ,

, or a pharmaceutically acceptable salt thereof. 65. A compound selected from the group consisting of: , or a pharmaceutically acceptable salt thereof. 66. A compound selected from the group consisting of:

, or a pharmaceutically acceptable salt thereof. 67. A compound selected from the group consisting of:

68. A compound that is or a pharmaceutically acceptable salt thereof. 69. A pharmaceutical composition comprising the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. 70. The pharmaceutical composition of claim 69, further comprising one or more additional therapeutic agents, or a pharmaceutically acceptable salt thereof. 71. The pharmaceutical composition of claim 70, wherein the one or more additional therapeutic agents comprises an anti-malarial agent. 72. The pharmaceutical composition of claim 71, wherein the anti-malarial agent is selected from chloroquine and hydroxychloroquine, or a pharmaceutically acceptable salt thereof. 73. A method of inhibiting toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 74. A method of inhibiting toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 75. A method of inhibiting toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 76. A method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the pharmaceutical compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the composition of any one of claims 69-72.

77. A method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 78. A method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 79. A method of treating an inflammatory condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 80. The method of claim 79, wherein the inflammatory condition is selected from inflammatory bowel disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, glomerulonephritis, mixed connective tissue disease (MCTD), dermatomyositis, polymyositis, systemic sclerosis, antineutrophil cytoplasmic antibody-associated vasculitis, anti-phospholipid syndrome, autoimmune hemolytic anemia, macrophage activation syndrome driven inflammatory anemia, IgA nephropathy, type I diabetes, non-alcoholic steatohepatitis, and Sjogren’s syndrome. 81. A method of treating systemic lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 82. A method of treating cutaneous lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72. 83. A method of treating lupus nephritis in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition of any one of claims 69-72 84. The method of any one of claims 73-83, further comprising administering a therapeutically effective amount of one or more additional therapeutic agents, or a pharmaceutically acceptable salt thereof. 85. The method of claim 84, wherein the one or more additional therapeutic agents is selected from the group consisting of veltuzumab, PF-06835375, eculizumab, milatuzumab, SM-06, SM-03, BT-063, QX-006-N, BOS-161721, AK-101, TNX-1500, theralizumab, daxdilimab, TAK-079, felzartamab, itolizumab, anifrolumab, iscalimab, dapirolizumab pegol, lanalumab, LY-3361237, JNJ-55920839, UBP-1213, DS-7011, PFI-102, BIIB-059, obexelimab, talacotuzumab, vobarilizumab, TE-2324, PRV-3279, chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, AVO-101, rozibafusp alfa, VRN-02, annexuzlimab, ALPN-101, bendamustine hydrochloride, BMS-986256, NKTR-35, atacicept, telitacicept, BMS-986256, M-5049, KZR-616, KPG-818, verdinexor, ALPN-303, valziflocept, LA-1, cenerimod, prednisone, corticotropin, deucravacitinib, CPL-409116, CS-12192, tofacitinib citrate, ISB-830, DV-1079, julemic acid, iberdomide, TAM-01, BML-258, brepocitinib, SDC-1801, SDC-1802, ICP-330, NTR-441, dalazatide, GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, HMPL-523, RSLV-132, interleukin-2 follow-on biologic, interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, CUG-252, DZ- 2002, PEGylated HLA-x (SLE), AC-0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, orelabrutinib, DWP-213388, INV-103, R-salbutamol sulphate, anchorins, NIK-SMI1, X-6, INV-17, Oshadi D, baricitinib, upadacitinib, filgotinib, itacitinib, INCB-54707, delgocitinib, DWP-212525, CKD-971, as mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT-013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, and AMG-592, or a pharmaceutically acceptable salt of any of the foregoing, or any combination thereof. 86. The method of any one of claims 73-85, wherein the subject is a human. 87. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in therapy. 88. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of inhibiting toll- like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 89. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of inhibiting toll- like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 90. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of inhibiting toll- like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 91. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 92. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 93. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition.

94. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating an inflammatory condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 95. The use of claim 94, wherein the inflammatory condition is selected from inflammatory bowel disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, glomerulonephritis, mixed connective tissue disease (MCTD), dermatomyositis, polymyositis, systemic sclerosis, antineutrophil cytoplasmic antibody-associated vasculitis, anti-phospholipid syndrome, autoimmune hemolytic anemia, macrophage activation syndrome driven inflammatory anemia, IgA nephropathy, type I diabetes, non-alcoholic steatohepatitis, and Sjogren’s syndrome. 96. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating systemic lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 97. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating cutaneous lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 98. A compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 69-72 for use in a method of treating lupus nephritis in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. 99. The use of any one of claims 87-98, further comprising administering one or more additional therapeutic agents. 100. The use of claim 99, wherein the one or more additional therapeutic agents is selected from the group consisting of veltuzumab, PF-06835375, eculizumab, milatuzumab, SM-06, SM- 03, BT-063, QX-006-N, BOS-161721, AK-101, TNX-1500, theralizumab, daxdilimab, TAK- 079, felzartamab, itolizumab, anifrolumab, iscalimab, dapirolizumab pegol, lanalumab, LY- 3361237, JNJ-55920839, UBP-1213, DS-7011, PFI-102, BIIB-059, obexelimab, talacotuzumab, vobarilizumab, TE-2324, PRV-3279, chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, AVO-101, rozibafusp alfa, VRN-02, annexuzlimab, ALPN- 101, bendamustine hydrochloride, BMS-986256, NKTR-35, atacicept, telitacicept, BMS- 986256, M-5049, KZR-616, KPG-818, verdinexor, ALPN-303, valziflocept, LA-1, cenerimod, prednisone, corticotropin, deucravacitinib, CPL-409116, CS-12192, tofacitinib citrate, ISB-830, DV-1079, julemic acid, iberdomide, TAM-01, BML-258, brepocitinib, SDC-1801, SDC-1802, ICP-330, NTR-441, dalazatide, GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, HMPL-523, RSLV-132, interleukin-2 follow-on biologic, interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, CUG-252, DZ-2002, PEGylated HLA-x (SLE), AC- 0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, orelabrutinib, DWP-213388, INV-103, R-salbutamol sulphate, anchorins, NIK-SMI1, X-6, INV- 17, Oshadi D, baricitinib, upadacitinib, filgotinib, itacitinib, INCB-54707, delgocitinib, DWP- 212525, CKD-971, as mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT-013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, and AMG-592, or a pharmaceutically acceptable salt thereof. 101. The use of any one of claims 87-100, wherein the subject is a human.

Description:
6-SUBSTITUTED INDOLE COMPOUNDS CROSS REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to U.S. Provisional Application No.63/129,223, filed December 22, 2020, which is incorporated herein in its entirety for all purposes. FIELD [0002] This disclosure relates generally to novel 6-substituted indole compounds, pharmaceutical compositions comprising said compounds, and methods of making and using said compounds and pharmaceutical compositions. In some embodiments, the novel 6- substituted indole compounds provided herein may be used in the treatment of certain diseases and disorders, including, but not limited to, an inflammatory condition, systemic lupus erythematosus, cutaneous lupus erythematosus, or lupus nephritis. BACKGROUND [0003] Toll-like receptors (TLRs) are a family of transmembrane immune receptors that sense pathogens, trigger innate immune responses and prime adaptive immunity. TLR7/8/9 are endosomally localized TLRs that respond to single-stranded RNAs (TLR7/8) or unmethylated DNA containing cytosine-phosphate-guanine (CpG) motifs (TLR9). Activation of TLR7/8/9 leads to inflammatory responses including the production of Type I interferons and proinflammatory cytokines, activation of B cells and antibody production, and neutrophil NETosis. Aberrant activation of TLR7/8/9 contributes to elevated Type I interferon response, increased pro-inflammatory cytokines, and sustained autoantibody production that may fuel the chronic progression of a variety of autoimmune disease and inflammatory conditions leading to broad inflammation and tissue damage. . (Kawai et al., 2010, Nat Immunol 11, 373; Joosten et al., 2016, Nat Rev Rheomatol 12, 344; Crow et al., 2019, Lupus Sci Med 6, e000336; Garcia- Romo et al., 2011, Sci Transl Med 3, 73ra20; Kono et al., 2009, PNAS 106, 12061; Koh et al., 2013, J Immunol 190, 4982). Therefore, there is a need for compounds that are potent TLR7, and/or TLR8, and/or TLR9 antagonists that are stable and exhibit effective pharmacokinetic and/or pharmacodynamic profiles. SUMMARY [0004] In one embodiment, provided herein is a compound of Formula I, Formula I or a pharmaceutically acceptable salt thereof, wherein R 1 is 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-4 R a groups; R 2 is H, -CN, C 1-6 alkyl, or C 3-7 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups; X 1 and X 3 are each independently N or CR 3 ; each R 3 independently is H, halogen, C 1-6 alkyl, C 3-6 monocyclic cycloalkyl, or -O(C 1-4 alkyl), wherein the C 1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 4 R 4 , and C 1-4 alkoxy; X 2 is N or CH; Y is C 1-10 alkyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, or L 1 , wherein the C 1-10 alkyl and C 2-6 alkynyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R a groups; L 1 is -OR 5 , -C(O)R 5 , -C(O)N(R 5 )(R 5 ), -NR 5 R 5 , -N(R 5 ) 2 (R 5 ) + , -N(R 5 )C(O)R 5 , -N(R 5 )C(O)OR 5 , -N(R 5 )C(O)N(R 5 )(R 5 ), -N(R 5 )S(O) 2 (R 5a ), -NR 5 S(O) 2 N(R 5 )(R 5 ), -NR 5 S(O) 2 O(R 5a ), -OC(O)N(R 5 )(R 5 ), -SR 5 , -S(O)R 5a , -S(O)(NH)R 5 , -S(O) 2 R 5a , -S(O) 2 N(R 5 )(R 5 ), or -N=S(R 5a )(R 5a )=O; Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -C(O)NR 6 R 7 ,-S(O) 2 R 6 , C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups; wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 R a groups; R 6 is C 1-6 alkyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; R 13 is C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6- 10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R 4 and R 7 independently is H or C 1-3 alkyl; each R 8 independently is halogen, -C(O)R 9 , -NR 10 R 10 , C 1-6 alkyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 5 , -C(O)OR 5 , -C(O)N(R 5 )(R 5 ), -N(R 5 ) 2 (R 5 ) + , -N(R 5 )C(O)R 5 , -N(R 5 )C(O)OR 5 , -N(R 5 )C(O)N(R 5 )(R 5 ), -N(R 5 )S(O) 2 (R 5a ), -NR 5 S(O) 2 N(R 5 )(R 5 ), -NR 5 S(O) 2 O(R 5a ), -OC(O)R 5 , -OC(O)OR 5 , -OC(O)N(R 5 )(R 5 ), -SR 5 , -S(O)R 5a , -S(O)(NH)R 5 , -S(O) 2 R 5a , -S(O) 2 N(R 5 )(R 5 ), or -N=S(R 5a )(R 5a )=O, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; R 9 is C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 2-6 alkenyl and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R 5 and R 10 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6- 10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R 5a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R a independently is oxo, imino, halogen, -NO 2 , -N 3 , -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , -N(R 11 ) 2 (R 11 ) + , -N(R 11 )C(O)R 11 , -N(R 11 )C(O)OR 11 , -N(R 11 )C(O)N(R 11 )(R 11 ), -N(R 11 )S(O) 2 (R 11a ), -NR 11 S(O) 2 N(R 11 )(R 11 ), -NR 11 S(O) 2 O(R 11a ), -OC(O)R 11 , -OC(O)OR 11 , -OC(O)N(R 11 )(R 11 ), -SR 11 , -S(O)R 11a , -S(O)(NH)R 11 , -S(O) 2 R 11a , -S(O) 2 N(R 11 )(R 11 ), or -N=S(R 11a )(R 11a )=O, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups, each R b independently is oxo, imino, halogen, -NO2, -N3, -CN, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , -N(R 11 ) 2 (R 11 ) + , -N(R 11 )C(O)R 11 , -N(R 11 )C(O)OR 11 , -N(R 11 )C(O)N(R 11 )(R 11 ), -N(R 11 )S(O) 2 (R 11a ), -NR 11 S(O) 2 N(R 11 )(R 11 ), -NR 11 S(O) 2 O(R 11a ), -OC(O)R 11 , -OC(O)OR 11 , -OC(O)N(R 11 )(R 11 ), -SR 11 , -S(O)R 11a , -S(O)(NH)R 11 , -S(O) 2 R 11a , -S(O) 2 N(R 11 )(R 11 ), or -N=S(R 11a )(R 11a )=O, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups; each R c independently is halogen, -CN, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 12 , -C(O)R 12 , -C(O)OR 12 , -C(O)N(R 12 )(R 12 ), -NR 12 R 12 , -N(R 12 ) 2 (R 12 ) + , -N(R 12 )C(O)R 12 , -N(R 12 )C(O)OR 12 , -N(R 12 )C(O)N(R 12 )(R 12 ), -N(R 12 )S(O) 2 (R 12a ), -NR 12 S(O) 2 N(R 12 )(R 12 ), -NR 12 S(O) 2 O(R 12a ), -OC(O)R 12 , -OC(O)OR 12 , -OC(O)N(R 12 )(R 12 ), -SR 12 , -S(O)R 12a , -S(O)(NH)R 12 , -S(O) 2 R 12a , -S(O) 2 N(R 12 )(R 12 ), or -N=S(R 12a )(R 12a )=O; each R 11 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups; each R 11a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups; each R 12 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; each R 12a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0005] In one embodiment, provided herein are pharmaceutical composition comprising a compound provided herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. [0006] In one embodiment, provided herein is a method of inhibiting toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0007] In one embodiment, provided herein is a method of inhibiting toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0008] In one embodiment, provided herein is a method of inhibiting toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0009] In one embodiment, provided herein is a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0010] In one embodiment, provided herein is a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0011] In one embodiment, provided herein is a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0012] In one embodiment, provided herein is a method of treating an inflammatory condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0013] In one embodiment, provided herein is a method of treating systemic lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0014] In one embodiment, provided herein is a method of treating cutaneous lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0015] In one embodiment, provided herein is a method of treating lupus nephritis in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0016] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in therapy. [0017] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of inhibiting toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0018] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of inhibiting toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0019] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of inhibiting toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0020] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0021] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0022] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0023] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of treating an inflammatory condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0024] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of treating systemic lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition. [0025] In one embodiment, provided herein is a compound provided herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in a method of treating cutaneous lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound, or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of the pharmaceutical composition therapeutically effective amount of the pharmaceutical composition. DETAILED DESCRIPTION I. Definitions [0026] The description below is made with the understanding that the present disclosure is to be considered as an exemplification of the claimed subject matter, and is not intended to limit the appended claims to the specific embodiments illustrated. The headings used throughout this disclosure are provided for convenience and are not to be construed to limit the claims in any way. Embodiments illustrated under any heading may be combined with embodiments illustrated under any other heading. [0027] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. It must be noted that as used herein and in the appended claims, the singular forms “a”, “and”, and “the” include plural referents unless the context clearly dictates otherwise. Thus, e.g., reference to “the compound” includes a plurality of such compounds and reference to “the assay” includes reference to one or more assays and equivalents thereof known to those skilled in the art, and so forth. [0028] As used in the present disclosure, the following words, phrases and symbols are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise. [0029] A dash ( ) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -CONH2 is attached through the carbon atom. A dash at the front or end of a chemical group is a matter of convenience; chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning. A wavy line drawn through a line in a structure indicates a point of attachment of a group. Unless chemically or structurally required, no directionality is indicated or implied by the order in which a chemical group is written or named. A solid line coming out of the center of a ring (including a fused, bridged or spirocyclic ring system) indicates that the point of attachment for a substituent on the ring can be at any ring atom. For example, R aa in the below structure can be attached to any of the five carbon ring atoms or R aa can replace the hydrogen attached to the nitrogen ring atom: . As another example, R aa in the below structure: , R aa can be attached to any of the numbered positions shown below: . [0030] A solid line coming out of the center of a ring (including a fused, bridged, or spirocyclic ring system) indicates that the point of attachment for the ring system to the rest of the compound can be at any ring atom of the fused, bridged, or spirocyclic ring system. For example, in the below structure: , the monocyclic heterocyclyl can be attached to the rest of the compound at any of the numbered positions shown below: . As another example, in the below fused bicyclic heterocyclic structure, , the fused bicyclic heterocyclyl can be attached to the rest of the compound at any of the eight numbered positions shown below: . [0031] The prefix “C u-v ” indicates that the following group has from u to v carbon atoms. For example, “C 1-6 alkyl” indicates that the alkyl group has from 1 to 6 carbon atoms. Likewise, the term “x-y membered” rings, wherein x and y are numerical ranges, such as “3 to12- membered heterocyclyl”, refers to a ring containing x-y atoms (i.e., 3-12), of which up to 80% may be heteroatoms, such as N, O, S, P, and the remaining atoms are carbon. [0032] Also, certain commonly used alternative chemical names may or may not be used. For example, a divalent group such as a divalent “alkyl” group, a divalent “aryl” group, etc., may also be referred to as an “alkylene” group or an “alkylenyl” group, or alkylyl group, an “arylene” group or an “arylenyl” group, or arylyl group, respectively. [0033] “A compound disclosed herein” or “a compound of the present disclosure” or “a compound provided herein” or “a compound described herein” refers to the compounds of Formula I. Also included are the specific compounds of Examples **1** to **157**. [0034] Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. In certain embodiments, the term “about” includes the indicated amount ± 10%. In other embodiments, the term “about” includes the indicated amount ± 5%. In certain other embodiments, the term “about” includes the indicated amount ± 1%. Also, the term “about X” includes description of “X”. [0035] “Alkyl” refers to an unbranched or branched saturated hydrocarbon chain. As used herein, alkyl has 1 to 20 carbon atoms (i.e., C 1-20 alkyl), 1 to 12 carbon atoms (i.e., C 1-12 alkyl), 1 to 8 carbon atoms (i.e., C 1-8 alkyl), 1 to 6 carbon atoms (i.e., C 1-6 alkyl), 1 to 4 carbon atoms (i.e., C 1-4 alkyl), 1 to 3 carbon atoms (i.e., C 1-3 alkyl), or 1 to 2 carbon atoms (i.e., C 1-2 alkyl). Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl, and 3- methylpentyl. When an alkyl residue having a specific number of carbons is named by chemical name or identified by molecular formula, all positional isomers having that number of carbons may be encompassed; thus, for example, “butyl” includes n-butyl (i.e. -(CH 2 ) 3 CH 3 ), sec-butyl (i.e. -CH(CH 3 )CH 2 CH 3 ), isobutyl (i.e. -CH 2 CH(CH 3 ) 2 ) and tert-butyl (i.e. -C(CH 3 ) 3 ); and “propyl” includes n-propyl (i.e. -(CH 2 ) 2 CH 3 ) and isopropyl (i.e. -CH(CH 3 ) 2 ). [0036] “Alkenyl” refers to an aliphatic group containing at least one carbon-carbon double bond and having from 2 to 20 carbon atoms (i.e., C 2-20 alkenyl), 2 to 8 carbon atoms (i.e., C 2-8 alkenyl), 2 to 6 carbon atoms (i.e., C 2-6 alkenyl), or 2 to 4 carbon atoms (i.e., C 2-4 alkenyl). Examples of alkenyl groups include ethenyl, propenyl, butadienyl (including 1,2-butadienyl and 1,3-butadienyl). [0037] “Alkynyl” refers to an aliphatic group containing at least one carbon-carbon triple bond and having from 2 to 20 carbon atoms (i.e., C 2-20 alkynyl), 2 to 8 carbon atoms (i.e., C 2-8 alkynyl), 2 to 6 carbon atoms (i.e., C 2-6 alkynyl), or 2 to 4 carbon atoms (i.e., C 2-4 alkynyl). The term “alkynyl” also includes those groups having one triple bond and one double bond. [0038] “Alkylene” refers to a divalent and unbranched saturated hydrocarbon chain. As used herein, alkylene has 1 to 20 carbon atoms (i.e., C 1-20 alkylene), 1 to 12 carbon atoms (i.e., C 1-12 alkylene), 1 to 8 carbon atoms (i.e., C 1-8 alkylene), 1 to 6 carbon atoms (i.e., C 1-6 alkylene), 1 to 4 carbon atoms (i.e., C 1-4 alkylene), 1 to 3 carbon atoms (i.e., C 1-3 alkylene), or 1 to 2 carbon atoms (i.e., C 1-2 alkylene). Examples of alkylene groups include methylene, ethylene, propylene, butylene, pentylene, and hexylene. In some embodiments, an alkylene is optionally substituted with an alkyl group. Examples of substituted alkylene groups include -CH(CH 3 )CH 2 -, -CH 2 CH(CH 3 )-, -CH 2 CH(CH 2 CH 3 )-, -CH 2 C(CH 3 ) 2 -, -C(CH 3 ) 2 CH 2 -, -CH(CH 3 )CH(CH 3 )-, -CH 2 C(CH 2 CH 3 )(CH 3 )-, and -CH 2 C(CH 2 CH 3 ) 2 . [0039] “Alkoxy” refers to the group “alkyl-O-”. Examples of alkoxy groups include methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n- hexoxy, and 1,2-dimethylbutoxy. “Haloalkoxy” refers to an alkoxy group as defined above, wherein one or more hydrogen atoms are replaced by a halogen. [0040] “Acyl” refers to a group -C(=O)R, wherein R is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be optionally substituted, as defined herein. Examples of acyl include formyl, acetyl, cylcohexylcarbonyl, cyclohexylmethyl-carbonyl, and benzoyl. [0041] “Amido” refers to both a “C-amido” group which refers to the group -C(═O)NR y R z and an “N-amido” group which refers to the group -NR y C(═O)R z , wherein R y and R z are independently selected from the group consisting of hydrogen, alkyl, aryl, haloalkyl, heteroaryl, cycloalkyl, or heterocyclyl; each of which may be optionally substituted. [0042] “Amino” refers to the group -NR y R z wherein R y and R z are independently selected from the group consisting of hydrogen, alkyl, haloalkyl, aryl, heteroaryl, cycloalkyl, or heterocyclyl; each of which may be optionally substituted. [0043] “Aryl” refers to an aromatic carbocyclic group having a single ring (e.g. monocyclic) or multiple rings (e.g. bicyclic or tricyclic) including fused systems. As used herein, aryl has 6 to 20 ring carbon atoms (i.e., C6-20 aryl), 6 to 12 carbon ring atoms (i.e., C6-12 aryl), or 6 to 10 carbon ring atoms (i.e., C6-10 aryl). Examples of aryl groups include phenyl, naphthyl, fluorenyl, and anthryl. Aryl, however, does not encompass or overlap in any way with heteroaryl defined below. If one or more aryl groups are fused with a heteroaryl ring, the resulting ring system is heteroaryl. [0044] “Cyano” or “carbonitrile” refers to the group -CN. [0045] “Cycloalkyl” refers to a saturated or partially saturated cyclic alkyl group having a single ring or multiple rings including fused, bridged, and spiro ring systems. The term “cycloalkyl” includes cycloalkenyl groups (i.e. the cyclic group having at least one double bond). As used herein, cycloalkyl has from 3 to 20 ring carbon atoms (i.e., C3-20 cycloalkyl), 3 to 12 ring carbon atoms (i.e., C 3-12 cycloalkyl), 3 to 10 ring carbon atoms (i.e., C 3-10 cycloalkyl), 3 to 8 ring carbon atoms (i.e., C 3-8 cycloalkyl), or 3 to 6 ring carbon atoms (i.e., C 3-6 cycloalkyl). Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. [0046] “Bridged” refers to a ring fusion wherein different atoms on a ring are joined by a divalent substituent, such as an alkylenyl group, an alkylenyl group containing one or two heteroatoms, or a single heteroatom. Quinuclidinyl and admantanyl are examples of bridged ring systems. [0047] The term “fused” refers to a ring which is bound to an adjacent ring. [0048] “Spiro” refers to a ring substituent which is joined by two bonds at the same carbon atom. Examples of spiro groups include 1,1-diethylcyclopentane, dimethyl-dioxolane, and 4-benzyl-4-methylpiperidine, wherein the cyclopentane and piperidine, respectively, are the spiro substituents. [0049] “Halogen” or “halo” includes fluoro, chloro, bromo, and iodo. “Haloalkyl” refers to an unbranched or branched alkyl group as defined above, wherein one or more hydrogen atoms are replaced by a halogen. For example, where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached. Dihaloalkyl and trihaloalkyl refer to alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be, but are not necessarily, the same halogen. Examples of haloalkyl include difluoromethyl (-CHF2) and trifluoromethyl (-CF3). [0050] “Heteroalkylene” refers to a divalent and unbranched saturated hydrocarbon chain having one, two, or three heteroatoms selected from NH, O, or S. As used herein, a heteroalkylene has 1 to 20 carbon atoms and one, two, or three heteroatoms selected from NH, O, and S (i.e., C 1-20 heteroalkylene); 1 to 8 carbon atoms and one, two, or three heteroatoms selected from NH, O, and S (i.e., C 1-8 heteroalkylene); 1 to 6 carbon atoms and one, two, or three heteroatoms selected from NH, O, and S S (i.e., C 1-6 heteroalkylene); 1 to 4 carbon atoms and one, two, or three heteroatoms selected from NH, O, and S (i.e., C 1-4 heteroalkylene); 1 to 3 carbon atoms and one, two, or three heteroatoms selected from NH, O, and S (i.e., C 1-3 heteroalkylene); or 1 to 2 carbon atoms and one, two, or three heteroatoms selected from NH, O, and S (i.e., C 1-3 heteroalkylene). For example, -CH 2 O- is a C1 heteroalkylene and -CH 2 SCH 2 - is a C2 heteroalkylene. Examples of heteroalkylene groups include -CH 2 CH 2 OCH 2 -, - CH 2 SCH 2 OCH 2 -, -CH 2 O-, and -CH 2 NHCH 2 -. In some embodiments, a heteroalkylene is optionally substituted with an alkyl group. Examples of substituted heteroalkylene groups include -CH(CH 3 )N(CH 3 )CH 2 -, -CH 2 OCH(CH 3 )-, -CH 2 CH(CH 2 CH 3 )S-, -CH 2 NHC(CH 3 ) 2 -, - C(CH 3 ) 2 SCH 2 -, -CH(CH 3 )N(CH 3 )CH(CH 3 )O-, -CH 2 SC(CH 2 CH 3 )(CH 3 )-, and - CH 2 C(CH 2 CH 3 ) 2 NH-. [0051] “Heteroaryl” refers to an aromatic group having a single ring, multiple rings, or multiple fused rings, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur. As used herein, heteroaryl includes 1 to 20 carbon ring atoms (i.e., C 1-20 heteroaryl), 3 to 12 carbon ring atoms (i.e., C3-12 heteroaryl), or 3 to 8 carbon ring atoms (i.e., C3-8 heteroaryl); and 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups include pyrimidinyl, purinyl, pyridyl, pyridazinyl, benzothiazolyl, and pyrazolyl. Heteroaryl does not encompass or overlap with aryl as defined above. [0052] “Heterocyclyl” or “heterocyclic ring” or “heterocycle” refers to a non-aromatic cyclic alkyl group, with one or more ring heteroatoms independently selected from nitrogen, oxygen and sulfur. As used herein, “heterocyclyl” or “heterocyclic ring” or “heterocycle” refer to rings that are saturated or partially saturated unless otherwise indicated, e.g., in some embodiments “heterocyclyl” or “heterocyclic ring” or “heterocycle” refers to rings that are partially saturated where specified. The term “heterocyclyl” or “heterocyclic ring” or “heterocycle” includes heterocycloalkenyl groups (i.e., the heterocyclyl group having at least one double bond). A heterocyclyl may be a single ring or multiple rings wherein the multiple rings may be fused, bridged, or spiro. As used herein, heterocyclyl has 2 to 20 carbon ring atoms (i.e., C 2-20 heterocyclyl), 2 to 12 carbon ring atoms (i.e., C 2-12 heterocyclyl), 2 to 10 carbon ring atoms (i.e., C 2-10 heterocyclyl), 2 to 8 carbon ring atoms (i.e., C 2-8 heterocyclyl), 3 to 12 carbon ring atoms (i.e., C3-12 heterocyclyl), 3 to 8 carbon ring atoms (i.e., C3-8 heterocyclyl), or 3 to 6 carbon ring atoms (i.e., C 3-6 heterocyclyl); having 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, sulfur or oxygen. Examples of heterocyclyl groups include pyrrolidinyl, piperidinyl, piperazinyl, oxetanyl, dioxolanyl, azetidinyl, and morpholinyl. As used herein, the term “bridged- heterocyclyl” refers to a four- to ten-membered cyclic moiety connected at two non-adjacent atoms of the heterocyclyl with one or more (e.g., 1 or 2) four- to ten-membered cyclic moiety having at least one heteroatom where each heteroatom is independently selected from nitrogen, oxygen, and sulfur. As used herein, “bridged- heterocyclyl” includes bicyclic and tricyclic ring systems. Also as used herein, the term “spiro- heterocyclyl” refers to a ring system in which a three- to ten-membered heterocyclyl has one or more additional ring, wherein the one or more additional ring is three- to ten-membered cycloalkyl or three- to ten-membered heterocyclyl, where a single atom of the one or more additional ring is also an atom of the three- to ten-membered heterocyclyl. Examples of the spiro- heterocyclyl include bicyclic and tricyclic ring systems, such as 2-oxa-7- azaspiro[3.5]nonanyl, 2-oxa-6-azaspiro[3.4]octanyl, and 6-oxa-1-azaspiro[3.3]heptanyl. As used herein, the terms “heterocycle”, “heterocyclyl”, and “heterocyclic ring” are used interchangeably. In some embodiments, a heterocyclyl is substituted with an oxo group. [0053] “Hydroxy” or “hydroxyl” refers to the group -OH. [0054] “Oxo” refers to the group (=O) or (O). [0055] “Sulfonyl” refers to the group -S(O) 2 R bb , where R bb is alkyl, haloalkyl, heterocyclyl, cycloalkyl, heteroaryl, or aryl. Examples of sulfonyl are methylsulfonyl, ethylsulfonyl, phenylsulfonyl, and toluenesulfonyl. [0056] Whenever the graphical representation of a group terminates in a singly bonded nitrogen atom, that group represents an -NH group unless otherwise indicated. Similarly, unless otherwise expressed, hydrogen atom(s) are implied and deemed present where necessary in view of the knowledge of one of skill in the art to complete valency or provide stability. [0057] The terms “optional” or “optionally” mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. Also, the term “optionally substituted” means that any one or more hydrogen atoms on the designated atom or group may or may not be replaced by a moiety other than hydrogen. [0058] The term “substituted” means that any one or more hydrogen atoms on the designated atom or group is replaced with one or more substituents other than hydrogen, provided that the designated atom’s normal valence is not exceeded. The one or more substituents include, but are not limited to, alkyl, alkenyl, alkynyl, alkoxy, acyl, amino, amido, amidino, aryl, azido, carbamoyl, carboxyl, carboxyl ester, cyano, guanidino, halo, haloalkyl, heteroalkyl, heteroaryl, heterocyclyl, hydroxy, hydrazino, imino, oxo, nitro, alkylsulfinyl, sulfonic acid, alkylsulfonyl, thiocyanate, thiol, thione, or combinations thereof. Polymers or similar indefinite structures arrived at by defining substituents with further substituents appended ad infinitum (e.g., a substituted aryl having a substituted alkyl which is itself substituted with a substituted aryl group, which is further substituted by a substituted heteroalkyl group, etc.) are not intended for inclusion herein. Unless otherwise noted, the maximum number of serial substitutions in compounds described herein is three. For example, serial substitutions of substituted aryl groups with two other substituted aryl groups are limited to ((substituted aryl)substituted aryl) substituted aryl. Similarly, the above definitions are not intended to include impermissible substitution patterns (e.g., methyl substituted with 5 fluorines or heteroaryl groups having two adjacent oxygen ring atoms). Such impermissible substitution patterns are well known to the skilled artisan. When used to modify a chemical group, the term “substituted” may describe other chemical groups defined herein. For example, the term “substituted aryl” includes, but is not limited to, “alkylaryl.” Unless specified otherwise, where a group is described as optionally substituted, any substituents of the group are themselves unsubstituted. [0059] In some embodiments, a substituted cycloalkyl, a substituted heterocyclyl, a substituted aryl, and/or a substituted heteroaryl includes a cycloalkyl, a heterocyclyl, an aryl, and/or a heteroaryl that has a substituent on the ring atom to which the cycloalkyl, heterocyclyl, aryl, and/or heteroaryl is attached to the rest of the compound. For example, in the below moiety, the cyclopropyl is substituted with a methyl group: . [0060] The compounds of the embodiments disclosed herein, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. Where compounds are represented in their chiral form, it is understood that the embodiment encompasses, but is not limited to, the specific diastereomerically or enantiomerically enriched form. Where chirality is not specified but is present, it is understood that the embodiment is directed to either the specific diastereomerically or enantiomerically enriched form; or a racemic or scalemic mixture of such compound(s). As used herein, “scalemic mixture” is a mixture of stereoisomers at a ratio other than 1:1. [0061] A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are non-superimposable mirror images of one another. [0062] "Enantiomers" are a pair of stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a "racemic" mixture. A mixture of enantiomers at a ratio other than 1:1 is a “scalemic” mixture. [0063] "Diastereoisomers" are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other. [0064] A “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any compounds provided herein. [0065] Some of the compounds provided herein exist as tautomeric isomers. Tautomeric isomers are in equilibrium with one another. For example, amide containing compounds may exist in equilibrium with imidic acid tautomers. Regardless of which tautomer is shown, and regardless of the nature of the equilibrium among tautomers, the compounds are understood by one of ordinary skill in the art to comprise both amide and imidic acid tautomers. Thus, the amide containing compounds are understood to include their imidic acid tautomers. Likewise, the imidic acid containing compounds are understood to include their amide tautomers. [0066] A “solvate” is formed by the interaction of a solvent and a compound. Solvates of salts of the compounds provided herein are also provided. Hydrates of the compounds provided herein are also provided. [0067] Any formula or structure provided herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as, but not limited to 2 H (deuterium, D), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 18 F, 31 P, 32 P, 35 S, 36 Cl and 125 I. Various isotopically labeled compounds of the present disclosure, for example those into which radioactive isotopes such as 2 H, 3 H, 13 C and 14 C are incorporated, are also provided herein. Such isotopically labelled compounds may be useful in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays or in radioactive treatment of patients. [0068] The present disclosure also includes compounds of Formula I, in which from 1 to n hydrogens attached to a carbon atom is/are replaced by deuterium, in which n is the number of hydrogens in the molecule. Such compounds exhibit increased resistance to metabolism and are thus useful for increasing the half-life of any compound of Formula I, II, or IIa, when administered to a mammal, particularly a human. See, for example, Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism,” Trends Pharmacol. Sci.5(12):524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium. [0069] Deuterium labelled or substituted therapeutic compounds of the present disclosure may have improved DMPK (drug metabolism and pharmacokinetics) properties, relating to absorption, distribution, metabolism and excretion (ADME). Substitution with heavier isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, reduced dosage requirements and/or an improvement in therapeutic index. An 18 F labeled compound may be useful for PET or SPECT studies. Isotopically labeled compounds of this disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. It is understood that deuterium in this context is regarded as a substituent in the compound of Formula I. [0070] The concentration of such a heavier isotope, specifically deuterium, may be defined by an isotopic enrichment factor. In the compounds of this disclosure, any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural abundance isotopic composition. Accordingly, in the compounds of this disclosure, any atom specifically designated as a deuterium (D) is meant to represent deuterium. [0071] In many cases, the compounds of this disclosure are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. [0072] The term “pharmaceutically acceptable salt” of a given compound refers to salts that retain the biological effectiveness and properties of the given compound, and which are not biologically or otherwise undesirable. Pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di(substituted alkyl) amines, tri(substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, substituted alkenyl amines, di(substituted alkenyl) amines, tri(substituted alkenyl) amines, mono, di or tri cycloalkyl amines, mono, di or tri arylamines or mixed amines, and the like. Specific examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like. [0073] Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like. [0074] As used herein, “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. [0075] “Treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired clinical results may include one or more of the following: a) inhibiting the disease or condition (i.e., decreasing one or more symptoms resulting from the disease or condition, and/or diminishing the extent of the disease or condition); b) slowing or arresting the development of one or more clinical symptoms associated with the disease or condition (i.e., stabilizing the disease or condition, preventing or delaying the worsening or progression of the disease or condition, and/or preventing or delaying the spread (i.e., metastasis) of the disease or condition); and/or c) relieving the disease, that is, causing the regression of clinical symptoms (i.e., ameliorating the disease state, providing partial or total remission of the disease or condition, enhancing effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival). [0076] “Prevention” or “preventing” means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. Compounds may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition. [0077] “Subject” refers to an animal, such as a mammal (including a human), that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in human therapy and/or veterinary applications. In some embodiments, the subject is a mammal. In one embodiment, the subject is a human. [0078] The term “therapeutically effective amount” or “effective amount” of a compound described herein or pharmaceutically acceptable salts, isomer, or a mixture thereof means an amount sufficient to effect treatment when administered to a subject, to provide a therapeutic benefit such as amelioration of symptoms or slowing of disease progression. For example, a therapeutically effective amount may be an amount sufficient to improve a symptom of a disease or condition responsive to inhibition of toll-like receptor 7, 8, and/or 9. The therapeutically effective amount may vary depending on the subject, and the disease or condition being treated, the weight and age of the subject, the severity of the disease or condition, and the manner of administering, which can readily be determined by one of ordinary skill in the art. II. Compounds [0079] In one embodiment, provided herein is a compound of Formula I, Formula I or a pharmaceutically acceptable salt thereof, wherein R 1 is 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-4 R a groups; R 2 is H, -CN, C 1-6 alkyl, or C 3-7 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups; X 1 and X 3 are each independently N or CR 3 ; each R 3 independently is H, halogen, C 1-6 alkyl, C 3-6 monocyclic cycloalkyl, or -O(C 1-4 alkyl), wherein the C 1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 4 R 4 , and C 1-4 alkoxy; X 2 is N or CH; Y is C1-10 alkyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, or L 1 , wherein the C1-10 alkyl and C 2-6 alkynyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R a groups; L 1 is -OR 5 , -C(O)R 5 , -C(O)N(R 5 )(R 5 ), -NR 5 R 5 , -N(R 5 ) 2 (R 5 ) + , -N(R 5 )C(O)R 5 , -N(R 5 )C(O)OR 5 , -N(R 5 )C(O)N(R 5 )(R 5 ), -N(R 5 )S(O) 2 (R 5a ), -NR 5 S(O) 2 N(R 5 )(R 5 ), -NR 5 S(O) 2 O(R 5a ), -OC(O)N(R 5 )(R 5 ), -SR 5 , -S(O)R 5a , -S(O)(NH)R 5 , -S(O) 2 R 5a , -S(O) 2 N(R 5 )(R 5 ), or -N=S(R 5a )(R 5a )=O; Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -C(O)NR 6 R 7 ,-S(O) 2 R 6 , C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups; wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 R a groups; R 6 is C 1-6 alkyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; R 13 is C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6- 10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R 4 and R 7 independently is H or C 1-3 alkyl; each R 8 independently is halogen, -C(O)R 9 , -NR 10 R 10 , C 1-6 alkyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 5 , -C(O)OR 5 , -C(O)N(R 5 )(R 5 ), -N(R 5 ) 2 (R 5 ) + , -N(R 5 )C(O)R 5 , -N(R 5 )C(O)OR 5 , -N(R 5 )C(O)N(R 5 )(R 5 ), -N(R 5 )S(O) 2 (R 5a ), -NR 5 S(O) 2 N(R 5 )(R 5 ), -NR 5 S(O) 2 O(R 5a ), -OC(O)R 5 , -OC(O)OR 5 , -OC(O)N(R 5 )(R 5 ), -SR 5 , -S(O)R 5a , -S(O)(NH)R 5 , -S(O) 2 R 5a , -S(O) 2 N(R 5 )(R 5 ), or -N=S(R 5a )(R 5a )=O, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; R 9 is C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 2-6 alkenyl and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R 5 and R 10 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6- 10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R 5a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups; each R a independently is oxo, imino, halogen, -NO2, -N3, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , -N(R 11 ) 2 (R 11 ) + , -N(R 11 )C(O)R 11 , -N(R 11 )C(O)OR 11 , -N(R 11 )C(O)N(R 11 )(R 11 ), -N(R 11 )S(O) 2 (R 11a ), -NR 11 S(O) 2 N(R 11 )(R 11 ), -NR 11 S(O) 2 O(R 11a ), -OC(O)R 11 , -OC(O)OR 11 , -OC(O)N(R 11 )(R 11 ), -SR 11 , -S(O)R 11a , -S(O)(NH)R 11 , -S(O) 2 R 11a , -S(O) 2 N(R 11 )(R 11 ), or -N=S(R 11a )(R 11a )=O, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups, each R b independently is oxo, imino, halogen, -NO2, -N3, -CN, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , -N(R 11 ) 2 (R 11 ) + , -N(R 11 )C(O)R 11 , -N(R 11 )C(O)OR 11 , -N(R 11 )C(O)N(R 11 )(R 11 ), -N(R 11 )S(O) 2 (R 11a ), -NR 11 S(O) 2 N(R 11 )(R 11 ), -NR 11 S(O) 2 O(R 11a ), -OC(O)R 11 , -OC(O)OR 11 , -OC(O)N(R 11 )(R 11 ), -SR 11 , -S(O)R 11a , -S(O)(NH)R 11 , -S(O) 2 R 11a , -S(O) 2 N(R 11 )(R 11 ), or -N=S(R 11a )(R 11a )=O, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups; each R c independently is halogen, -CN, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 12 , -C(O)R 12 , -C(O)OR 12 , -C(O)N(R 12 )(R 12 ), -NR 12 R 12 , -N(R 12 ) 2 (R 12 ) + , -N(R 12 )C(O)R 12 , -N(R 12 )C(O)OR 12 , -N(R 12 )C(O)N(R 12 )(R 12 ), -N(R 12 )S(O) 2 (R 12a ), -NR 12 S(O) 2 N(R 12 )(R 12 ), -NR 12 S(O) 2 O(R 12a ), -OC(O)R 12 , -OC(O)OR 12 , -OC(O)N(R 12 )(R 12 ), -SR 12 , -S(O)R 12a , -S(O)(NH)R 12 , -S(O) 2 R 12a , -S(O) 2 N(R 12 )(R 12 ), or -N=S(R 12a )(R 12a )=O; each R 11 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups; each R 11a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups; each R 12 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; each R 12a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0080] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 R a groups; R 2 is -CN, C 1-6 alkyl, or C3-5 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, wherein the C 3-5 monocyclic cycloalkyl is optionally substituted with 1-3 R a groups; X 1 is N or CR 3 ; X 3 is CR 3 ; each R 3 independently is H, halogen, or C 1-3 alkyl; X 2 is CH; Y is C2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8- 10 membered fused bicyclic heteroaryl, or L 1 , wherein the C2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R a groups; L 1 is -C(O)N(R 5 )(R 5 ); each R 5 independently is H or 4-7 membered monocyclic heterocyclyl; Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -S(O) 2 R 6 , 5-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 R a groups; R 6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 R a groups; R 13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 R a groups; each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , -NR 10 R 10 , 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 R a groups; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0081] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 R a groups; R 2 is -CN, C 1-6 alkyl, or C3-5 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, wherein the C 3-5 monocyclic cycloalkyl is optionally substituted with 1-3 R a groups; X 1 is N or CR 3 ; X 3 is CR 3 ; each R 3 independently is H, halogen, or C 1-3 alkyl; X 2 is CH; Y is C 2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the C 2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R a groups; Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -S(O) 2 R 6 , 5-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 R a groups; R 6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 R a groups; R 13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 R a groups; each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , -NR 10 R 10 , 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 R a groups; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0082] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups; R 2 is -CN, C 1-6 alkyl, or C 3-5 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy; X 1 is N or CR 3 ; X 3 is CR 3 ; each R 3 independently is H, halogen, or C 1-3 alkyl; X 2 is CH; Y is C 2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8- 10 membered fused bicyclic heteroaryl, or L 1 , wherein the C 2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups; L 1 is -C(O)N(R 5 )(R 5 ); each R 5 independently is H or 4-7 membered monocyclic heterocyclyl; Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -S(O) 2 R 6 , 5-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with one R b group, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; R 6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; R 13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , -NR 10 R 10 , 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; R 9 is 4-7 membered monocyclic heterocyclyl or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; each R 11 independently is H or C 1-4 alkyl, wherein the C 1-4 alkyl is optionally substituted with one group selected from -OH and -NR 12 R 12 ; R b is -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy; each R 12 independently is H or C 1-3 alkyl; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0083] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups; R 2 is -CN, C 1-6 alkyl, or C3-5 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy; X 1 is N or CR 3 ; X 3 is CR 3 ; each R 3 independently is H, halogen, or C 1-3 alkyl; X 2 is CH; Y is C2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the C2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups; Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -S(O) 2 R 6 , 5-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with one R b group, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; R 6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; R 13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , -NR 10 R 10 , 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1- 3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; R 9 is 4-7 membered monocyclic heterocyclyl or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl; each R 11 independently is H or C 1-4 alkyl, wherein the C 1-4 alkyl is optionally substituted with one group selected from -OH and -NR 12 R 12 ; R b is -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl; each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy; each R 12 independently is H or C 1-3 alkyl; wherein each 4-membered monocyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 5-7 membered monocyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; wherein each 6-membered bridged bicyclic heterocyclyl independently has 1 ring heteroatom selected from N, O, and S; wherein each 7-membered bridged bicyclic heterocyclyl independently has 1-2 ring heteroatoms independently selected from N, O, and S; and wherein each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl each independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0084] Unless specified otherwise, each 4-membered monocyclic heterocyclyl as used herein has 1 ring heteroatom selected from N, O, and S. Unless specified otherwise, each 5-7 membered monocyclic heterocyclyl as used herein has 1-2 ring heteroatoms independently selected from N, O, and S. Unless specified otherwise, each 6-membered bridged bicyclic heterocyclyl as used herein has 1 ring heteroatom selected from N, O, and S. Unless specified otherwise, each 7-membered bridged bicyclic heterocyclyl as used herein has 1-2 ring heteroatoms independently selected from N, O, and S. Unless specified otherwise, each 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl as used herein independently have 1-4 ring heteroatoms independently selected from N, O, and S. [0085] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-4 R a groups. [0086] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 R a groups. [0087] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. [0088] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups, wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0089] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0090] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0091] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 4-7 membered monocyclic heterocyclyl. [0092] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0093] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0094] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-7 membered monocyclic heterocyclyl. [0095] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0096] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, wherein the phenyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0097] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl. [0098] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0099] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl, wherein the naphthalenyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0100] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is naphthalenyl. [0101] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0102] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0103] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 5-6 membered monocyclic heteroaryl. [0104] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8- 10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8- 10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0105] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0106] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heterocyclyl. [0107] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8- 10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0108] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups and wherein each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. [0109] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is 8-10 membered fused bicyclic heteroaryl. [0110] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is

, each of which is optionally substituted with 1-4 R a groups. [0111] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, pyrazolyl, isoxazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinolinyl, , each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0112] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is phenyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinolinyl, , each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0113] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is substituted with 1-3 groups independently selected from -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1-3 halogen. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is substituted with 1-3 groups independently selected from -CN, oxo, -NH2, methoxy, methyl, and -CF3. [0114] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is pyridinyl substituted with 1-3 groups independently selected from -CN, methoxy, methyl, and -NH2. [0115] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is , , , , , ,

[0116] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is , [0117] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is [0118] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 1 is . [0119] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is H, -CN, C 1-6 alkyl, or C 3-7 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. [0120] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is -CN, C 1-6 alkyl, or C 3-5 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, wherein the C3-5 monocyclic cycloalkyl is optionally substituted with 1-3 R a groups. [0121] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is -CN, C 1-6 alkyl, or C 3-5 monocyclic cycloalkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy. [0122] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is -CN, C 1-4 alkyl, or cyclopropyl, wherein the C 1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy and wherein the cyclopropyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is -CN, -C(O)CH 3 , methyl, ethyl, isopropyl, or cyclopropyl. [0123] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is -CN. [0124] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-6 alkyl. [0125] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-4 alkyl, wherein the C 1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-4 alkyl, wherein the C 1-4 alkyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, and C 1-3 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is -C(O)CH 3 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-4 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is methyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is ethyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is propyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is isopropyl. [0126] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 3-7 monocyclic cycloalkyl. [0127] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is cyclopropyl, wherein the cyclopropyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is cyclopropyl, wherein the cyclopropyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is cyclopropyl. [0128] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 2 is C 1-3 alkyl and R 1 is . [0129] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 and X 3 are each independently N or CR 3 . [0130] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 is N or CH. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 is N. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 is CR 3 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 is CR 3 , wherein R 3 is H, halogen, or C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 is CH. [0131] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 3 is N or CH. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 3 is N. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 3 is CR 3 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 3 is CR 3 , wherein R 3 is H, halogen, or C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 3 is CH. [0132] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 and X 3 are CH. [0133] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 3 independently is H, halogen, C 1-6 alkyl, C 3-6 monocyclic cycloalkyl, or -O(C 1-4 alkyl), wherein the C 1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 4 R 4 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 3 independently is H, halogen, or C 1-3 alkyl. [0134] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is halogen. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is C 1-6 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is C 3-6 monocyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is -O(C 1-4 alkyl), wherein the C 1-4 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 4 R 4 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is -OCF3. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 3 is -OCHF 2 . [0135] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 2 is N or CH. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 2 is N. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 2 is CH. [0136] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, X 1 , X 2 , and X 3 are CH. [0137] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C1-10 alkyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, or L 1 , wherein the C1-10 alkyl and C 2-6 alkynyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R a groups. [0138] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the C 2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 R a groups. [0139] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-3 alkynyl, 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the C 2-3 alkynyl is substituted with one Z group, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with one Z group and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. [0140] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, and wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0141] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, or 8-10 membered fused bicyclic heteroaryl, wherein the 5-7 membered monocyclic heterocyclyl, phenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, and 8-10 membered fused bicyclic heteroaryl are each substituted with one Z group and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, and wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0142] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 1-10 alkyl, wherein the C 1-10 alkyl is optionally substituted with one Z group and is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C1-10 alkyl, wherein the C1-10 alkyl is substituted with one Z group and is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C1-10 alkyl, wherein the C1-10 alkyl is substituted with one Z group and is substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 1-10 alkyl, wherein the C 1-10 alkyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C1-10 alkyl, wherein the C1-10 alkyl is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C1-10 alkyl. [0143] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with one Z group and is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-6 alkynyl, wherein the C 2-6 alkynyl is substituted with one Z group and is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-6 alkynyl, wherein the C 2-6 alkynyl is substituted with one Z group and is substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-6 alkynyl, wherein the C 2-6 alkynyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2-6 alkynyl. [0144] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C2-3 alkynyl substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 2 alkynyl substituted with one Z group. [0145] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with one Z group and is substituted with 1- 3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 3-7 monocyclic cycloalkyl. [0146] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 7-10 fused bicyclic cycloalkyl. [0147] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C 5-10 bridged bicyclic cycloalkyl. [0148] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 4-7 membered monocyclic heterocyclyl. [0149] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5- 7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0150] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0151] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0152] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-7 membered monocyclic heterocyclyl. [0153] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0154] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0155] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0156] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl. [0157] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0158] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0159] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0160] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is naphthalenyl. [0161] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5- 6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0162] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0163] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0164] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 5-6 membered monocyclic heteroaryl. [0165] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0166] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0167] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0168] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8- 10 membered fused bicyclic heterocyclyl. [0169] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 6-10 membered bridged bicyclic heterocyclyl. [0170] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0171] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8- 10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0172] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. [0173] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the C 1-5 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 12 R 12 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 8- 10 membered fused bicyclic heteroaryl. [0174] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with one Z group and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with one Z group and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with one Z group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is 7-10 membered spirocyclic heterocyclyl. [0175] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is L 1 . [0176] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -OR 5 , -C(O)R 5 , -C(O)N(R 5 )(R 5 ), -NR 5 R 5 , -N(R 5 ) 2 (R 5 ) + , -N(R 5 )C(O)R 5 , -N(R 5 )C(O)OR 5 , -N(R 5 )C(O)N(R 5 )(R 5 ), -N(R 5 )S(O) 2 (R 5a ), -NR 5 S(O) 2 N(R 5 )(R 5 ), -NR 5 S(O) 2 O(R 5a ), -OC(O)N(R 5 )(R 5 ), -SR 5 , -S(O)R 5a , -S(O)(NH)R 5 , -S(O) 2 R 5a , -S(O) 2 N(R 5 )(R 5 ), or -N=S(R 5a )(R 5a )=O. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -OR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -C(O)R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -C(O)N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -NR 5 R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -N(R 5 ) 2 (R 5 ) + . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -N(R 5 )C(O)R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -N(R 5 )C(O)OR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -N(R 5 )C(O)N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -N(R 5 )S(O) 2 (R 5a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -NR 5 S(O) 2 N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -NR 5 S(O) 2 O(R 5a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -OC(O)N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -SR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -S(O)R 5a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -S(O)(NH)R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -S(O) 2 R 5a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -S(O) 2 N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is -N=S(R 5a )(R 5a )=O. [0177] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is . [0178] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C2 alkynyl and Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C2 alkynyl and Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C2 alkynyl and Z is 5-7 membered monocyclic heterocyclyl. [0179] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is C2 alkynyl and Z is piperazinyl. [0180] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is pyrrolidinyl, piperidinyl, pyrazolyl, pyridinyl, quinolinyl, triazolyl, pyrimidinyl, thiazolyl, , each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with -NH 2 . [0181] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is pyrrolidinyl, piperidinyl, pyrazolyl, pyridinyl, quinolinyl, triazolyl, pyrimidinyl, thiazolyl, each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with -NH2. [0182] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is pyrrolidinyl, piperidinyl, pyrazolyl, pyridinyl, quinolinyl, triazolyl, pyrimidinyl, thiazolyl, [0183] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is pyrrolidinyl, piperidinyl, pyrazolyl, pyridinyl, quinolinyl, triazolyl, pyrimidinyl, thiazolyl, each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with -NH2. [0184] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is optionally substituted with 1-2 groups independently selected from -CH 2 CH 2 NH2, methoxy, -N(CH 3 ) 2 , -NH2, -N(CH 3 )CH 2 CH 2 NHCH 3 , and methyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is substituted with 1-2 groups independently selected from -CH 2 CH 2 NH 2 , methoxy, -N(CH 3 ) 2 , -NH2, -N(CH 3 )CH 2 CH 2 NHCH 3 , and methyl. [0185] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is piperidinyl. [0186] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is [0187] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is [0188] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, pyrazolyl, triazolyl, thiazolyl, pyridinyl, pyrimidinyl, or , each of which is substituted with one Z group and independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0189] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is substituted with one Z group and is optionally substituted with C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is substituted with one Z group and is optionally substituted with methyl. [0190] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is pyridinyl, wherein the pyridinyl is substituted with one Z group and is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0191] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl, wherein the phenyl is substituted with one Z group and is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0192] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y substituted with Z is , each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0193] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y substituted with Z is each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0194] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -C(O)NR 6 R 7 ,-S(O) 2 R 6 , C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups; wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 R a groups. [0195] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -S(O) 2 R 6 , 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 R a groups. [0196] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , -S(O) 2 R 6 , 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with one R b group, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0197] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0198] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl each independently have one or two ring heteroatoms that is N. [0199] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0200] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, or 7-10 membered spirocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl are each substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, and 7-10 membered spirocyclic heterocyclyl each independently have one or two ring heteroatoms that is N. [0201] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl or 8-10 membered fused bicyclic heterocyclyl, wherein the 5-7 membered heterocyclyl and 8-10 membered fused bicyclic heterocyclyl are each substituted with 1-2 R 8 groups and are each independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0202] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, -NR 6 R 7 , -C(O)R 13 , or -S(O) 2 R 6 , wherein the C 1-6 alkyl is optionally substituted with one group selected from 4-7 membered monocyclic heterocyclyl, 8- 10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl. [0203] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -NHR 6 , -N(CH 3 )R 6 , -C(O)R 13 , -S(O) 2 R 6 , or methyl substituted with piperazinyl . [0204] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one oxo group and 1- 2 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one oxo group and one group selected from -NR 11 R 11 and 5-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one R b group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one group selected from 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is methyl optionally substituted with piperazinyl. [0205] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C1- 6 alkyl, wherein the C 1-6 alkyl is substituted with one oxo group and 1-2 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1- 6 alkyl, wherein the C 1-6 alkyl is substituted with one oxo group and one group selected from - NR 11 R 11 and 5-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with one R b group. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with one group selected from 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is methyl substituted with piperazinyl. [0206] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 1-6 alkyl. [0207] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -C(O)(C 1-4 alkyl), wherein the C 1-4 alkyl is substituted with -NR 11 R 11 or 5-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -C(O)(C 1-2 alkyl), wherein the C 1-2 alkyl is substituted with -NR 11 R 11 or 5-7 membered monocyclic heterocyclyl. [0208] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -NR 6 R 7 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -NHR 6 or -N(CH 3 )R 6 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -NHR 6 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -N(CH 3 )R 6 . [0209] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -C(O)NR 6 R 7 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -C(O)N(H)R 6 . [0210] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -S(O) 2 R 6 . [0211] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 1-6 alkyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0212] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 R a groups. [0213] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0214] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 1-6 alkyl. [0215] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 3-7 monocyclic cycloalkyl. [0216] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 7-10 fused bicyclic cycloalkyl. [0217] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is C 5-10 bridged bicyclic cycloalkyl. [0218] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0219] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0220] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 4-7 membered monocyclic heterocyclyl. [0221] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is phenyl, wherein the phenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is phenyl. [0222] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is naphthalenyl, wherein the naphthalenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is naphthalenyl. [0223] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 5-6 membered monocyclic heteroaryl. [0224] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8- 10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0225] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0226] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 8-10 membered fused bicyclic heterocyclyl. [0227] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6- 10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0228] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6- 10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0229] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 6-10 membered bridged bicyclic heterocyclyl. [0230] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 7- 10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 6 is 7-10 membered spirocyclic heterocyclyl. [0231] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Y is phenyl and Z is -C(O)R 13 . [0232] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is -C(O)R 13 . [0233] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0234] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 R a groups. [0235] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0236] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 3-7 monocyclic cycloalkyl. [0237] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 7-10 fused bicyclic cycloalkyl. [0238] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is C 5-10 bridged bicyclic cycloalkyl. [0239] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0240] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0241] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 4-7 membered monocyclic heterocyclyl. [0242] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is phenyl, wherein the phenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is phenyl. [0243] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is naphthalenyl, wherein the naphthalenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is naphthalenyl. [0244] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 5-6 membered monocyclic heteroaryl. [0245] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0246] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0247] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 8-10 membered fused bicyclic heterocyclyl. [0248] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0249] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0250] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 6-10 membered bridged bicyclic heterocyclyl. [0251] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 is 7-10 membered spirocyclic heterocyclyl. [0252] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 3-7 monocyclic cycloalkyl. [0253] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 7- 10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C7- 10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 7-10 fused bicyclic cycloalkyl. [0254] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C5- 10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 5- 10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is C 5-10 bridged bicyclic cycloalkyl. [0255] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 4-7 membered monocyclic heterocyclyl. [0256] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0257] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0258] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0259] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-7 membered monocyclic heterocyclyl. [0260] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is phenyl, wherein the phenyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is phenyl, wherein the phenyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is phenyl, wherein the phenyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is phenyl, wherein the phenyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is phenyl, wherein the phenyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is phenyl. [0261] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is naphthalenyl, wherein the naphthalenyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is naphthalenyl, wherein the naphthalenyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is naphthalenyl, wherein the naphthalenyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is naphthalenyl. [0262] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5- 6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 5- 6 membered monocyclic heteroaryl. [0263] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0264] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0265] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0266] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heterocyclyl. [0267] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0268] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0269] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0270] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6- 10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 6-10 membered bridged bicyclic heterocyclyl. [0271] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8- 10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 8-10 membered fused bicyclic heteroaryl. [0272] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0273] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-2 R 8 groups and is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0274] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-2 R 8 groups and is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0275] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-2 R 8 groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is 7-10 membered spirocyclic heterocyclyl. [0276] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-5 alkyl. [0277] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, , each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-5 alkyl. [0278] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, , each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0279] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0280] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl [0281] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is optionally substituted with -NH 2 . [0282] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is piperazinyl. [0283] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, each of which is substituted with 1-2 R 8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl [0284] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, diazepanyl, each of which is substituted with 1-2 R 8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0285] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z is pyrrolidinyl, piperidinyl, or piperazinyl, each of which is substituted with 1-2 R 8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0286] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, wherein Z is each of which is substituted with 1-2 R 8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0287] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, wherein Z is each of which is substituted with 1-2 R 8 groups and is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0288] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z substituted with 1-2 R 8 groups is: each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0289] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z substituted with 1-2 R 8 groups is: , each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0290] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, Z substituted with 1-2 R 8 groups is: . [0291] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is halogen, -C(O)R 9 , -NR 10 R 10 , C 1-6 alkyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 5 , -C(O)OR 5 , -C(O)N(R 5 )(R 5 ), -N(R 5 ) 2 (R 5 ) + , -N(R 5 )C(O)R 5 , -N(R 5 )C(O)OR 5 , -N(R 5 )C(O)N(R 5 )(R 5 ), -N(R 5 )S(O) 2 (R 5a ), -NR 5 S(O) 2 N(R 5 )(R 5 ), -NR 5 S(O) 2 O(R 5a ), -OC(O)R 5 , -OC(O)OR 5 , -OC(O)N(R 5 )(R 5 ), -SR 5 , -S(O)R 5a , -S(O)(NH)R 5 , -S(O) 2 R 5a , -S(O) 2 N(R 5 )(R 5 ), or -N=S(R 5a )(R 5a )=O, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, 4-7 membered monocyclic heterocyclyl, phenyl, naphthalenyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0292] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , -NR 10 R 10 , 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 R a groups. [0293] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , -NR 10 R 10 , 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0294] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is halogen, C 1-6 alkyl, -C(O)R 9 , or -NR 10 R 10 , wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy. [0295] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is halogen. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -OR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -C(O)OR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -C(O)N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -N(R 5 ) 2 (R 5 ) + . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -N(R 5 )C(O)R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -N(R 5 )C(O)OR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -N(R 5 )C(O)N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -N(R 5 )S(O) 2 (R 5a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -NR 5 S(O) 2 N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -NR 5 S(O) 2 O(R 5a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -OC(O)R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -OC(O)OR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -OC(O)N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -SR 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -S(O)R 5a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -S(O)(NH)R 5 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -S(O) 2 R 5a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -S(O) 2 N(R 5 )(R 5 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -N=S(R 5a )(R 5a )=O. [0296] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-3 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl, wherein the C 1-6 alkyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-3 alkyl, wherein the C 1-3 alkyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-3 alkyl, wherein the C 1-3 alkyl is substituted with 1-2 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , and C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-6 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 1-3 alkyl. [0297] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 3-7 monocyclic cycloalkyl. [0298] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 7-10 fused bicyclic cycloalkyl. [0299] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is C 5-10 bridged bicyclic cycloalkyl. [0300] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0301] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from - OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0302] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 4-7 membered monocyclic heterocyclyl. [0303] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is phenyl, wherein the phenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is phenyl. [0304] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is naphthalenyl, wherein the naphthalenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is naphthalenyl. [0305] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 5-6 membered monocyclic heteroaryl. [0306] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0307] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0308] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 8-10 membered fused bicyclic heterocyclyl. [0309] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0310] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0311] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 6-10 membered bridged bicyclic heterocyclyl. [0312] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is 7-10 membered spirocyclic heterocyclyl. [0313] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 and each R 8 independently is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0314] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 and each R 8 independently is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl, wherein the 4-membered monocyclic heterocyclyl has one ring heteroatom that is O, and wherein the 5-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, and 6-10 membered bridged bicyclic heterocyclyl each independently have one or two ring heteroatoms that is N. [0315] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 and each R 8 independently is oxetanyl, pyrrolidinyl, piperidinyl, piperazinyl, each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0316] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 and each R 8 independently is optionally substituted with one -NH 2 . [0317] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 13 and each R 8 independently is . [0318] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is imidazolyl or pyridinyl, each of which is independently optionally substituted with 1-2 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0319] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is -F, -NH 2 , -N(CH 3 ) 2 , methyl, [0320] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is -F, -NH 2 , -N(CH 3 ) 2 , methyl, [0321] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is -F, -NH2, methyl, , [0322] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 8 independently is -F, -NH 2 , methyl, [0323] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -C(O)R 9 . [0324] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 2-6 alkenyl and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0325] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 4-7 membered monocyclic heterocyclyl or 6-10 membered bridged bicyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. [0326] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-7 membered monocyclic heterocyclyl or 6-10 membered bridged bicyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl and 6-10 membered bridged bicyclic heterocyclyl are each independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0327] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-4 R b groups. [0328] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 3-7 monocyclic cycloalkyl. [0329] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 7-10 fused bicyclic cycloalkyl. [0330] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is C 5-10 bridged bicyclic cycloalkyl. [0331] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is phenyl, wherein the phenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is phenyl. [0332] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is naphthalenyl, wherein the naphthalenyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is naphthalenyl. [0333] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-6 membered monocyclic heteroaryl. [0334] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 8- 10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 8-10 membered fused bicyclic heterocyclyl. [0335] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 8- 10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 8-10 membered fused bicyclic heteroaryl. [0336] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 7- 10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 7-10 membered spirocyclic heterocyclyl. [0337] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 4- 7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 4-7 membered monocyclic heterocyclyl. [0338] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5- 7 membered monocyclic heterocyclyl, wherein the 5-7 membered monocyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 5-7 membered monocyclic heterocyclyl. [0339] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6- 10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and C 1-5 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is 6-10 membered bridged bicyclic heterocyclyl. [0340] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 9 is pyrrolidinyl, morpholinyl, or , each of which is independently optionally substituted with 1-3 groups independently selected from -OH, halogen, -CN, -NR 11 R 11 , C 1-3 alkoxy, and C 1-3 alkyl. [0341] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 8 is

. [0342] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 8 is -NR 10 R 10 . [0343] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 4 independently is H or C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 4 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 4 is C 1-3 alkyl. [0344] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 7 is H or C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 7 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, R 7 is C 1-3 alkyl. [0345] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 5 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0346] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 5 independently is H or 4-7 membered monocyclic heterocyclyl. [0347] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 5 independently is H or 6 membered monocyclic heterocyclyl, having 1-2 ring heteroatoms independently selected from N, O and S. [0348] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. [0349] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5 is 6 membered monocyclic heterocyclyl, wherein the 6 membered monocyclic heterocyclyl has 1-2 ring heteroatoms independently selected from N, O and S. [0350] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 10 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0351] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 10 independently is H or C 1-6 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 10 independently is H or C 1-3 alkyl. [0352] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6- 10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 10 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. [0353] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 5a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each independently optionally substituted with 1-4 R b groups, wherein the C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-4 R a groups. [0354] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-4 R b groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is phenyl, wherein the phenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-4 R a groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 5a is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-4 R a groups. [0355] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R a independently is oxo, imino, halogen, -NO2, -N3, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C5- 10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5- 6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , -N(R 11 ) 2 (R 11 ) + , -N(R 11 )C(O)R 11 , -N(R 11 )C(O)OR 11 , -N(R 11 )C(O)N(R 11 )(R 11 ), -N(R 11 )S(O) 2 (R 11a ), -NR 11 S(O) 2 N(R 11 )(R 11 ), -NR 11 S(O) 2 O(R 11a ), -OC(O)R 11 , -OC(O)OR 11 , -OC(O)N(R 11 )(R 11 ), -SR 11 , -S(O)R 11a , -S(O)(NH)R 11 , -S(O) 2 R 11a , -S(O) 2 N(R 11 )(R 11 ), or -N=S(R 11a )(R 11a )=O, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups. [0356] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is oxo. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is imino. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is halogen. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -NO 2 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N 3 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -CN. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -C(O)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -C(O)OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -C(O)N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -NR 11 R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N(R 11 ) 2 (R 11 ) + . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N(R 11 )C(O)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N(R 11 )C(O)OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N(R 11 )C(O)N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N(R 11 )S(O) 2 (R 11a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -NR 11 S(O) 2 N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -NR 11 S(O) 2 O(R 11a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -OC(O)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -OC(O)OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -OC(O)N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -SR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -S(O)R 11a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -S(O)(NH)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -S(O) 2 R 11a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -S(O) 2 N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is -N=S(R 11a )(R 11a )=O. [0357] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is phenyl, wherein the phenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R a is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-3 R c groups. [0358] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R b independently is oxo, imino, halogen, -NO2, -N3, -CN, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -C(O)N(R 11 )(R 11 ), -NR 11 R 11 , -N(R 11 ) 2 (R 11 ) + , -N(R 11 )C(O)R 11 , -N(R 11 )C(O)OR 11 , -N(R 11 )C(O)N(R 11 )(R 11 ), -N(R 11 )S(O) 2 (R 11a ), -NR 11 S(O) 2 N(R 11 )(R 11 ), -NR 11 S(O) 2 O(R 11a ), -OC(O)R 11 , -OC(O)OR 11 , -OC(O)N(R 11 )(R 11 ), -SR 11 , -S(O)R 11a , -S(O)(NH)R 11 , -S(O) 2 R 11a , -S(O) 2 N(R 11 )(R 11 ), or -N=S(R 11a )(R 11a )=O, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups. [0359] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R b independently is -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 8-10 membered bridged bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl. [0360] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R b independently is -OH, halogen, -CN, oxo, -NR 11 R 11 , C 1-4 alkoxy, and 5-7 membered monocyclic heterocyclyl. [0361] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R b independently is 4-7 membered monocyclic heterocyclyl, 8-10 membered fused bicyclic heterocyclyl, or 6-10 membered bridged bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 4-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 5-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 8-10 membered fused bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 6-10 membered bridged bicyclic heterocyclyl. [0362] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is oxo. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is imino. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is halogen. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -NO 2 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N3. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -CN. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -C(O)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -C(O)OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -C(O)N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -NR 11 R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N(R 11 ) 2 (R 11 ) + . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N(R 11 )C(O)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N(R 11 )C(O)OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N(R 11 )C(O)N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N(R 11 )S(O) 2 (R 11a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -NR 11 S(O) 2 N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -NR 11 S(O) 2 O(R 11a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -OC(O)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -OC(O)OR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -OC(O)N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -SR 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -S(O)R 11a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -S(O)(NH)R 11 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -S(O) 2 R 11a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -S(O) 2 N(R 11 )(R 11 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is -N=S(R 11a )(R 11a )=O. [0363] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is phenyl, wherein the phenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R b is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-3 R c groups. [0364] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R c independently is halogen, -CN, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, 7-10 membered spirocyclic heterocyclyl, -OR 12 , -C(O)R 12 , -C(O)OR 12 , -C(O)N(R 12 )(R 12 ), -NR 12 R 12 , -N(R 12 ) 2 (R 12 ) + , -N(R 12 )C(O)R 12 , -N(R 12 )C(O)OR 12 , -N(R 12 )C(O)N(R 12 )(R 12 ), -N(R 12 )S(O) 2 (R 12a ), -NR 12 S(O) 2 N(R 12 )(R 12 ), -NR 12 S(O) 2 O(R 12a ), -OC(O)R 12 , -OC(O)OR 12 , -OC(O)N(R 12 )(R 12 ), -SR 12 , -S(O)R 12a , -S(O)(NH)R 12 , -S(O) 2 R 12a , -S(O) 2 N(R 12 )(R 12 ), or -N=S(R 12a )(R 12a )=O. [0365] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is halogen. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -CN. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is C 7-10 fused bicyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is C 5-10 bridged bicyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is phenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is naphthalenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is 4-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is 5-6 membered monocyclic heteroaryl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is 8-10 membered fused bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is 6-10 membered bridged bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is 8-10 membered fused bicyclic heteroaryl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is 7-10 membered spirocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -OR 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -C(O)R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -C(O)OR 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -C(O)N(R 12 )(R 12 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -NR 12 R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -N(R 12 ) 2 (R 12 ) + . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -N(R 12 )C(O)R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -N(R 12 )C(O)OR 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -N(R 12 )C(O)N(R 12 )(R 12 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -N(R 12 )S(O) 2 (R 12a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -NR 12 S(O) 2 N(R 12 )(R 12 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -NR 12 S(O) 2 O(R 12a ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -OC(O)R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -OC(O)OR 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -OC(O)N(R 12 )(R 12 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -SR 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -S(O)R 12a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -S(O)(NH)R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -S(O) 2 R 12a . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -S(O) 2 N(R 12 )(R 12 ). In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -N=S(R 12a )(R 12a )=O. [0366] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R c independently is -OH, halogen, -CN, -NR 12 R 12 , or C 1-4 alkoxy. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is -OH. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R c is C 1-4 alkoxy. [0367] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 11 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups. [0368] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 11 independently is H or C 1-4 alkyl, wherein the C 1-4 alkyl is optionally substituted with one group selected from -OH and -NR 12 R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 11 independently is H or methyl. [0369] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 1-4 alkyl, wherein the C 1-4 alkyl is optionally substituted with one group selected from -OH and -NR 12 R 12 . In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is methyl. [0370] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is phenyl, wherein the phenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11 is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-3 R c groups. [0371] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 11a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl, wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8-10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, and 7-10 membered spirocyclic heterocyclyl are each independently optionally substituted with 1-3 R c groups. [0372] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is C 2-6 alkenyl, wherein the C 2-6 alkenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is C 2-6 alkynyl, wherein the C 2-6 alkynyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is C 3-7 monocyclic cycloalkyl, wherein the C 3-7 monocyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is C 7-10 fused bicyclic cycloalkyl, wherein the C 7-10 fused bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is C 5-10 bridged bicyclic cycloalkyl, wherein the C 5-10 bridged bicyclic cycloalkyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is phenyl, wherein the phenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is naphthalenyl, wherein the naphthalenyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is 4-7 membered monocyclic heterocyclyl, wherein the 4-7 membered monocyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is 5-6 membered monocyclic heteroaryl, wherein the 5-6 membered monocyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is 8-10 membered fused bicyclic heterocyclyl, wherein the 8-10 membered fused bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is 6-10 membered bridged bicyclic heterocyclyl, wherein the 6-10 membered bridged bicyclic heterocyclyl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is 8-10 membered fused bicyclic heteroaryl, wherein the 8-10 membered fused bicyclic heteroaryl is optionally substituted with 1-3 R c groups. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 11a is 7-10 membered spirocyclic heterocyclyl, wherein the 7-10 membered spirocyclic heterocyclyl is optionally substituted with 1-3 R c groups. [0373] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 12 independently is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl. [0374] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 12 independently is H or C 1-3 alkyl. [0375] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is H. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 1-6 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 1-3 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 2-6 alkenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 2-6 alkynyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 3-7 monocyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 7-10 fused bicyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is C 5-10 bridged bicyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is phenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is naphthalenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is 4-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is 5-6 membered monocyclic heteroaryl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is 8-10 membered fused bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is 6-10 membered bridged bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is 8-10 membered fused bicyclic heteroaryl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12 is 7-10 membered spirocyclic heterocyclyl. [0376] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 12a independently is C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 monocyclic cycloalkyl, C 7-10 fused bicyclic cycloalkyl, C 5-10 bridged bicyclic cycloalkyl, phenyl, naphthalenyl, 4-7 membered monocyclic heterocyclyl, 5-6 membered monocyclic heteroaryl, 8- 10 membered fused bicyclic heterocyclyl, 6-10 membered bridged bicyclic heterocyclyl, 8-10 membered fused bicyclic heteroaryl, or 7-10 membered spirocyclic heterocyclyl. [0377] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is C 1-6 alkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is C 2-6 alkenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is C 2-6 alkynyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is C 3-7 monocyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is C 7-10 fused bicyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is C 5-10 bridged bicyclic cycloalkyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is phenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is naphthalenyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is 4-7 membered monocyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is 5-6 membered monocyclic heteroaryl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is 8-10 membered fused bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is 6-10 membered bridged bicyclic heterocyclyl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is 8-10 membered fused bicyclic heteroaryl. In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, one R 12a is 7-10 membered spirocyclic heterocyclyl. [0378] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, each R 5 independently is H or 6 membered monocyclic heterocyclyl, having 1-2 ring heteroatoms independently selected from N, O and S. [0379] In some embodiments of a compound of Formula I, or a pharmaceutically acceptable salt thereof, L 1 is [0380] In one embodiment, provided herein is a compound selected from the group consisting of:

[0381] In one embodiment, provided herein is a compound selected from the group consisting of:

or a pharmaceutically acceptable salt thereof. [0382] In one embodiment, provided herein is a compound selected from the group consisting of: or a pharmaceutically acceptable salt thereof. [0383] In one embodiment, provided herein is a compound selected from the group consisting of: , or a pharmaceutically acceptable salt thereof. [0384] In one embodiment, provided herein is a compound selected from the group consisting of: , ,

, or a pharmaceutically acceptable salt thereof. [0385] In one embodiment, provided herein is a compound selected from the group consisting of: , or a pharmaceutically acceptable salt thereof. [0386] In one embodiment, provided herein is a compound: or a pharmaceutically acceptable salt thereof. III. Compositions and Kits [0387] Compounds provided herein, or pharmaceutically acceptable salts thereof, are usually administered in the form of pharmaceutical compositions. Thus, provided herein are also pharmaceutical compositions that comprise one or more of the compounds provided herein or pharmaceutically acceptable salts, isomer, or a mixture thereof and one or more pharmaceutically acceptable vehicles selected from carriers, adjuvants and excipients. The compounds provided herein, or pharmaceutically acceptable salts thereof, may be the sole active ingredient or one of the active ingredients of the pharmaceutical compositions. Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants. Such compositions are prepared in a manner well known in the pharmaceutical art. See, e.g., Remington’s Pharmaceutical Sciences, Mace Publishing Co., Philadelphia, Pa.17th Ed. (1985); and Modern Pharmaceutics, Marcel Dekker, Inc.3rd Ed. (G.S. Banker & C.T. Rhodes, Eds.). [0388] In one embodiment, provided herein are pharmaceutical compositions comprising a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. In some embodiments, the pharmaceutical compositions comprise a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. [0389] In some embodiments, the pharmaceutical compositions provided herein further comprise one or more (i.e., one, two, three, four; one or two; one to three; or one to four) additional therapeutic agents, or a pharmaceutically acceptable salt thereof. In some embodiments, the pharmaceutical compositions further comprise a therapeutically effective amount of the one or more (i.e., one, two, three, four; one or two; one to three; or one to four) additional therapeutic agents, or a pharmaceutically acceptable salt thereof. [0390] In some embodiments, the one or more additional therapeutic agents comprises an anti-malarial agent. In some embodiments, the anti-malarial agent is selected from chloroquine and hydroxychloroquine, or a pharmaceutically acceptable salt thereof. [0391] In some embodiments, the one or more additional therapeutic agents include agents that are therapeutic for an inflammatory condition. In some embodiments, the one or more additional therapeutic agents is selected from the group consisting of: veltuzumab, PF- 06835375, eculizumab, milatuzumab, SM-06, SM-03, BT-063, QX-006-N, BOS-161721, AK- 101, TNX-1500, theralizumab, daxdilimab, TAK-079, felzartamab, itolizumab, anifrolumab, iscalimab, dapirolizumab pegol, lanalumab, LY-3361237, JNJ-55920839, UBP-1213, DS-7011, PFI-102, BIIB-059, obexelimab, talacotuzumab, vobarilizumab, TE-2324, PRV-3279, chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, AVO-101, rozibafusp alfa, VRN-02, annexuzlimab, ALPN-101, bendamustine hydrochloride, BMS-986256, NKTR-35, atacicept, telitacicept, BMS-986256, M-5049, KZR-616, KPG-818, verdinexor, ALPN-303, valziflocept, LA-1, cenerimod, prednisone, corticotropin, deucravacitinib, CPL-409116, CS-12192, tofacitinib citrate, ISB-830, DV-1079, julemic acid, iberdomide, TAM-01, BML-258, brepocitinib, SDC-1801, SDC-1802, ICP-330, NTR-441, dalazatide, GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, HMPL-523, RSLV- 132, interleukin-2 follow-on biologic, interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, CUG-252, DZ-2002, PEGylated HLA-x (SLE), AC-0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, orelabrutinib, DWP-213388, INV-103, R-salbutamol sulphate, anchorins, NIK-SMI1, X-6, INV-17, Oshadi D, baricitinib, upadacitinib, filgotinib, itacitinib, INCB-54707, delgocitinib, DWP-212525, CKD-971, as mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT- 013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, and AMG-592, or a pharmaceutically acceptable salt of any of the foregoing, or any combinations thereof. [0392] The pharmaceutical compositions may be administered in either single or multiple doses. The pharmaceutical compositions may be administered by various methods including, for example, rectal, buccal, intranasal and transdermal routes. In some embodiments, the pharmaceutical compositions may be administered by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant. [0393] One mode for administration is parenteral, for example, by injection. The forms in which the pharmaceutical compositions described herein may be incorporated for administration by injection include, for example, aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles. In some embodiments, the compounds, or pharmaceutically acceptable salts thereof, and pharmaceutical compositions disclosed herein are administered by subcutaneous injection. [0394] The pharmaceutical compositions of the present disclosure may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned herein. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. [0395] In some embodiments, the sterile injectable preparation disclosed herein may also be a sterile injectable solution or suspension prepared from a reconstituted lyophilized powder in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. [0396] Formulations suitable for parenteral administration include aqueous and non- aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. In certain embodiments the suspension is a microsuspension. In certain embodiments the suspension is a nanosuspension. [0397] In some embodiments, formulations suitable for parenteral administration (e.g., intramuscular (IM) and subcutaneous (SC) administration) will include one or more excipients. Excipients should be compatible with the other ingredients of the formulation and physiologically innocuous to the recipient thereof. Examples of suitable excipients are well known to the person skilled in the art of parenteral formulation and may be found e.g., in Handbook of Pharmaceutical Excipients (eds. Rowe, Sheskey & Quinn), 6th edition 2009. Examples of solubilizing excipients in a parenteral formulation (e.g., an SC or IM formulation) include, but are not limited to, polysorbates (such as polysorbate 20 or 80) and poloxamers (such as poloxamer 338, 188, or 207). [0398] In some embodiments, the compounds, or pharmaceutically acceptable salts thereof, and pharmaceutical compositions disclosed herein are administered with implants. [0399] Oral administration may be another route for administration of the compounds provided herein or pharmaceutically acceptable salts thereof. Administration may be via, for example, capsule or enteric coated tablets. In making the pharmaceutical compositions that include at least one compound provided herein or pharmaceutically acceptable salts, isomer, or a mixture thereof, the active ingredient (such as a compound provided herein) is usually diluted by an excipient and/or enclosed within such a carrier that can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the pharmaceutical 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, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders. [0400] 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, sterile water, syrup, and methyl cellulose or any combinations thereof. The pharmaceutical compositions 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; or any combinations thereof. [0401] The pharmaceutical compositions that include at least one compound described herein or pharmaceutically acceptable salts, isomer, or a mixture thereof can be formulated so as to provide quick, sustained or delayed release of the active ingredient (such as a compound provided herein) after administration to the subject by employing procedures known in the art. Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Patent Nos.3,845,770; 4,326,525; 4,902,514; and 5,616,345. Another formulation for use in the methods of the present disclosure employs transdermal delivery devices (“patches”). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds provided herein in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent Nos.5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. [0402] For preparing solid compositions such as tablets, the principal active ingredient may be mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound described herein or pharmaceutically acceptable salts, isomer, or a mixture thereof. When referring to these preformulation compositions as homogeneous, the active ingredient may be dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. [0403] The tablets or pills of the compounds provided herein or pharmaceutically acceptable salts thereof may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action, or to protect from the acid conditions of the stomach. For example, the tablet or pill can include 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 that 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 materials such as shellac, cetyl alcohol, and cellulose acetate. [0404] Pharmaceutical compositions for inhalation or insufflation may 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. In other embodiments, compositions in pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a facemask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner. [0405] In one embodiment, provided herein are kits that comprise a compound provided herein, (i.e., a compound of Formula I), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, and suitable packaging. In some embodiments, the kit further comprises instructions for use. In some embodiments, the kit comprises a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, and a label and/or instructions for use of the compounds in the treatment of the indications, including the diseases or conditions, described herein. [0406] In some embodiments, the kits further comprise one or more (i.e., one, two, three, four; one or two; one to three; or one to four) additional therapeutic agents, or a pharmaceutically acceptable salt thereof. [0407] In one embodiment, provided herein are articles of manufacture that comprise a compound described herein or pharmaceutically acceptable salts, isomer, or a mixture thereof in a suitable container. In some embodiments, the container may be a vial, jar, ampoule, preloaded syringe, or intravenous bag. IV. Methods [0408] The methods provided herein may be applied to cell populations in vivo or ex vivo. “In vivo” means within a living individual, as within an animal or human. In this context, the methods provided herein may be used therapeutically in an individual. “Ex vivo” means outside of a living individual. Examples of ex vivo cell populations include in vitro cell cultures and biological samples including fluid or tissue samples obtained from individuals. Such samples may be obtained by methods well known in the art. Exemplary biological fluid samples include blood, cerebrospinal fluid, urine, and saliva. Exemplary tissue samples include tumors and biopsies thereof. In this context, the present disclosure may be used for a variety of purposes, including therapeutic and experimental purposes. For example, the present disclosure may be used ex vivo to determine the optimal schedule and/or dosing of administration of a TLR 7, 8, and/or 9 inhibitor as disclosed herein for a given cell type, individual, and other parameters. Information gleaned from such use may be used for experimental purposes or in the clinic to set protocols for in vivo treatment. Other ex vivo uses for which the present disclosure may be suited are described below or will become apparent to those skilled in the art. The selected compounds may be further characterized to examine the safety or tolerance dosage in human or non-human subjects. Such properties may be examined using commonly known methods to those skilled in the art. [0409] In one embodiment, the present disclosure provides a method of inhibiting toll- like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of inhibiting toll-like receptor 7, 8, and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of inhibiting toll-like receptor 7, 8, or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0410] In one embodiment, the present disclosure provides a method of inhibiting toll- like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of inhibiting toll-like receptor 7 and 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of inhibiting toll-like receptor 7 or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0411] In one embodiment, the present disclosure provides a method of inhibiting toll- like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of inhibiting toll-like receptor 7 and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of inhibiting toll-like receptor 7 or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0412] In one embodiment, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0413] In one embodiment, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7 and 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7 or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0414] In one embodiment, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7 and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the present disclosure provides a method of treating a disease or disorder associated with elevated toll-like receptor 7 or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0415] In one embodiment, the present disclosure provides a method of treating an inflammatory condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein [0416] Non-limiting examples of an inflammatory condition include, without limitation, acne, acid-induced lung injury, Addison's disease, adrenal hyperplasia, adrenocortical insufficiency, adult-onset Still's disease, adult respiratory distress syndrome (ARDS), age- related macular degeneration, aging, alcoholic hepatitis, alcoholic liver disease, allergen-induced asthma, allergic bronchopulmonary, allergic conjunctivitis, allergic contact dermatitis, allergies, allergic encephalomyelitis, allergic neuritis, allograft rejection, alopecia, alopecia areata, Alzheimer's disease, amyloidosis, amyotrophic lateral sclerosis, angina pectoris, angioedema, angiofibroma, anhidrotic ectodermal dysplasia-ill, anti-glomerular basement membrane disease, antigen-antibody complex mediated diseases, ankylosing spondylitis, antiphospholipid syndrome, aphthous stomatitis, appendicitis, arthritis, ascites, aspergillosis, asthma, atherosclerosis, atherosclerotic plaques, atopic dermatitis, atrophic thyroiditis, autoimmune diseases, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune polyendocrinopathies, autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), autoimmune hepatitis, autoimmune thyroid disorders, autoinflammatory diseases, back pain, Bacillus anthracis infection, Bechet's disease, bee sting-induced inflammation, Behçet’s syndrome, Bell’s palsy, berylliosis, Blau syndrome, bone pain, bronchiolitis, bullous pemphigoid (BP) asthma, burns, bursitis, cardiac hypertrophy, carpal tunnel syndrome, Castleman's disease, catabolic disorders, cataracts, Celiac disease, cerebral aneurysm, chemical irritant-induced inflammation, chorioretinitis, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome, chronic heart failure, chronic lung disease of prematurity, chronic obstructive pulmonary disease (COPD), chronic pancreatitis, chronic prostatitis, chronic recurrent multifocal osteomyelitis, cicatricial alopecia, colitis, complex regional pain syndrome, complications of organ transplantation, conjunctivitis, connective tissue disease, contact dermatitis, corneal graft neovascularization, corneal ulcer, Crohn's disease, cryopyrin-associated periodic syndromes, cutaneous lupus erythematosus (CLE), cryptococcosis, cystic fibrosis, deficiency of the interleukin-1 receptor antagonist (DIRA), dermatitis, dermatitis endotoxemia, dermatomyositis, diabetic macular edema, diverticulitis, eczema, encephalitis, endometriosis, endotoxemia, eosinophilic pneumonias, epicondylitis, epidermolysis bullosa, erythema multiforme, erythroblastopenia, esophagitis, familial amyloidotic polyneuropathy, familial cold urticarial, familial Mediterranean fever, fetal growth retardation, fibromyalgia, fistulizing Crohn’s disease, food allergies, giant cell arteritis, glaucoma, glioblastoma, glomerular disease, glomerular nephritis, glomerulonephritis, gluten-sensitive enteropathy, gout, gouty arthritis, graft-versus-host disease (GVHD), granulomatous hepatitis, Graves' disease, growth plate injuries, Guillain-Barre syndrome. gut diseases, hair loss, Hashimoto's thyroiditis, head injury, headache, hearing loss, heart disease, hemangioma, hemolytic anemia, hemophilic joints, Henoch-Scholein purpura, hepatitis, hereditary periodic fever syndrome, heritable disorders of connective tissue, herpes zoster and simplex, hidradenitis suppurativa (HS), hip replacement, Hodgkin's disease, Huntington's disease, hyaline membrane disease, hyperactive inflammatory response, hyperammonemia, hypercalcemia, hypercholesterolemia, hypereosinophilic syndrome (HES), hyperimmunoglobulinemia D with recurrent fever (HIDS), hypersensitivity pneumonitis, hypertropic bone formation, hypoplastic and other anemias, hypoplastic anemia, ichthyosis, idiopathic demyelinating polyneuropathy, Idiopathic inflammatory myopathies (dermatomyositis, polymyositis), idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura, immunoglobulin nephropathies, immune complex nephritis, immune thrombocytopenic purpura (ITP), incontinentia pigmenti (IP, Bloch–Siemens syndrome), infectious mononucleosis, infectious diseases including viral diseases such as AIDS (HIV infection), hepatitis A, B, C, D, and E, herpes; inflammation, inflammation of the CNS, inflammatory bowel disease (IBD), inflammatory disease of the lower respiratory tract including bronchitis or chronic obstructive pulmonary diseases, inflammatory disease of the upper respiratory tract including the nose and sinuses such as rhinitis or sinusitis, inflammatory diseases of the respiratory tract, inflammatory ischemic event such as stroke or cardiac arrest, inflammatory lung disease, inflammatory myopathy such as myocarditis, inflammatory liver disease, inflammatory neuropathy, inflammatory pain, insect bite-induced inflammation, interstitial cystitis, interstitial lung disease, iritis, irritant-induced inflammation, ischemia/reperfusion, joint replacement, juvenile arthritis, juvenile rheumatoid arthritis, keratitis, kidney injury caused by parasitic infections, kidney transplant rejection, leptospirosis, leukocyte adhesion deficiency, lichen sclerosus (LS), Lambert-Eaton myasthenic syndrome, Loeffler's syndrome, lupus, lupus nephritis, Lyme disease, Marfan syndrome (MFS), mast cell activation syndrome, mastocytosis, meningitis, meningioma, mesothelioma, mixed connective tissue disease, Muckle-Wells syndrome (urticaria deafness amyloidosis), mucositis, multiple organ injury syndrome, multiple sclerosis, muscle wasting, muscular dystrophy, myasthenia gravis (MG), myelodysplastic syndrome, myocarditis, myositis, nasal sinusitis, necrotizing enterocolitis, neonatal onset multisystem inflammatory disease (NOMID), neovascular glaucoma, nephrotic syndrome, neuritis, neuropathological diseases, non-allergen induced asthma, obesity, ocular allergy, optic neuritis, organ transplant rejection, Osier-Weber syndrome, osteoarthritis, osteogenesis imperfecta, osteonecrosis, osteoporosis, osterarthritis, otitis, pachyonychia congenita, Paget’s disease, Paget’s disease of bone, pancreatitis, Parkinson's disease, pediatric rheumatology, pelvic inflammatory disease, pemphigus, pemphigus vulgaris (PV), bullous pemphigoid (BP), pericarditis, periodic fever, periodontitis, peritoneal endometriosis, pernicious anemia (Addison's disease), pertussis, PFAPA (periodic fever aphthous pharyngitis and cervical adenopathy), pharyngitis and adenitis (PFAPA syndrome), plant irritant-induced inflammation, pneumocystis infection, pneumonia, pneumonitis, poison ivy/ urushiol oil-induced inflammation, polyarthritis nodosa, polychondritis, polycystic kidney disease, polymyalgia rheumatic, giant cell arteritis, polymyositis, pouchitis, reperfusion injury and transplant rejection, primary biliary cirrhosis, primary pulmonary hypertension, primary sclerosing cholangitis (PSC), proctitis, psoriasis, psoriasis vulgaris, psoriatic arthritis, psoriatic epidermis, psychosocial stress diseases, pulmonary disease, pulmonary fibrosis, pulmonary hypertension, pyoderma gangrenosum, pyogenic granuloma retrolental fibroplasias, pyogenic sterile arthritis, Raynaud’s syndrome, Reiter's disease, reactive arthritis, renal disease, renal graft rejection, reperfusion injury, respiratory distress syndrome, retinal disease, retrolental fibroplasia, Reynaud's syndrome, rheumatic carditis, rheumatic diseases, rheumatic fever, rheumatoid arthritis, rhinitis, rhinitis psoriasis, rosacea, sarcoidosis, Schnitzler syndrome, scleritis, sclerosis, scleroderma, scoliosis, seborrhea, sepsis, septic shock, severe pain, Sézary syndrome, sickle cell anemia, silica-induced disease (Silicosis), Sjogren's syndrome, skin diseases, skin irritation, skin rash, skin sensitization (contact dermatitis or allergic contact dermatitis), sleep apnea, spinal cord injury, spinal stenosis, spondyloarthropathies, sports injuries, sprains and strains, Stevens-Johnson syndrome (SJS), stroke, subarachnoid hemorrhage, sunburn, synovial inflammation, systemic inflammatory response syndrome (SIRS), systemic lupus erythematosus (SLE), systemic mast cell disease (SMCD), systemic vasculitis, systemic-onset juvenile idiopathic arthritis, temporal arteritis, tendinitis, tenosynovitis, thrombocytopenia, thyroditis, thyroiditis, tissue transplant, toxoplasmosis, trachoma, transplantation rejection, traumatic brain injury, tuberculosis, tubulointerstitial nephritis, tumor necrosis factor (TNF) receptor associated periodic syndrome (TRAPS), type 1 diabetes, type 2 diabetes, complications from type 1 or type 2 diabetes, ulcerative colitis, urticaria, uterine fibroids, uveitis, uveoretinitis, vascular restenosis, vasculitis, vasculitis (NHLBI), vitiligo, Wegener's granulomatosis, and Whipple's disease. [0417] In some embodiments, the inflammatory condition is selected from inflammatory bowel disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, glomerulonephritis, mixed connective tissue disease (MCTD), dermatomyositis, polymyositis, systemic sclerosis, antineutrophil cytoplasmic antibody-associated vasculitis, anti-phospholipid syndrome, autoimmune hemolytic anemia, macrophage activation syndrome driven inflammatory anemia, IgA nephropathy, type I diabetes, non-alcoholic steatohepatitis, and Sjogren’s syndrome. In some embodiments, the inflammatory condition is inflammatory bowel disease. In some embodiments, the inflammatory condition is psoriasis. In some embodiments, the inflammatory condition is psoriatic arthritis. In some embodiments, the inflammatory condition is rheumatoid arthritis. In some embodiments, the inflammatory condition is glomerulonephritis. In some embodiments, the inflammatory condition is mixed connective tissue disease (MCTD). In some embodiments, the inflammatory condition is dermatomyositis. In some embodiments, the inflammatory condition is polymyositis. In some embodiments, the inflammatory condition is systemic sclerosis. In some embodiments, the inflammatory condition is antineutrophil cytoplasmic antibody-associated vasculitis. In some embodiments, the inflammatory condition is anti-phospholipid syndrome. In some embodiments, the inflammatory condition is autoimmune hemolytic anemia. In some embodiments, the inflammatory condition is macrophage activation syndrome driven inflammatory anemia. In some embodiments, the inflammatory condition is IgA nephropathy. In some embodiments, the inflammatory condition is type I diabetes. In some embodiments, the inflammatory condition is non-alcoholic steatohepatitis. In some embodiments, the inflammatory condition is Sjogren’s syndrome. [0418] The compounds provided herein, or pharmaceutically acceptable salts thereof, or the pharmaceutical composition provided herein may treat or ameliorate systemic lupus erythematosus (SLE), cutaneous lupus erythematosus (CLE), lupus nephritis, lupus-related, symptom of SLE, symptom of CLE, or other autoimmune disorder. Symptoms of systemic lupus erythematosus include joint pain, joint swelling, arthritis, fatigue, hair loss, mouth sores, swollen lymph nodes, sensitivity to sunlight, skin rash, headaches, numbness, tingling, seizures, vision problems, personality changes, abdominal pain, nausea, vomiting, abnormal heart rhythms, coughing up blood and difficulty breathing, patchy skin color and Raynaud's phenomenon. [0419] In one embodiment, the present disclosure provides a method of treating systemic lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0420] In one embodiment, the present disclosure provides a method of treating cutaneous lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0421] In one embodiment, the present disclosure provides a method of treating lupus nephritis in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0422] In some embodiments, the methods provided herein further comprise administering a therapeutically effective amount of one or more additional therapeutic agents, or a pharmaceutically acceptable salt thereof. [0423] In some embodiments, the one or more additional therapeutic agents is selected from the group consisting of veltuzumab, PF-06835375, eculizumab, milatuzumab, SM-06, SM- 03, BT-063, QX-006-N, BOS-161721, AK-101, TNX-1500, theralizumab, daxdilimab, TAK- 079, felzartamab, itolizumab, anifrolumab, iscalimab, dapirolizumab pegol, lanalumab, LY- 3361237, JNJ-55920839, UBP-1213, DS-7011, PFI-102, BIIB-059, obexelimab, talacotuzumab, vobarilizumab, TE-2324, PRV-3279, chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, AVO-101, rozibafusp alfa, VRN-02, annexuzlimab, ALPN- 101, bendamustine hydrochloride, BMS-986256, NKTR-35, atacicept, telitacicept, BMS- 986256, M-5049, KZR-616, KPG-818, verdinexor, ALPN-303, valziflocept, LA-1, cenerimod, prednisone, corticotropin, deucravacitinib, CPL-409116, CS-12192, tofacitinib citrate, ISB-830, DV-1079, julemic acid, iberdomide, TAM-01, BML-258, brepocitinib, SDC-1801, SDC-1802, ICP-330, NTR-441, dalazatide, GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, HMPL-523, RSLV-132, interleukin-2 follow-on biologic, interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, CUG-252, DZ-2002, PEGylated HLA-x (SLE), AC- 0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, orelabrutinib, DWP-213388, INV-103, R-salbutamol sulphate, anchorins, NIK-SMI1, X-6, INV- 17, Oshadi D, baricitinib, upadacitinib, filgotinib, itacitinib, INCB-54707, delgocitinib, DWP- 212525, CKD-971, as mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT-013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, and AMG-592, or a pharmaceutically acceptable salt of any of the foregoing, or any combination thereof. [0424] In some embodiments, the one or more additional therapeutic agents is selected from chloroquine and hydroxychloroquine, or a pharmaceutically acceptable salt thereof. In some embodiments, the one or more additional therapeutic agents is chloroquine. In some embodiments, the one or more additional therapeutic agents is hydroxychloroquine. In some embodiments, the one or more additional therapeutic agent is a pharmaceutically acceptable salt of hydroxychloroquine. In some embodiments, the one or more additional therapeutic agent is hydroxychloroquine sulfate. [0425] In some embodiments of the methods provided herein, the subject is a human. [0426] In some embodiments, the methods provided herein comprise administering a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof. In some embodiments, the methods provided herein comprise administering a therapeutically effective amount of a pharmaceutical composition provided herein. [0427] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein for use in therapy. [0428] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of inhibiting toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of inhibiting toll-like receptor 7, 8, and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of inhibiting toll-like receptor 7, 8, or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0429] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of inhibiting toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of inhibiting toll-like receptor 7 and 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of inhibiting toll-like receptor 7 or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0430] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of inhibiting toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of inhibiting toll-like receptor 7 and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of inhibiting toll-like receptor 7 or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0431] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7, 8, or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0432] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 or 8 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0433] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and/or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 and 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. In some embodiments, the compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition provided herein is for use in a method of treating a disease or disorder associated with elevated toll-like receptor 7 or 9 activity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0434] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of treating an inflammatory condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0435] Non-limiting examples of an inflammatory condition include, without limitation, acne, acid-induced lung injury, Addison's disease, adrenal hyperplasia, adrenocortical insufficiency, adult-onset Still's disease, adult respiratory distress syndrome (ARDS), age- related macular degeneration, aging, alcoholic hepatitis, alcoholic liver disease, allergen-induced asthma, allergic bronchopulmonary, allergic conjunctivitis, allergic contact dermatitis, allergies, allergic encephalomyelitis, allergic neuritis, allograft rejection, alopecia, alopecia areata, Alzheimer's disease, amyloidosis, amyotrophic lateral sclerosis, angina pectoris, angioedema, angiofibroma, anhidrotic ectodermal dysplasia-ill, anti-glomerular basement membrane disease, antigen-antibody complex mediated diseases, ankylosing spondylitis, antiphospholipid syndrome, aphthous stomatitis, appendicitis, arthritis, ascites, aspergillosis, asthma, atherosclerosis, atherosclerotic plaques, atopic dermatitis, atrophic thyroiditis, autoimmune diseases, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune polyendocrinopathies, autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), autoimmune hepatitis, autoimmune thyroid disorders, autoinflammatory diseases, back pain, Bacillus anthracis infection, Bechet's disease, bee sting-induced inflammation, Behçet’s syndrome, Bell’s palsy, berylliosis, Blau syndrome, bone pain, bronchiolitis, bullous pemphigoid (BP) asthma, burns, bursitis, cardiac hypertrophy, carpal tunnel syndrome, Castleman's disease, catabolic disorders, cataracts, Celiac disease, cerebral aneurysm, chemical irritant-induced inflammation, chorioretinitis, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome, chronic heart failure, chronic lung disease of prematurity, chronic obstructive pulmonary disease (COPD), chronic pancreatitis, chronic prostatitis, chronic recurrent multifocal osteomyelitis, cicatricial alopecia, colitis, complex regional pain syndrome, complications of organ transplantation, conjunctivitis, connective tissue disease, contact dermatitis, corneal graft neovascularization, corneal ulcer, Crohn's disease, cryopyrin-associated periodic syndromes, cutaneous lupus erythematosus (CLE), cryptococcosis, cystic fibrosis, deficiency of the interleukin-1 receptor antagonist (DIRA), dermatitis, dermatitis endotoxemia, dermatomyositis, diabetic macular edema, diverticulitis, eczema, encephalitis, endometriosis, endotoxemia, eosinophilic pneumonias, epicondylitis, epidermolysis bullosa, erythema multiforme, erythroblastopenia, esophagitis, familial amyloidotic polyneuropathy, familial cold urticarial, familial Mediterranean fever, fetal growth retardation, fibromyalgia, fistulizing Crohn’s disease, food allergies, giant cell arteritis, glaucoma, glioblastoma, glomerular disease, glomerular nephritis, glomerulonephritis, gluten-sensitive enteropathy, gout, gouty arthritis, graft-versus-host disease (GVHD), granulomatous hepatitis, Graves' disease, growth plate injuries, Guillain-Barre syndrome. gut diseases, hair loss, Hashimoto's thyroiditis, head injury, headache, hearing loss, heart disease, hemangioma, hemolytic anemia, hemophilic joints, Henoch-Scholein purpura, hepatitis, hereditary periodic fever syndrome, heritable disorders of connective tissue, herpes zoster and simplex, hidradenitis suppurativa (HS), hip replacement, Hodgkin's disease, Huntington's disease, hyaline membrane disease, hyperactive inflammatory response, hyperammonemia, hypercalcemia, hypercholesterolemia, hypereosinophilic syndrome (HES), hyperimmunoglobulinemia D with recurrent fever (HIDS), hypersensitivity pneumonitis, hypertropic bone formation, hypoplastic and other anemias, hypoplastic anemia, ichthyosis, idiopathic demyelinating polyneuropathy, Idiopathic inflammatory myopathies (dermatomyositis, polymyositis), idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura, immunoglobulin nephropathies, immune complex nephritis, immune thrombocytopenic purpura (ITP), incontinentia pigmenti (IP, Bloch–Siemens syndrome), infectious mononucleosis, infectious diseases including viral diseases such as AIDS (HIV infection), hepatitis A, B, C, D, and E, herpes; inflammation, inflammation of the CNS, inflammatory bowel disease (IBD), inflammatory disease of the lower respiratory tract including bronchitis or chronic obstructive pulmonary diseases, inflammatory disease of the upper respiratory tract including the nose and sinuses such as rhinitis or sinusitis, inflammatory diseases of the respiratory tract, inflammatory ischemic event such as stroke or cardiac arrest, inflammatory lung disease, inflammatory myopathy such as myocarditis, inflammatory liver disease, inflammatory neuropathy, inflammatory pain, insect bite-induced inflammation, interstitial cystitis, interstitial lung disease, iritis, irritant-induced inflammation, ischemia/reperfusion, joint replacement, juvenile arthritis, juvenile rheumatoid arthritis, keratitis, kidney injury caused by parasitic infections, kidney transplant rejection, leptospirosis, leukocyte adhesion deficiency, lichen sclerosus (LS), Lambert-Eaton myasthenic syndrome, Loeffler's syndrome, lupus, lupus nephritis, Lyme disease, Marfan syndrome (MFS), mast cell activation syndrome, mastocytosis, meningitis, meningioma, mesothelioma, mixed connective tissue disease, Muckle-Wells syndrome (urticaria deafness amyloidosis), mucositis, multiple organ injury syndrome, multiple sclerosis, muscle wasting, muscular dystrophy, myasthenia gravis (MG), myelodysplastic syndrome, myocarditis, myositis, nasal sinusitis, necrotizing enterocolitis, neonatal onset multisystem inflammatory disease (NOMID), neovascular glaucoma, nephrotic syndrome, neuritis, neuropathological diseases, non-allergen induced asthma, obesity, ocular allergy, optic neuritis, organ transplant rejection, Osier-Weber syndrome, osteoarthritis, osteogenesis imperfecta, osteonecrosis, osteoporosis, osterarthritis, otitis, pachyonychia congenita, Paget’s disease, Paget’s disease of bone, pancreatitis, Parkinson's disease, pediatric rheumatology, pelvic inflammatory disease, pemphigus, pemphigus vulgaris (PV), bullous pemphigoid (BP), pericarditis, periodic fever, periodontitis, peritoneal endometriosis, pernicious anemia (Addison's disease), pertussis, PFAPA (periodic fever aphthous pharyngitis and cervical adenopathy), pharyngitis and adenitis (PFAPA syndrome), plant irritant-induced inflammation, pneumocystis infection, pneumonia, pneumonitis, poison ivy/ urushiol oil-induced inflammation, polyarthritis nodosa, polychondritis, polycystic kidney disease, polymyalgia rheumatic, giant cell arteritis, polymyositis, pouchitis, reperfusion injury and transplant rejection, primary biliary cirrhosis, primary pulmonary hypertension, primary sclerosing cholangitis (PSC), proctitis, psoriasis, psoriasis vulgaris, psoriatic arthritis, psoriatic epidermis, psychosocial stress diseases, pulmonary disease, pulmonary fibrosis, pulmonary hypertension, pyoderma gangrenosum, pyogenic granuloma retrolental fibroplasias, pyogenic sterile arthritis, Raynaud’s syndrome, Reiter's disease, reactive arthritis, renal disease, renal graft rejection, reperfusion injury, respiratory distress syndrome, retinal disease, retrolental fibroplasia, Reynaud's syndrome, rheumatic carditis, rheumatic diseases, rheumatic fever, rheumatoid arthritis, rhinitis, rhinitis psoriasis, rosacea, sarcoidosis, Schnitzler syndrome, scleritis, sclerosis, scleroderma, scoliosis, seborrhea, sepsis, septic shock, severe pain, Sézary syndrome, sickle cell anemia, silica-induced disease (Silicosis), Sjogren's syndrome, skin diseases, skin irritation, skin rash, skin sensitization (contact dermatitis or allergic contact dermatitis), sleep apnea, spinal cord injury, spinal stenosis, spondyloarthropathies, sports injuries, sprains and strains, Stevens-Johnson syndrome (SJS), stroke, subarachnoid hemorrhage, sunburn, synovial inflammation, systemic inflammatory response syndrome (SIRS), systemic lupus erythematosus (SLE), systemic mast cell disease (SMCD), systemic vasculitis, systemic-onset juvenile idiopathic arthritis, temporal arteritis, tendinitis, tenosynovitis, thrombocytopenia, thyroditis, thyroiditis, tissue transplant, toxoplasmosis, trachoma, transplantation rejection, traumatic brain injury, tuberculosis, tubulointerstitial nephritis, tumor necrosis factor (TNF) receptor associated periodic syndrome (TRAPS), type 1 diabetes, type 2 diabetes, complications from type 1 or type 2 diabetes, ulcerative colitis, urticaria, uterine fibroids, uveitis, uveoretinitis, vascular restenosis, vasculitis, vasculitis (NHLBI), vitiligo, Wegener's granulomatosis, and Whipple's disease. [0436] In some embodiments, the inflammatory condition is selected from inflammatory bowel disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, glomerulonephritis, mixed connective tissue disease (MCTD), dermatomyositis, polymyositis, systemic sclerosis, antineutrophil cytoplasmic antibody-associated vasculitis, anti-phospholipid syndrome, autoimmune hemolytic anemia, macrophage activation syndrome driven inflammatory anemia, IgA nephropathy, type I diabetes, non-alcoholic steatohepatitis, and Sjogren’s syndrome. In some embodiments, the inflammatory condition is inflammatory bowel disease. In some embodiments, the inflammatory condition is psoriasis. In some embodiments, the inflammatory condition is psoriatic arthritis. In some embodiments, the inflammatory condition is rheumatoid arthritis. In some embodiments, the inflammatory condition is glomerulonephritis. In some embodiments, the inflammatory condition is mixed connective tissue disease (MCTD). In some embodiments, the inflammatory condition is dermatomyositis. In some embodiments, the inflammatory condition is polymyositis. In some embodiments, the inflammatory condition is systemic sclerosis. In some embodiments, the inflammatory condition is antineutrophil cytoplasmic antibody-associated vasculitis. In some embodiments, the inflammatory condition is anti-phospholipid syndrome. In some embodiments, the inflammatory condition is autoimmune hemolytic anemia. In some embodiments, the inflammatory condition is macrophage activation syndrome driven inflammatory anemia. In some embodiments, the inflammatory condition is IgA nephropathy. In some embodiments, the inflammatory condition is type I diabetes. In some embodiments, the inflammatory condition is non-alcoholic steatohepatitis. In some embodiments, the inflammatory condition is Sjogren’s syndrome. [0437] The compounds provided herein, or pharmaceutically acceptable salts thereof, or the pharmaceutical composition provided herein may treat or ameliorate systemic lupus erythematosus (SLE), cutaneous lupus erythematosus (CLE), lupus nephritis, lupus-related, symptom of SLE, symptom of CLE, or other autoimmune disorder. Symptoms of systemic lupus erythematosus include joint pain, joint swelling, arthritis, fatigue, hair loss, mouth sores, swollen lymph nodes, sensitivity to sunlight, skin rash, headaches, numbness, tingling, seizures, vision problems, personality changes, abdominal pain, nausea, vomiting, abnormal heart rhythms, coughing up blood and difficulty breathing, patchy skin color and Raynaud's phenomenon. [0438] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of treating systemic lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0439] In one embodiment, the present disclosure provides a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein, for use in a method of treating cutaneous lupus erythematosus in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof, or a therapeutically effective amount of a pharmaceutical composition provided herein. [0440] In some embodiments, the uses provided herein further comprise administering a therapeutically effective amount of one or more additional therapeutic agents, or a pharmaceutically acceptable salt thereof. [0441] In some embodiments, the one or more additional therapeutic agents is selected from the group consisting of veltuzumab, PF-06835375, eculizumab, milatuzumab, SM-06, SM- 03, BT-063, QX-006-N, BOS-161721, AK-101, TNX-1500, theralizumab, daxdilimab, TAK- 079, felzartamab, itolizumab, anifrolumab, iscalimab, dapirolizumab pegol, lanalumab, LY- 3361237, JNJ-55920839, UBP-1213, DS-7011, PFI-102, BIIB-059, obexelimab, talacotuzumab, vobarilizumab, TE-2324, PRV-3279, chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, AVO-101, rozibafusp alfa, VRN-02, annexuzlimab, ALPN- 101, bendamustine hydrochloride, BMS-986256, NKTR-35, atacicept, telitacicept, BMS- 986256, M-5049, KZR-616, KPG-818, verdinexor, ALPN-303, valziflocept, LA-1, cenerimod, prednisone, corticotropin, deucravacitinib, CPL-409116, CS-12192, tofacitinib citrate, ISB-830, DV-1079, julemic acid, iberdomide, TAM-01, BML-258, brepocitinib, SDC-1801, SDC-1802, ICP-330, NTR-441, dalazatide, GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, HMPL-523, RSLV-132, interleukin-2 follow-on biologic, interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, CUG-252, DZ-2002, PEGylated HLA-x (SLE), AC- 0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, orelabrutinib, DWP-213388, INV-103, R-salbutamol sulphate, anchorins, NIK-SMI1, X-6, INV- 17, Oshadi D, baricitinib, upadacitinib, filgotinib, itacitinib, INCB-54707, delgocitinib, DWP- 212525, CKD-971, as mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT-013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, and AMG-592, or a pharmaceutically acceptable salt of any of the foregoing, or any combination thereof. [0442] In some embodiments, the one or more additional therapeutic agents is selected from chloroquine and hydroxychloroquine, or a pharmaceutically acceptable salt thereof. In some embodiments, the one or more additional therapeutic agents is chloroquine. In some embodiments, the one or more additional therapeutic agents is hydroxychloroquine. In some embodiments, the one or more additional therapeutic agent is a pharmaceutically acceptable salt of hydroxychloroquine. In some embodiments, the one or more additional therapeutic agent is hydroxychloroquine sulfate. [0443] In some embodiments of the uses provided herein, the subject is a human. [0444] In some embodiments, the uses provided herein comprise administering a therapeutically effective amount of a compound provided herein (i.e., a compound of Formula I), or a pharmaceutically acceptable salt thereof. In some embodiments, the methods provided herein comprise administering a therapeutically effective amount of a pharmaceutical composition provided herein. V. Administration [0445] The compounds of the present disclosure or pharmaceutically acceptable salts thereof (also referred to herein as the active ingredients) can be administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), transdermal, vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with, for example, the condition of the recipient. An advantage of certain compounds disclosed herein, or pharmaceutically acceptable salts thereof, is that they are orally bioavailable and can be dosed orally. [0446] A compound of the present disclosure, or a pharmaceutically acceptable salt thereof, may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer. In some embodiments, the compound, or a pharmaceutically acceptable salt thereof, is administered on a daily or intermittent schedule for the duration of the individual’s life. [0447] The specific dose level of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease in the subject undergoing therapy. For example, a dosage may be expressed as a number of milligrams of a compound provided herein, or a pharmaceutically acceptable salt thereof, per kilogram of the subject’s body weight (mg/kg). Dosages of between about 0.1 and 150 mg/kg may be appropriate. In some embodiments, about 0.1 and 100 mg/kg may be appropriate. In other embodiments a dosage of between 0.5 and 60 mg/kg may be appropriate. Normalizing according to the subject’s body weight is particularly useful when adjusting dosages between subjects of widely disparate size, such as occurs when using the drug in both children and adult humans or when converting an effective dosage in a non-human subject such as dog to a dosage suitable for a human subject. [0448] The daily dosage may also be described as a total amount of a compound described herein, or a pharmaceutically acceptable salt thereof, administered per dose or per day. Daily dosage of a compound of Formula I, or a pharmaceutically acceptable salt or pharmaceutically acceptable tautomer thereof, may be between about 1 mg and 4,000 mg, between about 2,000 to 4,000 mg/day, between about 1 to 2,000 mg/day, between about 1 to 1,000 mg/day, between about 10 to 500 mg/day, between about 20 to 500 mg/day, between about 50 to 300 mg/day, between about 75 to 200 mg/day, or between about 15 to 150 mg/day. [0449] The dosage or dosing frequency of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, may be adjusted over the course of the treatment, based on the judgment of the administering physician. [0450] The compounds of the present disclosure, or pharmaceutically acceptable salts thereof, may be administered to an individual (e.g., a human) in a therapeutically effective amount. In some embodiments, the compound, or a pharmaceutically acceptable salt thereof, is administered once daily. [0451] The compounds provided herein, or pharmaceutically acceptable salts thereof, can be administered by any useful route and means, such as by oral or parenteral (e.g., intravenous) administration. Therapeutically effective amounts of the compound, or a pharmaceutically acceptable salt thereof, may include from about 0.00001 mg/kg body weight per day to about 10 mg/kg body weight per day, such as from about 0.0001 mg/kg body weight per day to about 10 mg/kg body weight per day, or such as from about 0.001 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.01 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.05 mg/kg body weight per day to about 0.5 mg/kg body weight per day. In some embodiments, a therapeutically effective amount of the compounds provided herein, or pharmaceutically acceptable salts thereof, include from about 0.3 mg to about 30 mg per day, or from about 30 mg to about 300 mg per day, or from about 0.3 ^g to about 30 mg per day, or from about 30 ^g to about 300 ^g per day. [0452] A compound of the present disclosure, or a pharmaceutically acceptable salt thereof, may be combined with one or more additional therapeutic agents in any dosage amount of the compound of the present disclosure or a pharmaceutically acceptable salt thereof (e.g., from 1 mg to 1000 mg of compound). Therapeutically effective amounts may include from about 0.1 mg per dose to about 1000 mg per dose, such as from about 50 mg per dose to about 500 mg per dose, or such as from about 100 mg per dose to about 400 mg per dose, or such as from about 150 mg per dose to about 350 mg per dose, or such as from about 200 mg per dose to about 300 mg per dose, or such as from about 0.01 mg per dose to about 1000 mg per dose, or such as from about 0.01 mg per dose to about 100 mg per dose, or such as from about 0.1 mg per dose to about 100 mg per dose, or such as from about 1 mg per dose to about 100 mg per dose, or such as from about 1 mg per dose to about 10 mg per dose, or such as from about 1 mg per dose to about 1000 mg per dose. Other therapeutically effective amounts of the compound of Formula I, or a pharmaceutically acceptable salt thereof, are about 1 mg per dose, or about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or about 100 mg per dose. Other therapeutically effective amounts of the compound of the present disclosure, or pharmaceutically acceptable salts thereof, are about 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, or about 1000 mg per dose. [0453] In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 600 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 500 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 400 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 300 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 200 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 100 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 75 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 50 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 25 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 20 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 15 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 10 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg to about 5 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, about 21 mg, about 22 mg, about 23 mg, about 24 mg, or about 25 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 5 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 10 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 15 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 20 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 25 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 30 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 35 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 40 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 45 mg. In some embodiments, a therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof, is about 50 mg. [0454] In some embodiments, the methods described herein comprise administering to the subject an initial daily dose of about 1 to 500 mg of a compound provided herein, or a pharmaceutically acceptable salt thereof, and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, once per week, once every two weeks, once every three weeks, or once a month. [0455] When administered orally, the total daily dosage for a human subject may be between about 1 mg and 1,000 mg, between about 10-500 mg/day, between about 50-300 mg/day, between about 75-200 mg/day, or between about 100-150 mg/day. In some embodiments, the total daily dosage for a human subject may be about 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 200, 300, 400, 500, 600, 700, or 800 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 300, 400, 500, or 600 mg/day administered in a single dose. [0456] In some embodiments, the total daily dosage for a human subject may be about 100 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 150 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 200 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 250 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 300 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 350 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 400 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 450 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 500 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 550 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 600 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 650 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 700 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 750 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 800 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 850 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 900 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 950 mg/day administered in a single dose. In some embodiments, the total daily dosage for a human subject may be about 1000 mg/day administered in a single dose. [0457] A single dose can be administered hourly, daily, weekly, or monthly. For example, a single dose can be administered once every 1 hour, 2, 3, 4, 6, 8, 12, 16 or once every 24 hours. A single dose can also be administered once every 1 day, 2, 3, 4, 5, 6, or once every 7 days. A single dose can also be administered once every 1 week, 2, 3, or once every 4 weeks. In certain embodiments, a single dose can be administered once every week. A single dose can also be administered once every month. In some embodiments, a compound provided herein, or a pharmaceutically acceptable salt thereof, is administered once daily in a method disclosed herein. In some embodiments, a compound provided herein, or a pharmaceutically acceptable salt thereof, is administered twice daily in a method disclosed herein. [0458] The frequency of dosage of the compound of the present disclosure, or a pharmaceutically acceptable salt thereof, will be determined by the needs of the individual patient and can be, for example, once per day or twice, or more times, per day. Administration of the compound, or a pharmaceutically acceptable salt thereof, continues for as long as necessary to treat the inflammatory condition, or any other indication described herein. For example, a compound, or a pharmaceutically acceptable salt thereof, can be administered to a human suffering from an inflammatory condition for a period of from 20 days to 180 days or, for example, for a period of from 20 days to 90 days or, for example, for a period of from 30 days to 60 days. [0459] Administration can be intermittent, with a period of several or more days during which a patient receives a daily dose of the compound of the present disclosure, or a pharmaceutically acceptable salt thereof, followed by a period of several or more days during which a patient does not receive a daily dose of the compound or a pharmaceutically acceptable salt thereof. For example, a patient can receive a dose of the compound, or a pharmaceutically acceptable salt thereof, every other day, or three times per week. Again by way of example, a patient can receive a dose of the compound, or a pharmaceutically acceptable salt thereof, each day for a period of from 1 to 14 days, followed by a period of 7 to 21 days during which the patient does not receive a dose of the compound, or a pharmaceutically acceptable salt thereof, followed by a subsequent period (e.g., from 1 to 14 days) during which the patient again receives a daily dose of the compound, or a pharmaceutically acceptable salt thereof. Alternating periods of administration of the compound, or a pharmaceutically acceptable salt thereof, followed by non-administration of the compound, or a pharmaceutically acceptable salt thereof, can be repeated as clinically required to treat the patient. [0460] The compounds of the present disclosure, or pharmaceutically acceptable salts thereof, or the pharmaceutical compositions of the present disclosure may be administered once, twice, three, or four times daily, using any suitable mode described above. Also, administration or treatment with the compounds, or pharmaceutically acceptable salts thereof, may be continued for a number of days; for example, commonly treatment would continue for at least 7 days, 14 days, or 28 days, for one cycle of treatment. Treatment cycles are well known for inflammatory conditions and other indications described herein. In some embodiments, treatment cycles are frequently alternated with resting periods of about 1 to 28 days, commonly about 7 days or about 14 days, between cycles. The treatment cycles, in other embodiments, may also be continuous. VI. Combination Therapy [0461] Patients being treated by administration of the compounds provided herein, or pharmaceutically acceptable salts thereof, often exhibit diseases or conditions that benefit from treatment with other therapeutic agents. These diseases or conditions can be of an inflammatory nature or can be related to cancer, metabolic disorders, gastrointestinal disorders and the like. Thus, one embodiment of the disclosure is a method of treating an inflammation related disease or condition, or a metabolic disorder, gastrointestinal disorder, or cancer and the like comprising administering a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, in combination with one or more compounds useful for the treatment of such diseases to a subject, particularly a human subject, in need thereof. [0462] In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is combined with one, two, three, four or more additional therapeutic agents. In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is combined with two additional therapeutic agents. In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is combined with three additional therapeutic agents. In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is combined with four additional therapeutic agents. The one, two, three, four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, and/or they can be selected from different classes of therapeutic agents. [0463] In some embodiments, when a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is combined with one or more additional therapeutic agents as described herein, the components of the composition are administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. [0464] In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is combined with one or more additional therapeutic agents in a unitary dosage form for simultaneous administration to a patient, for example as a solid dosage form for oral administration. [0465] In some embodiments, a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is co-administered with one or more additional therapeutic agents. [0466] Co-administration includes administration of unit dosages of the compounds provided herein, or pharmaceutically acceptable salts thereof, before or after administration of unit dosages of one or more additional therapeutic agents. The compounds provided herein, or pharmaceutically acceptable salts thereof, may be administered within seconds, minutes, or hours of the administration of one or more additional therapeutic agents. For example, in some embodiments, a unit dose of a compound provided herein, or a pharmaceutically acceptable salt thereof, is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents. Alternatively, in other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound provided herein, or a pharmaceutically acceptable salt thereof, within seconds or minutes. In some embodiments, a unit dose of a compound provided herein, or a pharmaceutically acceptable salt thereof, is administered first, followed, after a period of hours (i.e., 1-12 hours), by administration of a unit dose of one or more additional therapeutic agents. In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (i.e., 1-12 hours), by administration of a unit dose of a compound provided herein or a pharmaceutically acceptable salt thereof. [0467] In some embodiments, a compound of Formula I, or a pharmaceutically acceptable salt thereof, is formulated as a tablet, which may optionally contain one or more other compounds useful for treating the disease being treated. In certain embodiments, the tablet can contain another active ingredient for treating an inflammatory condition or other indication described herein. In some embodiments, such tablets are suitable for once daily dosing. [0468] Also provided herein are methods of treatment in which a compound of Formula I, or a tautomer or pharmaceutically acceptable salt thereof, is given to a patient in combination with one or more additional therapeutic agents or therapy. In some embodiments, the total daily dosage of a compound of Formula I, or a tautomer, or a pharmaceutically acceptable salt thereof, may be about 1 to about 500 mg/day administered in a single dose for a human subject. Inflammatory Condition or Disease Combination Therapy [0469] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one or more additional therapeutic agents that treat or ameliorate an inflammatory condition. Non-limiting examples of an inflammatory condition include, without limitation, acne, acid-induced lung injury, Addison's disease, adrenal hyperplasia, adrenocortical insufficiency, adult-onset Still's disease, adult respiratory distress syndrome (ARDS), age-related macular degeneration, aging, alcoholic hepatitis, alcoholic liver disease, allergen-induced asthma, allergic bronchopulmonary, allergic conjunctivitis, allergic contact dermatitis, allergies, allergic encephalomyelitis, allergic neuritis, allograft rejection, alopecia, alopecia areata, Alzheimer's disease, amyloidosis, amyotrophic lateral sclerosis, angina pectoris, angioedema, angiofibroma, anhidrotic ectodermal dysplasia-ill, anti-glomerular basement membrane disease, antigen-antibody complex mediated diseases, ankylosing spondylitis, antiphospholipid syndrome, aphthous stomatitis, appendicitis, arthritis, ascites, aspergillosis, asthma, atherosclerosis, atherosclerotic plaques, atopic dermatitis, atrophic thyroiditis, autoimmune diseases, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune polyendocrinopathies, autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), autoimmune hepatitis, autoimmune thyroid disorders, autoinflammatory diseases, back pain, Bacillus anthracis infection, Bechet's disease, bee sting-induced inflammation, Behçet’s syndrome, Bell’s palsy, berylliosis, Blau syndrome, bone pain, bronchiolitis, bullous pemphigoid (BP) asthma, burns, bursitis, cardiac hypertrophy, carpal tunnel syndrome, Castleman's disease, catabolic disorders, cataracts, Celiac disease, cerebral aneurysm, chemical irritant-induced inflammation, chorioretinitis, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome, chronic heart failure, chronic lung disease of prematurity, chronic obstructive pulmonary disease (COPD), chronic pancreatitis, chronic prostatitis, chronic recurrent multifocal osteomyelitis, cicatricial alopecia, colitis, complex regional pain syndrome, complications of organ transplantation, conjunctivitis, connective tissue disease, contact dermatitis, corneal graft neovascularization, corneal ulcer, Crohn's disease, cryopyrin-associated periodic syndromes, cutaneous lupus erythematosus (CLE), cryptococcosis, cystic fibrosis, deficiency of the interleukin-1 receptor antagonist (DIRA), dermatitis, dermatitis endotoxemia, dermatomyositis, diabetic macular edema, diverticulitis, eczema, encephalitis, endometriosis, endotoxemia, eosinophilic pneumonias, epicondylitis, epidermolysis bullosa, erythema multiforme, erythroblastopenia, esophagitis, familial amyloidotic polyneuropathy, familial cold urticarial, familial Mediterranean fever, fetal growth retardation, fibromyalgia, fistulizing Crohn’s disease, food allergies, giant cell arteritis, glaucoma, glioblastoma, glomerular disease, glomerular nephritis, glomerulonephritis, gluten- sensitive enteropathy, gout, gouty arthritis, graft-versus-host disease (GVHD), granulomatous hepatitis, Graves' disease, growth plate injuries, Guillain-Barre syndrome. gut diseases, hair loss, Hashimoto's thyroiditis, head injury, headache, hearing loss, heart disease, hemangioma, hemolytic anemia, hemophilic joints, Henoch-Scholein purpura, hepatitis, hereditary periodic fever syndrome, heritable disorders of connective tissue, herpes zoster and simplex, hidradenitis suppurativa (HS), hip replacement, Hodgkin's disease, Huntington's disease, hyaline membrane disease, hyperactive inflammatory response, hyperammonemia, hypercalcemia, hypercholesterolemia, hypereosinophilic syndrome (HES), hyperimmunoglobulinemia D with recurrent fever (HIDS), hypersensitivity pneumonitis, hypertropic bone formation, hypoplastic and other anemias, hypoplastic anemia, ichthyosis, idiopathic demyelinating polyneuropathy, Idiopathic inflammatory myopathies (dermatomyositis, polymyositis), idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura, immunoglobulin nephropathies, immune complex nephritis, immune thrombocytopenic purpura (ITP), incontinentia pigmenti (IP, Bloch– Siemens syndrome), infectious mononucleosis, infectious diseases including viral diseases such as AIDS (HIV infection), hepatitis A, B, C, D, and E, herpes; inflammation, inflammation of the CNS, inflammatory bowel disease (IBD), inflammatory disease of the lower respiratory tract including bronchitis or chronic obstructive pulmonary diseases, inflammatory disease of the upper respiratory tract including the nose and sinuses such as rhinitis or sinusitis, inflammatory diseases of the respiratory tract, inflammatory ischemic event such as stroke or cardiac arrest, inflammatory lung disease, inflammatory myopathy such as myocarditis, inflammatory liver disease, inflammatory neuropathy, inflammatory pain, insect bite-induced inflammation, interstitial cystitis, interstitial lung disease, iritis, irritant-induced inflammation, ischemia/reperfusion, joint replacement, juvenile arthritis, juvenile rheumatoid arthritis, keratitis, kidney injury caused by parasitic infections, kidney transplant rejection, leptospirosis, leukocyte adhesion deficiency, lichen sclerosus (LS), Lambert-Eaton myasthenic syndrome, Loeffler's syndrome, lupus, lupus nephritis, Lyme disease, Marfan syndrome (MFS), mast cell activation syndrome, mastocytosis, meningitis, meningioma, mesothelioma, mixed connective tissue disease, Muckle-Wells syndrome (urticaria deafness amyloidosis), mucositis, multiple organ injury syndrome, multiple sclerosis, muscle wasting, muscular dystrophy, myasthenia gravis (MG), myelodysplastic syndrome, myocarditis, myositis, nasal sinusitis, necrotizing enterocolitis, neonatal onset multisystem inflammatory disease (NOMID), neovascular glaucoma, nephrotic syndrome, neuritis, neuropathological diseases, non-allergen induced asthma, obesity, ocular allergy, optic neuritis, organ transplant rejection, Osier-Weber syndrome, osteoarthritis, osteogenesis imperfecta, osteonecrosis, osteoporosis, osterarthritis, otitis, pachyonychia congenita, Paget’s disease, Paget’s disease of bone, pancreatitis, Parkinson's disease, pediatric rheumatology, pelvic inflammatory disease, pemphigus, pemphigus vulgaris (PV), bullous pemphigoid (BP), pericarditis, periodic fever, periodontitis, peritoneal endometriosis, pernicious anemia (Addison's disease), pertussis, PFAPA (periodic fever aphthous pharyngitis and cervical adenopathy), pharyngitis and adenitis (PFAPA syndrome), plant irritant-induced inflammation, pneumocystis infection, pneumonia, pneumonitis, poison ivy/ urushiol oil-induced inflammation, polyarthritis nodosa, polychondritis, polycystic kidney disease, polymyalgia rheumatic, giant cell arteritis, polymyositis, pouchitis, reperfusion injury and transplant rejection, primary biliary cirrhosis, primary pulmonary hypertension, primary sclerosing cholangitis (PSC), proctitis, psoriasis, psoriasis vulgaris, psoriatic arthritis, psoriatic epidermis, psychosocial stress diseases, pulmonary disease, pulmonary fibrosis, pulmonary hypertension, pyoderma gangrenosum, pyogenic granuloma retrolental fibroplasias, pyogenic sterile arthritis, Raynaud’s syndrome, Reiter's disease, reactive arthritis, renal disease, renal graft rejection, reperfusion injury, respiratory distress syndrome, retinal disease, retrolental fibroplasia, Reynaud's syndrome, rheumatic carditis, rheumatic diseases, rheumatic fever, rheumatoid arthritis, rhinitis, rhinitis psoriasis, rosacea, sarcoidosis, Schnitzler syndrome, scleritis, sclerosis, scleroderma, scoliosis, seborrhea, sepsis, septic shock, severe pain, Sézary syndrome, sickle cell anemia, silica-induced disease (Silicosis), Sjogren's syndrome, skin diseases, skin irritation, skin rash, skin sensitization (contact dermatitis or allergic contact dermatitis), sleep apnea, spinal cord injury, spinal stenosis, spondyloarthropathies, sports injuries, sprains and strains, Stevens-Johnson syndrome (SJS), stroke, subarachnoid hemorrhage, sunburn, synovial inflammation, systemic inflammatory response syndrome (SIRS), systemic lupus erythematosus (SLE), systemic mast cell disease (SMCD), systemic vasculitis, systemic-onset juvenile idiopathic arthritis, temporal arteritis, tendinitis, tenosynovitis, thrombocytopenia, thyroditis, thyroiditis, tissue transplant, toxoplasmosis, trachoma, transplantation rejection, traumatic brain injury, tuberculosis, tubulointerstitial nephritis, tumor necrosis factor (TNF) receptor associated periodic syndrome (TRAPS), type 1 diabetes, type 2 diabetes, complications from type 1 or type 2 diabetes, ulcerative colitis, urticaria, uterine fibroids, uveitis, uveoretinitis, vascular restenosis, vasculitis, vasculitis (NHLBI), vitiligo, Wegener's granulomatosis, and Whipple's disease. [0470] Non-limiting examples of therapeutic agents for treatment of an inflammatory disease or condition that can be used in combination with the compounds provided herein, or pharmaceutically acceptable salts thereof, include alpha-fetoprotein modulators; adenosine A3 receptor antagonist; adrenomedullin ligands; AKT1 gene inhibitors; antibiotics; antifungals; ASK1 inhibitors; ATPase inhibitors; beta adrenoceptor antagonists; BTK inhibitors; calcineurin inhibitors; carbohydrate metabolism modulators; cathepsin S inhibitors; CCR9 chemokine antagonists; CD233 modulators; CD29 modulators; CD3 antagonists; CD40 ligand inhibitors; CD40 ligand receptor antagonists; chemokine CXC ligand inhibitors; CHST15 gene inhibitors; collagen modulators; COT protein kinase inhibitors; CSF-1 agonist; CSF-1 antagonists; CX3CR1 chemokine modulators DYRK-1 alpha protein kinase inhibitor, eotaxin ligand inhibitors; EP4 prostanoid receptor agonists; F1F0 ATP synthase modulators; farnesoid X receptor (FXR, NR1H4) agonists or modulators; fecal microbiota transplantation (FMT), fractalkine ligand inhibitors; free fatty acid receptor 2 antagonists; GATA 3 transcription factor inhibitors; glucagon-like peptide 2 agonists; glucocorticoid agonists; Glucocorticoid receptor modulators; guanylate cyclase receptor agonists; HIF prolyl hydroxylase inhibitors; histone deacetylase inhibitors; HLA class II antigen modulators; hypoxia inducible factor-1 stimulator; ICAM1 gene inhibitors; IL-1 beta ligand modulators; IL-12 antagonists; IL-13 antagonists; IL- 18 antagonists; IL-18 receptor accessory protein antagonist, IL-22 agonists; IL-23 antagonists; IL-23A inhibitors; IL-6 antagonists; IL-7 receptor antagonists; IL-8 receptor antagonists; IL-36 inhibitors, integrin alpha-4/beta-1 antagonists; integrin alpha-4/beta-7 antagonists; integrin antagonists; interleukin ligand inhibitors; interleukin receptor 17A antagonists; interleukin-1 beta ligands; interleukin 1 like receptor 2 inhibitors; IL-6 receptor modulators; JAK tyrosine kinase inhibitors; Jak1 tyrosine kinase inhibitors; Jak3 tyrosine kinase inhibitors; lactoferrin stimulators; LanC like protein 2 modulators; leukocyte elastate inhibitors; leukocyte proteinase- 3 inhibitors; MAdCAM inhibitors; melanin concentrating hormone (MCH-1) antagonist; melanocortin agonists; metalloprotease-9 inhibitors; microbiome-targeting therapeutics; natriuretic peptide receptor C agonists; neuregulin-4 ligands; NLRP3 inhibitors; NKG2 D activating NK receptor antagonists; NR1H4 receptor (FXR) agonists or modulators(deleted); nuclear factor kappa B inhibitors; opioid receptor antagonists; OX40 ligand inhibitors; oxidoreductase inhibitors; P2X7 purinoceptor modulators; PDE 4 inhibitors; Pellino homolog 1 inhibitors; PPAR alpha/delta agonists; PPAR gamma agonists; Protein arginine deiminase IV inhibitor, protein fimH inhibitors; P-selectin glycoprotein ligand-1 inhibitors; Ret tyrosine kinase receptor inhibitors; RIP-1 kinase inhibitors; RIP-2 kinase inhibitors; RNA polymerase inhibitors; sphingosine 1 phosphate phosphatase 1 stimulators; sphingosine-1-phosphate receptor-1 agonists; sphingosine-1-phosphate receptor-5 agonists; sphingosine-1-phosphate receptor-1 antagonists; sphingosine-1-phosphate receptor-1 modulators; stem cell antigen-1 inhibitors; superoxide dismutase modulators; SYK inhibitors; tissue transglutaminase inhibitor; TLR-3 antagonists; TLR-4 antagonists; Toll- like receptor 8 (TLR8) inhibitors; TNF alpha ligand inhibitors; TNF ligand inhibitors; TNF alpha ligand modulators; TNF antagonists; TPL-2 inhibitors; tumor necrosis factor 14 ligand modulators; tumor necrosis factor 15 ligand inhibitors; Tyk2 tyrosine kinase inhibitors; type I IL-1 receptor antagonists; vanilloid VR1 agonists; and zonulin inhibitors; or any combination thereof. [0471] Adenosine A3 receptor antagonists include but are not limited to PBF-677. [0472] Adrenomedullin ligands include but are not limited to adrenomedullin. [0473] Antibiotics include but are not limited to ciprofloxacin, clarithromycin, metronidazole, vancomycin, rifamycin, rifaximin, and tosufloxacin. [0474] ASK1 inhibitors include but are not limited to GS-4997. [0475] Alpha-fetoprotein modulators include but are not limited to ACT-101. [0476] Anti-CD28 inhibitors include but are not limited to JNJ-3133 and abatacept. [0477] Beta adrenoceptor antagonists include but are not limited to NM-001. [0478] BTK inhibitors include but are not limited to GS-4059. [0479] Calcineurin inhibitors include but are not limited to tacrolimus and ciclosporin. [0480] Carbohydrate metabolism modulators include but are not limited to ASD-003. [0481] Cathepsin S inhibitors include but are not limited to VBY-129. [0482] CCR9 chemokine antagonists include but are not limited to CCX-507. [0483] CD233 modulators include but are not limited to GSK-2831781. [0484] CD29 modulators include but are not limited to PF-06687234. [0485] CD3 antagonists include but are not limited to NI-0401, muromonab-CD3, and teplizumab. [0486] CD4 antagonists include but are not limited to IT-1208. [0487] CD40 ligand inhibitors include but are not limited to SAR-441344 and letolizumab. [0488] CD40 gene inhibitors include but are not limited to NJA-730. [0489] CD40 ligand receptor antagonists include but are not limited to FFP-104, BI- 655064, ABBV-323, and VIB-4920. [0490] Chaperonin binding immunoglobulin protein include but are not limited to IRL- 201805. [0491] Chemokine CXC ligand inhibitors include but are not limited to LY-3041658. [0492] CHST15 gene inhibitors include but are not limited to STNM-01. [0493] Collagen modulators include but are not limited to ECCS-50 (DCCT-10). [0494] COT protein kinase inhibitors include but are not limited to GS-4875. [0495] CSF-1 antagonists include but are not limited to JNJ-40346527 (PRV-6527) and SNDX-6352. [0496] CX3CR1 chemokine modulators include but are not limited to E-6130. [0497] DYRK-1 alpha protein kinase inhibitor include but are not limited to VRN-02 [0498] Microbiome-targeting therapeutics include but are not limited to SER-287, SER- 301, and SER-155. [0499] Eotaxin ligand inhibitors include but are not limited to bertilimumab. [0500] EP4 prostanoid receptor agonists include but are not limited to KAG-308. [0501] F1F0 ATP synthase modulators include but are not limited to LYC-30937 EC. [0502] Fractalkine ligand inhibitors include but are not limited to quetmolimab (E-6011). [0503] Free fatty acid receptor 2 antagonists include but are not limited to GLPG-0974. [0504] GATA 3 transcription factor inhibitors include but are not limited to SB-012. [0505] Glucagon-like peptide 2 agonists include but are not limited to teduglutide and apraglutide. [0506] Glucocorticoid receptor agonists include but are not limited to budesonide, beclomethasone dipropionate, and dexamethasone sodium phosphate. [0507] Glucocorticoid receptor modulators /TNF ligand inhibitors include but are not limited to ABBV-3373. [0508] Guanylate cyclase receptor agonists include but are not limited to dolcanatide. [0509] HIF prolyl hydroxylase inhibitors include but are not limited to DS-1093 and AKB-4924. [0510] HIF prolyl hydroxylase-2 inhibitors /hypoxia inducible factor-1 stimulators include but are not limited to GB-004. [0511] Histone deacetylase inhibitors include but are not limited to givinostat and NIPEP-CARE. [0512] Histone deacetylase-6 inhibitors include but are not limited to CKD-506. [0513] HLA class II antigen modulators include but are not limited to HLA class II protein modulators. [0514] ICAM1 gene inhibitors include but are not limited to alicaforsen. [0515] IL-12 antagonists include but are not limited to ustekinumab (IL12/IL23). [0516] IL-13 antagonists include but are not limited to tralokinumab. [0517] IL-18 antagonists include but are not limited to GSK-1070806. [0518] IL-18 receptor accessory protein antagonist include but are not limited to anti-IL- 1R7 canonical antibody. [0519] IL-22 agonists include but are not limited to AMT-126 and RG-7880. [0520] IL-23 antagonists include but are not limited to tildrakizumab, risankizumab (BI- 655066), mirikizumab (LY-3074828), brazikumab (AMG-139), IBI-112, and PTG-200. [0521] IL-23A inhibitors include but are not limited to guselkumab. [0522] IL-6 antagonists include but are not limited to olokizumab. [0523] IL-7 receptor antagonists include but are not limited to OSE-127. [0524] IL-8 receptor antagonists include but are not limited to clotrimazole. [0525] Integrin alpha-4/beta-1 antagonists include but are not limited to natalizumab. [0526] Integrin alpha-4/beta-7 antagonists include but are not limited to etrolizumab (a4b7/aEb7), vedolizumab, carotegrast methyl, TRK-170 (a4b7/a4b1), PTG-100, and PN-10943. [0527] Integrin antagonists include but are not limited to E-6007. [0528] Interleukin ligand inhibitors include but are not limited to bimekizumab (IL- 17A/IL-17F). [0529] Interleukin receptor 17A antagonists include but are not limited to brodalumab. [0530] Interleukin-1 beta ligands include but are not limited to K(D)PT. [0531] Interleukin 1 like receptor 2 inhibitors include but are not limited to BI-655130. [0532] IL-6 receptor modulators include but are not limited to Amilo-5MER and olamkicept. [0533] JAK tyrosine kinase inhibitors include but are not limited to tofacitinib (1/3), peficitinib (1/3), TD-3504, and TD-1473. [0534] Jak1 tyrosine kinase inhibitors include but are not limited to a compound disclosed in US Patent No.9238628. [0535] Jak3 tyrosine kinase inhibitors include but are not limited to OST-122 and PF- 06651600. [0536] Jak3 tyrosine kinase inhibitor/ TrkA receptor antagonist include but are not limited to SNA-125. [0537] Examples of other JAK inhibitors include but are not limited to AT9283, AZD1480, baricitinib, BMS-911543, fedratinib, filgotinib (GLPG0634), gandotinib (LY2784544), INCB039110, lestaurtinib, momelotinib (CYT0387), NS-018, pacritinib (SB1518), peficitinib (ASP015K), ruxolitinib, tofacitinib (formerly tasocitinib), XL019, upadacitinib (ABT-494), LPG-0555, SHR-0302, and brepocitinib (PF-06700841) (JAK1/Tyk2). [0538] Lactoferrin stimulators include but are not limited to recombinant human lactoferrin (VEN-100). [0539] LanC like protein 2 modulators include but are not limited to BT-11 and BT-104. [0540] Leukocyte elastase inhibitors/Leukocyte proteinase-3 inhibitors include but are not limited to tiprelestat. [0541] MAdCAM inhibitors include but are not limited to SHP-647 (PF-547659). [0542] Melanin concentrating hormone (MCH-1) antagonists include but are not limited to CSTI-100. [0543] Melanocortin MC1 receptor agonists include but are not limited to ASP-3291 and PL-8177. [0544] Metalloprotease-9 inhibitors include but are not limited to GS-5745. [0545] Microbiome modulator include but are not limited to ABI-M201 [0546] Natriuretic peptide receptor C agonists include but are not limited to plecanatide. [0547] Neuregulin-4 ligands include but are not limited to NRG-4. [0548] NKG2 D activating NK receptor antagonists include but are not limited to JNJ- 4500. [0549] NLRP3 inhibitors include but are not limited to dapansutrile, BMS-986299, SB- 414, MCC-950, IFM-514, JT-194, PELA-167, and NBC-6. [0550] Farnesoid X receptor (FXR, NR1H4) agonists or modulators include but are not limited to AGN-242266, cilofexor tromethamine (GS-9674), EDP-305, EYP-001, GNF-5120, MET-409, MET-642, nidufexor (LMB-763), obeticholic acid, TERN-101, and tropifexor. [0551] Nuclear factor kappa B inhibitors include but are not limited to Thetanix. [0552] Opioid receptor antagonists include but are not limited to naltrexone and IRT- 103. [0553] OX40 ligand inhibitors include but are not limited to KHK-4083. [0554] Oxidoreductase inhibitors include but are not limited to olsalazine. [0555] Pellino homolog 1 inhibitors include but are not limited to BBT-401. [0556] P2X7 purinoceptor modulators include but are not limited to SGM-1019. [0557] PDE 4 inhibitors include but are not limited to apremilast. [0558] PPAR alpha/delta agonists include but are not limited to elafibranor (GFT-1007). [0559] PPAR gamma agonists include but are not limited to GED-0507-34-Levo. [0560] Protein fimH inhibitors include but are not limited to sibofimloc (EB-8018). [0561] P-selectin glycoprotein ligand-1 inhibitors include but are not limited to SEL-K2, AbGn-168H, and neihulizumab. [0562] Ret tyrosine kinase receptor inhibitors include but are not limited to GSK- 3179106. [0563] RIP-1 kinase inhibitors include but are not limited to GSK-2982772 and VRN- 04. [0564] RIP-2 kinase inhibitors include but are not limited to GSK-2983559. [0565] Sphingosine 1 phosphate phosphatase 1 stimulators include but are not limited to etrasimod. [0566] Sphingosine-1-phosphate receptor-1 agonists include but are not limited to mocravimod (KRP-203) and BMS-986166. [0567] Sphingosine-1-phosphate receptor-1 agonists/Sphingosine-1-phosphate receptor- 5 agonists include but are not limited to ozanimod. [0568] Sphingosine-1-phosphate receptor-1 antagonists include but are not limited to amiselimod (MT-1303). [0569] Sphingosine-1-phosphate receptor-1 modulators include but are not limited to OPL-002, SK1-I. [0570] Stem cell antigen-1 inhibitors include but are not limited to Ampion (DMI-9523). [0571] Superoxide dismutase modulators include but are not limited to midismase. [0572] Syk inhibitors include but are not limited to GS-9876. [0573] tissue transglutaminase inhibitor include but are not limited to zampilimab [0574] TLR-3 antagonists include but are not limited to PRV-300. [0575] TLR-4 antagonists include but are not limited to JKB-122. [0576] Toll- like receptor 8 (TLR8) inhibitors include but are not limited to E-6887, IMO-4200, IMO-8400, IMO-9200, MCT-465, MEDI-9197, motolimod, resiquimod, VTX-1463, and VTX-763. [0577] TNF alpha ligand inhibitors include but are not limited to adalimumab, certolizumab pegol, infliximab, golimumab, DLX-105, Debio-0512, HMPL-004, CYT-020- TNFQb, Hemay-007, and V-565. [0578] TNF alpha ligand modulators/ IL-1 beta ligand modulators include but are not limited to PUR-0110. [0579] TNF antagonists include but are not limited to AVX-470, tulinercept, and etanercept. [0580] Tumor necrosis factor 14 ligand modulators include but are not limited to AEVI- 002. [0581] Tumor necrosis factor 15 ligand inhibitors include but are not limited to PF- 06480605. [0582] Tyk2 tyrosine kinase inhibitors include but are not limited to PF-06826647 and BMS-986165. [0583] Type I IL-1 receptor antagonists include but are not limited to anakinra. [0584] Zonulin inhibitors include but are not limited to larazotide acetate. [0585] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one or more anti-inflammatory agents. Anti- inflammatory agents include but are not limited to non-steroidal anti-inflammatory drugs (NSAIDs), non-specific and COX-2 specific cyclooxgenase enzyme inhibitors, gold compounds, corticosteroids, methotrexate, tumor necrosis factor receptor (TNF) receptors antagonists, and immunosuppressants. [0586] Examples of NSAIDs include, but are not limited to ibuprofen, flurbiprofen, naproxen and naproxen sodium, diclofenac, combinations of diclofenac sodium and misoprostol, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, sodium nabumetone, sulfasalazine, tolmetin sodium, and hydroxychloroquine. Additional examples of NSAIDs also include but are not limited to COX-2 specific inhibitors (i.e., a compound that inhibits COX-2 with an IC 50 that is at least 50-fold lower than the IC 50 for COX-1), such as celecoxib, valdecoxib, lumiracoxib, etoricoxib and/or rofecoxib. [0587] In some embodiments, the anti-inflammatory agent is a salicylate. Salicylates include but are not limited to acetylsalicylic acid or aspirin, sodium salicylate, choline, and magnesium salicylates. [0588] In some embodiments, the anti-inflammatory agent is a corticosteroid. Non- limiting examples of a corticosteroid include cortisone, dexamethasone, methylprednisolone, prednisolone, prednisolone sodium phosphate, and prednisone. [0589] In some embodiments, the anti-inflammatory agent is a gold compound, e.g., gold sodium thiomalate or auranofin. [0590] In some embodiments, the anti-inflammatory agent is a metabolic inhibitor. Non- limiting examples of a metabolic inhibitor include a dihydrofolate reductase inhibitor, such as methotrexate, or a dihydroorotate dehydrogenase (DHODH) inhibitor, such as leflunomide. [0591] In some embodiments, the anti-inflammatory agent is an anti-C5 monoclonal antibody (such as eculizumab or pexelizumab), a TNF antagonist (such as entanercept), or infliximab, which is an anti-TNF alpha monoclonal antibody. [0592] In some embodiments, the anti-inflammatory agent is an immunosuppressant. Non-limiting examples of an immunosuppressant include methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine, mycophenolate sodium, mercaptopurine, and mycophenolate mofetil. Lupus Combination Therapy [0593] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, is combined with one or more additional therapeutic agents that target adenosylhomocysteinase, ADP ribosyl cyclase-1 (CD38), adrenocorticotrophic hormone ligands, AIMP multisynthetase complex protein 1, annexin A1 modulators, B and T lymphocyte attenuator (BTLA), BDCA2, beta 2 adrenoceptor, B-lymphocyte antigen CD19, B-lymphocyte antigen CD20, B-lymphocyte cell adhesion molecule (CD22), B-lymphocyte stimulator ligand (BAFF), btk tyrosine kinase, cannabinoid CB2 receptor, CD11b agonists, CD38 Activation- inducible TNF receptor, CD40 (CD154) ligand, CD74, CD79b modulators, CDw123, Collagen VII (Col VII), Complement C5 factor, C-type lectin domain protein 4C, CXCR5 chemokine modulators, deoxyribonuclease modulators, DNA binding protein Ikaros, DYRK-1 alpha protein kinase, dndoplasmin, Exportin 1, FK506 binding protein, glucocorticoid receptor, HLA antigen, IL-10, IL-23m IL-12 receptors, IL-2 receptor, IL-2 receptor alpha subunit, IL-21 modulators, IL- 6R, immunoglobulin gamma Fc receptor II modulators, immunoglobulin gamma Fc receptor IIB, inducible T-cell co-stimulator, interferon alpha ligand (INF-alpha), interferon omega ligand (INF omega), interferon type I receptor, interleukin-2 ligand, Itk tyrosine kinase, JAK tyrosine kinase, Jak1 tyrosine kinase, Jak2 tyrosine kinase, Jak3 tyrosine kinase, KCNA voltage-gated potassium channel-3, leukocyte Ig like receptor A4 modulators, mitochondrial 10 kDa heat shock protein, mTOR, non receptor tyrosine kinase TYK2, nuclear export, nuclear factor kappa B inducing kinase, nuclease stimulators, OX-40 receptors, PARP modulators, proteasome modulators, protein arginine deiminase IV (PAD4), protein cereblon modulators, protein MB21D1, retinoid Z receptor gamma inverse, rho associated protein kinase 1, rho associated protein kinase 2, serine threonine protein kinase TBK1 (TBK1), sphingosine kinase 1, sphingosine-1-phosphate receptor-1 modulators, stimulator of interferon genes protein, Syk tyrosine kinase, T cell surface glycoprotein CD28, T-cell differentiation antigen CD6, TLR-7 modulators, TLR-8 modulators, TLR-9 modulators, transcription factor modulators, tumor necrosis factor ligand 13 (APRIL), Tyk2 tyrosine kinase, ubiquitin ligase modulators, and/or zinc finger binding protein Aiolos. [0594] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from: ● activation-inducible TNF receptor agonists, including but not limited to BMS-986256; ● adenosylhomocysteinase inhibitors, including but not limited to DZ-2002; ● adrenocorticotrophic hormone ligands, including but not limited to corticotropin; ● AIMP multisynthetase complex protein 1 stimulator/Endoplasmin inhibitors, including but not limited to anchorins; ● aniti-CDw123 antibodies, including but not limited to talacotuzumab; ● annexin A1 modulators, including but not limited to annexuzlimab; ● anti- IL-12/IL23 antibodies, including but not limited to AK-101; ● anti-BAFF-R antibodies, including but not limited to lanalumab; ● anti-BDCA2 antibodies, including but not limited to BIIB-059; ● anti-BLys antibodies, including but not limited to belimumab and UBP-1213; ● anti-BTLA modulator antibodies, including but not limited to LY-3361237; ● anti-C5 antibodies, including but not limited to eculizumab; ● anti-CD154 antibodies, including but not limited to TNX-1500; ● anti-CD19/CD32b antibodies, including but not limited to obexelimab; ● anti-CD20 antibodies, including but not limited to veltuzumab; ● anti-CD22 antibodies, including but not limited to SM-06, SM-03; ● anti-CD28 antibodies, including but not limited to theralizumab; ● anti-CD38 antibodies, including but not limited to TAK-079 and felzartamab; ● anti-CD40 antibodies, including but not limited to iscalimab and dapirolizumab pegol; ● anti-CD6 antibodies, including but not limited to itolizumab; ● anti-CD74 antibodies, including but not limited to milatuzumab; ● anti-CXCR5 antibodies, including but not limited to PF-06835375; ● anti-IFN-alpha antibodies, including but not limited to QX-006-N; ● anti-IFN-alpha/omega antibodies, including but not limited to JNJ-55920839; ● anti-IL-10 antibodies, including but not limited to BT-063; ● anti-IL-21 antibodies, including but not limited to BOS-161721; ● anti-IL-6R nanobodies, including but not limited to vobarilizumab; ● anti-ILT7 antibodies, including but not limited to daxdilimab; ● anti-interferon alpha vaccines, including but not limited to CKD-971; ● anti-interferon receptor type I antibodies, including but not limited to anifrolumab; ● anti-PAD4 antibodies, including but not limited to PFI-102; ● anti-TLR-7 antibodies, including but not limited to DS-7011; ● BAFF/APRIL inhibitors, including but not limited to ALPN-303; ● Beta 2 adrenoceptor agonists, including but not limited to R-salbutamol sulphate; ● bi-specific antibodies targeting BAFF/ICOSL, including but not limited to rozibafusp alfa; ● bi-specific antibodies targeting CD32B/CD79B, including but not limited to PRV-3279; ● bi-specific antibodies targeting Col VII/BAFF, including but not limited to TE-2324; ● B-lymphocyte stimulator ligand inhibitors, including but not limited to atacicept and telitacicept; ● Btk tyrosine kinase inhibitors, including but not limited to AC-0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, and orelabrutinib; ● Btk/itk tyrosine kinase inhibitors, including but not limited to DWP-213388; ● Btk/Jak3 tyrosine kinase inhibitors, including but not limited to DWP-212525; ● cannabinoid CB2 receptor agonists, including but not limited to julemic acid; ● CD11b agonists, including but not limited to LA-1; ● deoxyribonuclease gamma stimulators, including but not limited to NTR-441; ● deoxyribonuclease modulators, including but not limited to Oshadi D; ● DYRK-1 alpha protein kinase inhibitors, including but not limited to VRN-02; ● exportin 1 inhibitors, including but not limited to SINE compounds; ● glucocorticoid receptor agonists, including but not limited to prednisone; ● HLA antigen modulators, including but not limited to PEGylated HLA-x (SLE); ● IL-2 receptor alpha subunit stimulators, including but not limited to NKTR-35; ● imunoglobulin gamma Fc receptor IIB modulators, including but not limited to valziflocept; ● inducible T-cell co-stimulator inhibitor (ICOS)/T cell surface glycoprotein CD28 inhibitors, including but not limited to ALPN-101; ● interferon alpha ligand modulator/ TLR-7 /TLR-9 modulators, including but not limited to DV-1079; ● interleukin-2 ligands, including but not limited to interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, and CUG-252; ● interleukin-2 ligands/ IL-2 receptor agonists, including but not limited to interleukin-2 follow-on biologic; ● JAK 1/2/3 and ROCK 1/2 inhibitors, including but not limited to CPL-409116; ● JAK tyrosine kinase inhibitors, including but not limited to delgocitinib; ● Jak1 /Jak2 tyrosine kinase inhibitors, including but not limited to baricitinib; ● Jak1 tyrosine kinase inhibitors, including but not limited to upadacitinib, filgotinib, itacitinib, and INCB-54707; ● Jak1/Tyk2 tyrosine kinase inhibitors, including but not limited to brepocitinib, SDC- 1801, and SDC-1802; ● JAK3/1 and TBK1 kinase inhibitors, including but not limited to CS-12192; ● JAK3/JAK1 tyrosine kinase inhibitors, including but not limited to tofacitinib citrate; ● KCNA voltage-gated potassium channel-3 inhibitors, including but not limited to dalazatide; ● mitochondrial 10 kDa heat shock protein stimulators, including but not limited to INV- 103; ● mTOR inhibitors, including but not limited to TAM-01; ● non-receptor tyrosine kinase TYK2 antagonists, including but not limited to ICP-330; ● nuclear export inhibitors, including but not limited to verdinexor; ● nuclear factor kappa B inducing kinase inhibitors, including but not limited to NIK- SMI1; ● nuclease stimulators, including but not limited to RSLV-132; ● OX-40 receptor antagonists, including but not limited to ISB-830; ● PARP modulators, including but not limited to bendamustine hydrochloride; ● PD-L1 CAR-expressing NK-92 cell therapy; ● proteasome inhibitors, including but not limited to KZR-616; ● protein cereblon modulators, including but not limited to iberdomide; ● protein MB21D1 inhibitors, including but not limited to X-6; ● retinoid Z receptor gamma inverse agonists, including but not limited to INV-17; ● sphingosine kinase 1 inhibitors, including but not limited to BML-258; ● sphingosine-1-phosphate receptor-1 modulator, including but not limited to cenerimod; ● Syk tyrosine kinase inhibitors, including but not limited to GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, and HMPL-523; ● TLR-9 antagonists, including but not limited to chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, and AVO-101; ● TLR7/8 antagonists, including but not limited to M-5049, E-6887, and BMS-986256; ● TLR-8 antagonists, including but not limited to ZG-170607; ● TLR7/8/9 antagonists, including but not limited to IMO-8400 and IMO-9200; ● Tyk2 tyrosine kinase inhibitors, including but not limited to deucravacitinib; ● ubiquitin ligase modulators, including but not limited to KPG-818; and ● other drugs for lupus, including but not limited to mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT-013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, AMG-592, phosphatidylserine-liposome- based immunotherapy, and CD4+CD127lo/-CD25+ polyclonal regulatory T cells. [0595] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from veltuzumab, PF-06835375, eculizumab, milatuzumab, SM-06, SM-03, BT- 063, QX-006-N, BOS-161721, AK-101, TNX-1500, theralizumab, daxdilimab, TAK-079, felzartamab, itolizumab, anifrolumab, iscalimab, dapirolizumab pegol, lanalumab, LY-3361237, JNJ-55920839, UBP-1213, DS-7011, PFI-102, BIIB-059, obexelimab, talacotuzumab, vobarilizumab, TE-2324, PRV-3279, chloroquine, hydroxychloroquine, hydroxychloroquine sulfate, COV-08-0064; GNKS-356, AVO-101, rozibafusp alfa, VRN-02, annexuzlimab, ALPN- 101, bendamustine hydrochloride, BMS-986256, NKTR-35, atacicept, telitacicept, BMS- 986256, M-5049, KZR-616, KPG-818, verdinexor, ALPN-303, valziflocept, LA-1, cenerimod, prednisone, corticotropin, deucravacitinib, CPL-409116, CS-12192, tofacitinib citrate, ISB-830, DV-1079, julemic acid, iberdomide, TAM-01, BML-258, brepocitinib, SDC-1801, SDC-1802, ICP-330, NTR-441, dalazatide, GSK-2646264, SKI-O-703, lanraplenib (GS-9876), GNS-1653, HMPL-523, RSLV-132, interleukin-2 follow-on biologic, interleukin-2 Anteluke, interking recombinant human interleukin-2, ILT-101, CUG-252, DZ-2002, PEGylated HLA-x (SLE), AC- 0058, fenebrutinib, XNW-1011, tirabrutinib hydrochloride, branebrutinib, elsubrutinib, orelabrutinib, DWP-213388, INV-103, R-salbutamol sulphate, anchorins, NIK-SMI1, X-6, INV- 17, Oshadi D, baricitinib, upadacitinib, filgotinib, itacitinib, INCB-54707, delgocitinib, DWP- 212525, CKD-971, as mometasone, betamethasone, forigerimod, anandamide, DCB-SLE1, arsenic trioxide, tairuimide, TV-4710 (edratide), allogeneic human umbilical cord-derived mesenchymal stem cell therapy (hUC-MSCs), LC-200, BI-705564, SM-934, GX-101, TXR-712, TXR-711, CIT-013, MHV-370, Panzyga®, TPX-6001, TPX-7001, artenimol, and AMG-592, or a pharmaceutically acceptable salt of any of the foregoing, or any combination thereof. Psoriasis Combination Therapy [0596] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents that are useful for treating or ameliorating psoriasis. In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from acetaldehyde dehydrogenase inhibitor, adenosine A1 receptor antagonist, adenosine A3 receptor antagonist, adenosine A3 receptor agonists, ADP ribosyl cyclase-1 inhibitors, alpha 2 adrenoceptor modulator, apolipoprotein A antagonist, aryl hydrocarbon receptor agonist, Bcl-xL Bcl-2 associated death promotor modulators, beta amyloid antagonist, beta-catenin inhibitors, bromodomain containing protein inhibitor, Ca2+ release activated Ca2+ channel 1 inhibitors, calcineurin inhibitors, calcium channel inhibitors, cannabinoid CB1 receptor antagonist, cathepsin S inhibitors, CCR3 chemokine antagonists, CXCR2 chemokine antagonist, CXCR1/2 chemokine, CCR6 chemokine antagonist, CD223 modulators, CD40 ligand receptor antagonists, cell adhesion molecule inhibitors, cell surface glycoprotein MUC18 inhibitors, CREB binding protein inhibitors, CXCR4 chemokine modulators, cytokine receptor antagonist, cytosolic phospholipase A2 inhibitors, DHFR inhibitors, DYRK-1 alpha protein kinase inhibitor, EGFR family tyrosine kinase receptor inhibitors, enolase 1 inhibitor, eotaxin ligand inhibitors, F1F0 ATP synthase modulator, free fatty acid receptor 2 agonist, free fatty acid receptor 3 agonist, galectin-3 inhibitors, glucocorticoid agonists, GM-CSF ligand inhibitors, GNRH receptor modulators, 5-HT 1a receptor antagonist, FGF receptor antagonist, GroEL protein 2 inhibitor, histamine H1 receptor antagonists, histamine H4 receptor antagonists, histone deacetylase-1 inhibitors, histone deacetylase-2 inhibitors, histone deacetylase-3 inhibitors, histone deacetylase-6 inhibitors, Hsp 90 inhibitor, IL-1 receptor antagonist, interleukin 1 like receptor 2 inhibitor, IL-2 receptor alpha subunit stimulator, IL-2 modulator, IL-10 antagonists, IL-12 antagonists, IL-17 agonist, IL17RA gene inhibitor, IL-17 antagonists, IL-23 antagonists, IL-8 antagonists, immunoglobulin like domain receptor 2 antagonist, insulin receptor substrate-1 inhibitors, interferon gamma receptor antagonists, interleukin 17 ligand inhibitors, interleukin 17A ligand inhibitors, interleukin 17A ligand modulators, interleukin 17F ligand inhibitors, interleukin 23A inhibitors, interleukin receptor 17A antagonists, interleukin receptor 17A modulators, interleukin-1 alpha ligand inhibitors, interleukin-1 beta ligand modulators, IRAK-4 protein kinase inhibitor, Itk tyrosine kinase inhibitor, JAK tyrosine kinase inhibitors, Jak1 tyrosine kinase inhibitors, Jak2 tyrosine kinase inhibitors, Jak3 tyrosine kinase inhibitors, KCNA voltage-gated potassium channel-3 inhibitors, Lck tyrosine kinase inhibitors, lysophosphatidate-1 receptor antagonists, MALT protein 1 inhibitors, MAP kinase inhibitors, membrane copper amine oxidase inhibitors, metalloprotease-1 inhibitors, mitochondrial 10 kDa heat shock protein stimulators, mTOR complex 1 inhibitor, mTOR complex 2 inhibitor, non receptor tyrosine kinase TYK2 antagonists, nuclear erythroid 2-related factor 2 stimulators, nuclear factor kappa B inhibitors, nucleoside reverse transcriptase inhibitors, oncostatin M receptor subunit beta inhibitor, opioid receptor delta antagonists, OX40 ligand inhibitor, parathyroid hormone ligand inhibitors, PDE 4 inhibitors, PDE 4b inhibitor, P2Y6 purinoceptor modulator; P-glycoprotein inhibitors, phosphoinositide-3 kinase delta inhibitors, phosphoinositide-3 kinase gamma inhibitors, phospholipase A2 inhibitors, programmed cell death ligand 1 modulators, programmed cell death protein 1 stimulator, P- selectin glycoprotein ligand-1 stimulators, retinoic acid receptor agonists, retinoic acid receptor gamma antagonists, retinoic acid receptor gamma inverse agonists, retinoid receptor agonists, retinoid X receptor agonists, retinoid X receptor modulators, retinoid Z receptor gamma agonists, retinoid Z receptor gamma inverse agonists, retinoid Z receptor gamma antagonist, rho associated protein kinase 2 inhibitors, ribonuclease P inhibitors, RIP-1 kinase inhibitor, sphingosine-1-phosphate receptor-1 antagonists, sphingosine-1-phosphate receptor-1 modulators, Src tyrosine kinase inhibitors, STAT-3 inhibitors, Syk tyrosine kinase inhibitor, T-box transcription factor TBX21 modulators, T-cell differentiation antigen CD6 inhibitors, T- cell surface glycoprotein CD8 inhibitors, T cell surface glycoprotein CD28 stimulator, TGF beta agonists, TLR-7 antagonists, TLR-8 antagonists, TLR-9 antagonists, TNF alpha ligand inhibitors, TNF alpha ligand modulators, TNF antagonists, TNF binding agents, TNF gene inhibitor, topoisomerase II inhibitors, TrkA receptor antagonists, tubulin binding agents, Tyk2 tyrosine kinase inhibitor, type II TNF receptor modulators, unspecified cytokine receptor antagonists, vitamin D3 receptor agonists, vitamin D3 receptor modulators, Wnt ligand inhibitor, and Wnt 5A ligand inhibitor. [0597] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from AP-005, 18C3 (anti-IL-1 alpha true human antibody), ABX-464, acitretin, adalimumab, adipocell, AFB-035, aganirsen, AKP-11, alefacept, alitretinoin, Amilo-5mer, aminopterin, amiselimod, apremilast, ASKP-1240, AST-005, ATI-2138, AVX-001, baricitinib, belapectin (GR-MD-02), bertilimumab, betamethasone, BI-655066, BI-730357, BI-730460, BI- 730460, bimekizumab, BMS-986165, BMX-010, briakinumab, brodalumab, BTT-1023, C-82, calcipotriol, calcitriol, CC-90005, CCL-20LD, CD-10367, certolizumab pegol, CF-101, ciclosporin, CJM-112, CKBA, clobetasol propionate + tretinoin, CM-2489, CPL-409116, crisaborole, CS-12192, CT-327, CTX-101, dalazatide, DFD-06, dimethyl fumarate, dithranol, DLX-105, DSXS-1411, DSXS-1535, DUR-928, EDP-1815, etanercept, fluocinonide, FPP-003, GK-664-S, GLG-801, GLPG-3121, GLPG-3667, GLPG-3970, GLY-2028, GMDP, GSK- 2800528, GSK-2831781, GSK-2981278A, guselkumab, halomethasone, HAT-1, IMO-3100, IMO-8400, inecalcitol, infliximab, INV-103, IR-444, IR-502, itolizumab, ixekizumab, JN-2528, KBL-697, KD-025, LAS-41004, LEO-124249, LEO-29102, LEO-32731, LEO-35299, lithium succinate, LNP-1955, LP-0200, M-1095, maxacalcitol, MDX-018, methotrexate, MOL-4249, mometasone, MP-1032, MSB-03, myristyl nicotinate, namilumab, neihulizumab, niclosamide, NLP-91, NP-000888, NVN-1000, olopatadine, orilotimod, P-3072, P-3073, PAT-1657, Pc4, pefcalcitol, PF-06700841, Prurisol, PRX-003, PRX-167700, PUR-0110, recombinant human LFA-3/antibody fusion protein , RON-2315, RTU-1096, S-414114, secukinumab, SHP-141, SMET-D1, SNK-01, SP-14019, SSS-07, tacalcitol, tazarotene, tildrakizumab, tirbanibulin (KX- 01), tofacitinib, toreforant, tregalizumab, TU-2100, UCB-5857, UHE-105, ulobetasol, ustekinumab, VBY-891, voclosporin, VTP-43742, WBI-1001, and ZPL-389, or a pharmaceutically acceptable salt of any of the foregoing, or any combination thereof. [0598] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from: ● acetaldehyde dehydrogenase inhibitor, including but not limited to ADX-629; ● adenosine A3 receptor agonists, including but not limited to piclidenoson (CF-101); ● adenosine A3 receptor antagonist, including but not limited to PBF-1650; ● ADP ribosyl cyclase-1 inhibitors, including but not limited to IMO-3100; ● 5-HT 1a receptor antagonist, including but not limited to AX-1602; ● apolipoprotein A antagonist, including but not limited to orticumab; ● cytokine receptor antagonist, including but not limited to tapinarof; ● aryl hydrocarbon receptor modulator, including but not limited to NTI-528 and RLV- 102; ● Bcl-xL Bcl-2 associated death promotor modulators, including but not limited to Pc4; ● beta-catenin inhibitors, including but not limited to C-82; ● bromodomain containing protein inhibitor, including but not limited to BOS-475; ● Ca2+ release activated Ca2+ channel 1 inhibitors, including but not limited to CM-2489 and PRCL-02; ● calcineurin inhibitors, including but not limited to voclosporin, pimecrolimus, tacrolimus, ciclosporin, HS-378, oxeclosporin, OLO-400, ADV-P3, and CTX-006; ● calcium channel inhibitors, including but not limited to RP-3128; ● cathepsin S inhibitors, including but not limited to VBY-129, VBY-891, RWJ-445380, and CRA-028129; ● CCR3 chemokine antagonists, including but not limited to bertilimumab; ● CXCR2 chemokine antagonist, including but not limited to CCX-624; ● CD223 modulators, including but not limited to GSK-2831781; ● CD40 ligand receptor antagonists, including but not limited to ASKP-1240, lucatumumab, and toralizumab; ● cell adhesion molecule inhibitors, including but not limited to BIRT-2584, PC-114, alicaforsen, IC-747, ICM-3, and ISIS-2302; ● cell surface glycoprotein MUC18 inhibitors, including but not limited to PRX-003 and imaprelimab; ● CREB binding protein inhibitors, including but not limited to C-82; ● CXCR1/2 chemokine, including but not limited to LY-3041658; ● CXCR4 chemokine modulators, including but not limited to CD184-FK506 ADC; ● cytosolic phospholipase A2 inhibitors, including but not limited to AVX-001; ● DHFR inhibitors, including but not limited to methotrexate, CH-4051, CePep, CH-1504, MQX-5902, and MPI-2505; ● DYRK-1 alpha protein kinase inhibitor, including but not limited to VRN-02; ● EGFR family tyrosine kinase receptor inhibitors, including but not limited to erlotinib, icotinib hydrochloride, and SGT-210; ● Enolase 1 inhibitor, including but not limited to HuL-001; ● Eotaxin ligand inhibitors, including but not limited to bertilimumab; ● F1F0 ATP synthase modulator, including but not limited to LYC-30937; ● FGF receptor antagonist, including but not limited to potassium dobesilate; ● free fatty acid receptor 2, 3 agonist, including but not limited to SFA-002; ● galectin-3 inhibitors, including but not limited to belapectin (GR-MD-02); ● glucocorticoid agonists, including but not limited to betamethasone, clobetasol, auranofin, NM-135, DSXS-1538b, and SEGRA; ● GM-CSF ligand inhibitors, including but not limited to namilumab; ● GNRH receptor modulators, including but not limited to NL-001; ● GroEL protein 2 inhibitor, including but not limited to prozumab; ● histamine H1 receptor antagonists, including but not limited to olopatadine and loratadine + nortriptyline; ● histamine H4 receptor antagonists, including but not limited to toreforant and ZPL-389; ● histone deacetylase-2 inhibitors, including but not limited to KAR-1880; ● histone deacetylase 1, 6, 2, 3 inhibitors, including but not limited to remetinostat (SHP- 141); ● Hsp 90 inhibitor, including but not limited to CTXT-102; ● IL-2 receptor alpha subunit stimulator, including but not limited to NKTR-358; ● IL-2 modulator; including but not limited to CC-92252; ● IL-10 antagonists, including but not limited to pimecrolimus; ● IL-12 antagonists, including but not limited to BOW-090, briakinumab, FM-202, and apilimod; ● IL-17 antagonists, including but not limited to ixekizumab, secukinumab, AFB-035, KD- 025, DLX-3003, EBI-028, M-1095, IMO-3100, GR-1501, 608, vunakizumab, sonelokimab, AK-111, HB-0017, and SIM-335; ● IL-17 agonist, including but not limited to ZL-1102; ● IL17RA gene inhibitor, including but not limited to XCUR-17; ● IL-23 antagonists, including but not limited to tildrakizumab, BI-655066, AMG-139, briakinumab, mirikizumab (LY-3074828),FM-202, apilimod, LY-2525623, risankizumab, and IBI-112; ● IL-23 antagonist, including but not limited to ustekinumab and AK-101; ● IL-8 antagonists, including but not limited to BMS-986253 (MDX-018), AS-101, ABX- IL8, LI-312, SB-332235, and LF-216; ● immunoglobulin like domain receptor 2 antagonist, including but not limited to CGEN- 15001; ● insulin receptor substrate-1 inhibitors, including but not limited to aganirsen; ● interferon gamma receptor antagonists, including but not limited to pimecrolimus, AMG- 811, OA-1, AGT-1, mometasone + nortriptyline, and fontolizumab; ● interleukin 17 ligand inhibitors, including but not limited to CJM-112, netakimab, and AFB-035; ● interleukin 17A ligand inhibitors, including but not limited to COVA-322, JS-005, and ABY-035/AFO2; ● interleukin 17A ligand modulators, including but not limited to QX-002-N; ● interleukin 17A/17F ligand inhibitors, including but not limited to bimekizumab; ● interleukin 23A inhibitors, including but not limited to guselkumab and QX-004-N; ● interleukin receptor 17A antagonists, including but not limited to brodalumab and LZM- 012; ● interleukin 1 like receptor 2 inhibitor, including but not limited to spesolimab and imsidolimab; ● interleukin-1 alpha ligand inhibitors, including but not limited to bermekimab (CA- 18C3); ● interleukin-1 beta ligand modulators, including but not limited to PUR-0110 and AR- 100; ● IRAK-4 protein kinase inhibitor, including but not limited to BAY-1834845; ● Itk tyrosine kinase inhibitor, including but not limited to JTE-051; ● JAK tyrosine kinase inhibitors, including but not limited to CS-17380; ● Jak1 tyrosine kinase inhibitors, including but not limited to itacitinib, abrocitinib (PF- 04965842), solcitinib, SHR-0302, and filgotinib; ● JAK1,2,3 tyrosine kinase inhibitor, including but not limited to jaktinib; ● JAk1,2 tyrosine kinase inhibitor, including but not limited to baricitinib and ruxolitinib; ● TYk2 tyrosine kinase inhibitor, including but not limited to brepocitinib; ● Jak1 tyrosine kinase inhibitor, including but not limited to PF-06263276; ● JAk 1, 3 tyrosine kinase inhibitor, including but not limited to CS-944X, tofacitinib, and peficitinib; ● KCNA voltage-gated potassium channel-3 inhibitors, including but not limited to KPI- 150, dalazatide, BNC-164, and SPS-4251; ● Lck tyrosine kinase inhibitors, including but not limited to BMS-350751 and NTRC- 0625-0; ● lysophosphatidate-1 receptor antagonists, including but not limited to BMS-986202; ● MAP kinase inhibitors, including but not limited to AIK-33 and KIN-3032; ● membrane copper amine oxidase inhibitors, including but not limited to vepalimomab, BTT-1023, RTU-1096, and PRX-167700; ● metalloprotease-1 inhibitors, including but not limited to KIN-3032 and HMR-1571; ● mitochondrial 10 kDa heat shock protein stimulators, including but not limited to INV- 103; ● Non receptor tyrosine kinase TYK2 antagonists, including but not limited to SAR-20347, ICP-332, and SDC-1801; ● nuclear erythroid 2-related factor 2 stimulators, including but not limited to dimethyl fumarate and XP-23829; ● nuclear factor kappa B inhibitors, including but not limited to S-414114, VGX-1027, AKBA, SP-100030, and YP-008; ● nucleoside reverse transcriptase inhibitors, including but not limited to Prurisol; ● oncostatin M receptor subunit beta inhibitor, including but not limited to vixarelimab; ● Opioid receptor delta antagonists, including but not limited to HS-378; ● OX40 ligand inhibitor, including but not limited to KY-1005; ● P38 MAP kinase inhibitor, including but not limited to AMG-101, AIK-3, VGX-1027, AIK-a1, BMS-582949, doramapimod, semapimod, TA-5493, HEP-689, and RWJ- 68354; ● parathyroid hormone ligand inhibitors, including but not limited to inecalcitol; ● PDE 4 inhibitors, including but not limited to apremilast, roflumilast, orismilast, MK- 0873, Ro-20-1724, HMR-1571, RPR-122818, HPP-737, crisaborole, and DC-591042; ● PDE 4b inhibitor, including but not limited to GRT-6015; ● TNF alpha ligand inhibitor, including but not limited to Hemay-005; ● P-Glycoprotein inhibitors, including but not limited to boningmycin; ● Beta amyloid antagonist, including but not limited to GC-021109; ● phosphoinositide-3 kinase delta inhibitors, including but not limited to seletalisib (UCB- 5857); ● mTOR complex 2 inhibitor, including but not limited to bimiralisib; ● phosphoinositide-3 kinase gamma inhibitors, including but not limited to TAT-N25 peptide; ● phospholipase A2 inhibitor, including but not limited to ZPL-521, Project P-0229, BMS- 181162, and BMS-188184; ● programmed cell death ligand 1 modulators, including but not limited to GX-P2; ● programmed cell death protein 1 stimulator, including but not limited to LY-3462817 and CC-90006; ● P-selectin glycoprotein ligand-1 stimulators, including but not limited toneihulizumab; ● P-selectin glycoprotein ligand-1, including but not limited to AbGn-168H; ● retinoic acid receptor agonists, including but not limited to acitretin, tazarotene, tretinoin, tazarotene arotinoid trometamol, CD-1599, AM-580, BMS-181163, and CPR-2005; ● retinoic acid receptor gamma antagonists, including but not limited to VTP-43742 and BBI-6000; ● retinoic acid receptor gamma inverse agonists, including but not limited to GSK- 2981278A and JNJ-3534; ● retinoid receptor agonists, including but not limited to RASP; ● retinoid X receptor agonists, including but not limited to LGD-1550; ● retinoid X receptor modulators, including but not limited to bexarotene, alitretinoin, ALRT-1069, LGD-1069, and Net-4IB; ● retinoid Z receptor gamma agonists, including but not limited to NCE-407; ● retinoid Z receptor gamma inverse agonists, including but not limited to ARN-6039, IMU-935, BOS-172767, SAR-441169, and INV-17; ● retinoid Z receptor gamma antagonist, including but not limited to AUR-101, JTE-451, ESR-114, ABBV-157, and AZD-0284; ● rho associated protein kinase 2 inhibitors, including but not limited to KD-025; ● RIP-1 kinase inhibitor, including but not limited to GSK-2982772, DNL-758, and VRN- 04; ● ribonuclease P inhibitors, including but not limited to RASP; ● sphingosine-1-phosphate receptor-1 modulators, including but not limited to amiselimod, AKP-11, FP-253, and CS-0777; ● sphingosine-1-phosphate receptor-1 agonist, including but not limited to AK-119, SCD- 044, and SYL-927; ● sphingosine-1-phosphate receptor-5 modulator, including but not limited to CBP-307; ● Src tyrosine kinase inhibitors, including but not limited to tirbanibulin (KX-01); ● STAT-3 inhibitors, including but not limited to TAK-114, GLG-801, and MOL-4249; ● Syk tyrosine kinase inhibitor, including but not limited to HMPL-523; ● T-box transcription factor TBX21 modulators, including but not limited to SB-020; ● T-cell differentiation antigen CD6 inhibitors, including but not limited to itolizumab; ● T-cell surface glycoprotein CD8 inhibitors, including but not limited to tregalizumab; ● T cell surface glycoprotein CD28 stimulator, including but not limited to theralizumab; ● TGF beta agonists, including but not limited to tregalizumab; ● TLR-7 antagonists, including but not limited to IMO-3100; ● TLR-9 antagonists, including but not limited to IMO-3100 and GNKS-356; ● TLR 7,8,9 antagonist, including but not limited to IMO-8400; ● TNF alpha ligand inhibitors, including but not limited to adalimumab, CHS-1420, BAX- 2923, MSB-11022, ABP-501, MYL-1401A, infliximab, certolizumab pegol, AST-005, etanercept, opinercept, , ISIS-104838, DLX-105, SSS-07, DLX-2751, DLX-105 , Debio- 0512, TAQ-588, adalimumab , placulumab, PMI-001, CYT-020-TNFQb, AN-0128, CYT-007-TNFQb, SYI-2074, , YP-008, SCT-640A, SBT-104, and T-1649; ● TNF alpha ligand modulators, including but not limited to PUR-0110, CDP-571, and ACU-D2; ● TNF antagonists, including but not limited to certolizumab pegol, SCB-808, BAX-2200, CT-P05, SCB-131, GSK-2800528, onercept, and ALS-00T2-0501; ● TNF binding agents, including but not limited to adalimumab, certolizumab pegol SCB- 131, onercept, CT-P17, SBC-808, ABP-501, MYL-1401A, MSB-11022, BAX-2923, CHS-1420, and BCD-057; ● TNF gene inhibitor, including but not limited to AST-005; ● topoisomerase II inhibitors, including but not limited to GPX-150; ● TrkA receptor antagonists, including but not limited to VM-902A, CT-327, K-252a, and lestaurtinib; ● tubulin binding agents, including but not limited to KX-01 and paclitaxel; ● Tyk2 tyrosine kinase inhibitor, including but not limited to deucravacitinib, PF- 06826647, ABBV-712, and CS-43001; ● type II TNF receptor modulators, including but not limited to TNR-001, BAX-2200, and SCB-131; ● unspecified cytokine receptor antagonists, including but not limited to tetrathiomolybdate, JD-4000, X-083-NAB, SPHD-400, pimecrolimus, and HMPL-010; ● vitamin D3 receptor agonists, including but not limited to inecalcitol, maxacalcitol, calcipotriol , falecalcitriol, maxacalcitol , calcitriol NS-78, tacalcitol, calcipotriol, calcithiazol, ecalcidene, lexacalcitol, atocalcitol, and Ro-65-2299; ● vitamin D, D3 receptor modulators, including but not limited to VS-320 and VS-105; ● Wnt ligand inhibitor, including but not limited to SM-04755; and ● Wnt 5A ligand inhibitor, including but not limited to Box-5. Rheumatoid Arthritis Combination Therapy [0599] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents that are useful for treating or ameliorating rheumatoid arthritis. In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from 14-3-3 protein eta inhibitors, 5-lipoxygenase inhibitors, abl tyrosine kinase inhibitors, ACTH receptor agonists, adenosine A3 receptor agonists, adenosine deaminase inhibitors, ADP ribosyl cyclase-1 inhibitors, ADP ribosyl cyclase-1 modulators, ADP ribosylation factor 6 inhibitors, adrenocorticotrophic hormone ligands, aggrecanase-2 inhibitors, albumin modulators, anti- TNF steroid conjugate, adenosine A1 receptor antagonist, annexin A1 modulator, AP1 transcription factor inhibitors, apolipoprotein B modulator, aryl hydrocarbon receptor agonist plus autoantigen, basigin inhibitors, bcr protein inhibitors, B-lymphocyte antigen CD19 inhibitors, B-lymphocyte antigen CD20 inhibitors, B-lymphocyte antigen CD20 modulators, B-lymphocyte cell adhesion molecule inhibitor, B-lymphocyte stimulator ligand inhibitors, bradykinin receptor modulators, BRAF gene inhibitors, branched amino acid aminotransferase 1 inhibitors, bromodomain containing protein inhibitors, Btk tyrosine kinase inhibitors, cadherin-11 antagonists, calcineurin inhibitors, calcium channel inhibitors, calreticulin inhibitor, carbonic anhydrase inhibitors, cathepsin K inhibitors, cathepsin S inhibitors, CCR1 chemokine antagonists, CCR2 chemokine antagonists, CCR3 gene modulators, CCR5 chemokine antagonists, CD126 antagonists, CD29 modulators, CD3 modulators, CD39 agonists, CD4 agonists, CD4 antagonists, CD40 ligand inhibitors, CD40 ligand receptor antagonists, CD40 ligand receptor modulators, CD52 antagonists, CD73 agonists, CD79b modulators, CD80 antagonists, CD86 antagonists, CD95 antagonists, cell adhesion molecule inhibitors, chaperonin modulator, choline kinase inhibitors, clusterin stimulators, complement C5 factor inhibitors, complement factor stimulators, C-reactive protein inhibitors, CSF-1 antagonists, CXC10 chemokine ligand inhibitors, CXCR4 chemokine antagonists, cyclin- dependent kinase inhibitor 1 inhibitors, cyclin-dependent kinase-2 inhibitors, cyclin-dependent kinase-4 inhibitors, cyclin-dependent kinase-5 inhibitors, cyclin-dependent kinase-6 inhibitors, cyclin-dependent kinase-7 inhibitors, cyclin-dependent kinase-9 inhibitors, cyclooxygenase 2 inhibitors, cyclooxygenase 2 modulators, cyclooxygenase inhibitors, cytosolic phospholipase A2 inhibitors, cytotoxic T-lymphocyte protein-4 modulators, cytotoxic T-lymphocyte protein-4 stimulators, deoxyribonuclease gamma stimulator, DHFR inhibitors, diamine acetyltransferase inhibitors, dihydroorotate dehydrogenase inhibitors, DYRK-1 alpha protein kinase inhibitor, elongation factor 2 inhibitors, enolase 1 inhibitor, eotaxin 2 ligand inhibitors, EP4 prostanoid receptor antagonists, erythropoietin receptor agonists, factor XIIa antagonist, Fas ligands, FGF-2 ligand inhibitors, FK506 binding protein-12 modulators, folate antagonists, folate receptor agonists, folate receptor beta antagonists, folate receptor modulators, fractalkine ligand inhibitors, fyn tyrosine kinase inhibitors, G protein coupled receptor 15 antagonists, GABA A receptor modulators, glucocorticoid agonists, glucocorticoid antagonists, glucocorticoid induced leucine zipper stimulators, GM-CSF ligand inhibitors, GM-CSF receptor antagonists, GM-CSF receptor modulators, growth regulated protein alpha ligand inhibitors, H+ K+ ATPase inhibitors, histamine H4 receptor antagonists, histone deacetylase inhibitors, histone deacetylase-6 inhibitors, HIV-1 gp120 protein inhibitors, HLA class II antigen DQ-2 alpha modulators, HLA class II antigen inhibitors, HLA class II antigen modulators, Hsp 70 family inhibitors, hypoxia inducible factor-1 inhibitors, IFNB gene stimulators, I-kappa B kinase beta inhibitors, I-kappa B kinase inhibitors, IL-1 antagonists, IL-10 agonists, IL-11 agonists, IL-12 antagonists, IL-15 antagonists, IL-17 antagonists, IL-17 receptor modulators, IL-18 receptor accessory protein antagonist, IL-8 ligand inhibitors, IL-2 agonists, IL-2 antagonists, IL-21 antagonists, IL-23 antagonists, IL-3 antagonists, IL-4 agonists, IL-6 antagonists, IL-6 receptor modulators, IL-6 neutralizing human antibodies, anti- IL6 antibody, immunoglobulin antagonists, immunoglobulin G1 agonists, immunoglobulin G1 antagonists, immunoglobulin G1 modulators, immunoglobulin G2 antagonists, immunoglobulin G2 modulators, immunoglobulin gamma Fc receptor II modulators, immunoglobulin gamma Fc receptor IIB antagonists, immunoglobulin kappa modulators, immunoglobulin M antagonists, inducible nitric oxide synthase inhibitors (iNOS inhibitors), inosine monophosphate dehydrogenase inhibitors, insulin sensitizers, integrin alpha-1/beta-1 antagonists, integrin alpha-4/beta-1 antagonists, integrin alpha-9 antagonist, integrin antagonists, interferon beta ligands, interferon gamma ligands, interleukin 17A ligand inhibitors, interleukin 17F ligand inhibitors, interleukin 23A inhibitors, interleukin ligands, interleukin receptor 17A antagonists, interleukin-1 beta ligand inhibitors, interleukin-10 ligands, interleukin-2 ligands, interleukin-4 ligands, Interleukin-6 ligand inhibitors, Itk tyrosine kinase inhibitors, JAK tyrosine kinase inhibitors, Jak1 tyrosine kinase inhibitors, Jak2 tyrosine kinase inhibitors, JAK3 gene inhibitors, Jak3 tyrosine kinase inhibitors, Jun N terminal kinase inhibitors, KCNA voltage-gated potassium channel-3 modulators, kelch like ECH associated protein 1 modulators, kit tyrosine kinase inhibitors, LanC like protein 2 modulators, leukotriene BLT receptor antagonist, LITAF gene inhibitors, lymphocyte function antigen-3 receptor antagonists, Lyn tyrosine kinase inhibitors, macrophage-drug conjugate (MDC), macrophage mannose receptor 1 modulators, MAdCAM inhibitors, MAP kinase modulators, MAP3K2 gene inhibitors, MAPKAPK5 inhibitors, matrix metalloprotease inhibitors, MCL1 gene inhibitors, MEK protein kinase inhibitors, MEK-1 protein kinase inhibitors, MEK-2 protein kinase inhibitors, membrane copper amine oxidase inhibitors, metalloprotease-2 inhibitors, metalloprotease-9 inhibitors, methylprednisolone, midkine ligand inhibitors, mitochondrial 10 kDa heat shock protein stimulators, mTOR complex 1 inhibitors, mTOR inhibitors, NAD ADP ribosyltransferase stimulators, NAMPT gene inhibitors, NF kappa B inhibitor stimulators, NFAT gene inhibitors, NFE2L2 gene stimulators, nicotinic acetylcholine receptor antagonists, NK cell receptor modulators, NKG2 A B activating NK receptor antagonists, NKG2 D activating NK receptor antagonists, nuclear erythroid 2-related factor 2 stimulators, nuclear factor kappa B inhibitors, nuclear factor kappa B modulators, nuclear factor kappa B p105 inhibitors, opioid growth factor receptor agonists, opioid receptor delta antagonists, osteoclast differentiation factor antagonists, osteoclast differentiation factor ligand inhibitors, oxidoreductase inhibitors, P2X7 purinoceptor agonists, p38 MAP kinase alpha inhibitors, p38 MAP kinase inhibitors, PDE 4 inhibitors, PDE 5 inhibitors, PDGF receptor agonists, PDGF receptor antagonists, PDGF-B ligand inhibitors, PERK gene inhibitors, phosphoinositide-3 kinase delta inhibitors, phosphoinositide-3 kinase gamma inhibitors, phospholipase A2 inhibitors, platelet activating factor receptor antagonists, PPAR gamma agonists, programmed cell death protein 1 modulators, prostaglandin D synthase stimulators, protein arginine deiminase inhibitors, protein tyrosine kinase inhibitors, protease- activated receptor-2 antagonist, PurH purine biosynthesis protein inhibitors, rho associated protein kinase 2 inhibitors, seprase inhibitors, signal transducer CD24 modulators, signal transduction inhibitors, sodium glucose transporter-2 inhibitors, sphingosine 1 phosphate phosphatase modulators, STAT3 gene inhibitors, serum amyloid A protein modulator, superoxide dismutase stimulators, SYK family tyrosine kinase inhibitors, Syk tyrosine kinase inhibitors, syndecan-1 inhibitors, T cell receptor antagonists, T cell receptor modulators, T cell surface glycoprotein CD28 inhibitors, T cell surface glycoprotein CD28 stimulators, TAK1 binding protein modulators, talin modulators, T-cell differentiation antigen CD6 inhibitors, T- cell surface glycoprotein CD8 inhibitors, tenascin modulators, TGF beta agonists, thymulin agonists, TLR-2 antagonists, TLR-4 antagonists, TLR-9 antagonists, TNF alpha ligand inhibitors, TNF alpha ligand modulators, TNF antagonists, TNF gene inhibitors, TNF receptor modulators, TNFSF11 gene inhibitors, transcription factor p65 inhibitors, transcription factor RelB inhibitors, transferrin modulators, transthyretin modulator, tumor necrosis factor 13C receptor antagonists, tumor necrosis factor 15 ligand inhibitors, tumor necrosis factor ligand 13 inhibitors, tumor necrosis factor ligand inhibitors, type I IL-1 receptor antagonists, type I TNF receptor antagonists, type II TNF receptor modulators, unspecified GPCR agonists, VEGF receptor antagonists, VEGF-2 receptor antagonists, VEGF-2 receptor modulators, VEGF-B ligand inhibitors, X-linked inhibitor of apoptosis protein inhibitors, and zap70 tyrosine kinase inhibitors. [0600] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from 99mTc labelled annexin V-128, abatacept, abatacept biosimilar, ABBV- 257, ABT-122, ABT-494, acalabrutinib, aceclofenac, actarit, AdMSCs, MS-392, adalimumab, adalimumab biosimilar, adalimumab follow-on biologic, AK-106, ALX-0061, Amilo-5MER, aminopterin, AMT-101, anakinra, anakinra biosimilar, anakinra follow-on biologic, annexuzlimab, ARG-301, ARQ-250, ASLAN-003, ASP-5094, AT-132, AZD-9567, baricitinib, BI-655064, bimekizumab, BiP (rheumatoid arthritis), BLHP-006, blisibimod, BMS-986104, BMS-986142, ABBV-105, BTT-1023, canakinumab, Cartistem, CCX-354, CD24-IgFc, celecoxib, cerdulatinib, certolizumab pegol, CF-101, CFZ-533, CHR-5154, cibinetide, ciclosporin, clazakizumab, CNTO-6785, corticotropin, CR-6086, CreaVax-RA, CWG-92, CWG-940, Cx-611, DE-098, DEN-181, deflazacort, Rheumavax, denosumab, diacerein, diclofenac, DWJ-1421, E-6011, eicosapentaenoic acid monoglycerides, etanercept, etanercept biosimilar, etanercept follow-on biologic, etodolac, etoricoxib, filgotinib, fosdagrocorat, GLPG- 3970, gerilimzumab, ginsenoside C-K, givinostat, GLPG-4399, goat polyclonal antibodies, golimumab, GS-5745, GS-9876, GSK-3196165, HHT-109, HM-71224, HMPL-523, HST-003, hyaluronate sodium, (S)-hydroxychloroquine, IB-RA (injectable, rheumatoid arthritis), IB-RA (oral, rheumatoid arthritis), IcanoMAB, ICP-022, iguratimod, IMD-2560, imidazole salicylate, infliximab, infliximab biobetter, infliximab biosimilar, CT-P13, INSIX RA, interferon gamma follow-on biologic, interleukin-2 (injectable), interleukin-2 follow-on biologic, INV-103, IR- 501, itolizumab, JNJ-40346527, Ka Shu Ning, KB-312, KD-025, ketoprofen + omeprazole, KINE-101, LB-600, leflunomide, lenzilumab, LLDT-8, LNK-01001, LNP-1955, lumiracoxib, LY-3090106, masitinib, mavrilimumab, MBS-2320, MEDI-5117, meloxicam, methotrexate, MGD-010, misoprostol + diclofenac, MM-A01-01, monalizumab, MORAb-022, MPC-300-IV, MRC-375, nabumetone, namilumab, naproxen + esomeprazole, naproxen + esomeprazole strontium, NIP-046, ocaratuzumab, ofatumumab, OHR-118 , olokizumab, OM-89, once-daily naproxen (oral controlled release, pain), ONO-4059, Oralgam, ozoralizumab, PAR-2 inhibitors, peficitinib, pelubiprofen, PF-06687234, piperidone hydrochloridum, piroxicam, prednisolone, prednisone, Procell, Prosorba, PRT-2607, PRTX-100, PRX-167700, QBSAU, rabeximod, RCT- 18, recombinant human CD22 monoclonal antibody (iv infusion), Lonn Ryonn Pharma/SinoMab Bioscience (Shenzhen), RA-Curcusome, recombinant human interleukin-1 receptor antagonist (rheumatoid arthritis), , recombinant human interleukin-2 recombinant TNF receptor 2-Fc fusion protein mutant , RG-6125, RhuDex, rifabutin + clarithromycin + clofazimine, rituximab, rituximab biosimilar, Toritz, rituximab follow-on biologic, RPI-78, SAN-300, sarilumab, SBI-087, seliciclib, SHR-0302, sirukumab, spebrutinib, SR-047, SSS-07, KDDF-201110-06, Syn-1002, T-5224, TAB-08, tacrolimus, TAK-020, TAK-079, tarenflurbil (transdermal spraygel, skin disease/rheumatoid arthritis), technetium Tc 99m tilmanocept, technetium[99Tc] methylenediphosphonate, tenoxicam, Debio-0512, tocilizumab, tofacitinib, tofacitinib citrate, TQG-2813, Trichuris suis ova, umbilical cord-derived mesenchymal stem cells (iv, RA/liver disease), ustekinumab, VAY-736, VB-201, WF-10, XmAb-5871, YH-1713, YHB-1411-2, YRA-1909, and ZM-008, or a pharmaceutically acceptable salt of any of the foregoing, or any combination thereof. [0601] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one, two, three, or four additional therapeutic agents selected from: ● 14-3-3 protein eta inhibitors, including but not limited to anti-AGX-020 mAbs (rheumatoid arthritis) and Augurex; ● 5-Lipoxygenase inhibitors, including but not limited to darbufelone, tebufelone, ZD- 2138, etalocib, PGV-20229, L-708780, T-0757, T-0799, ZM-216800, L-699333, BU- 4601A, and SKF-104351; ● 5-Lipoxygenase/Cyclooxygenase inhibitors, including but not limited to tenoxicam, licofelone, tenidap, tepoxalin, flobufen, SKF-86002, WY-28342, and CI-986; ● 5-Lipoxygenase/PPAR gamma agonists, including but not limited to etalocib; ● Abl tyrosine kinase inhibitors/Bcr protein inhibitors/Kit tyrosine kinase inhibitors/PDGF receptor antagonists/ Signal transduction inhibitors, including but not limited to imatinib; ● ACTH receptor agonists/Adrenocorticotrophic hormone ligands/Opioid growth factor receptor agonists, including but not limited to FAR-404 and metenkefalin acetate + tridecactide acetate; ● adenosine A1 receptor antagonist, including but not limited to CP-25; ● adenosine A3 receptor agonists, including but not limited to CF-101 (piclidenoson); ● adenosine deaminase inhibitors, cladribine, pentostatin, and FR-221647; ● ADP ribosyl cyclase-1 inhibitors, including but not limited to daratumumab; ● ADP ribosyl cyclase-1 modulators/syndecan-1 inhibitors, including but not limited to indatuximab ravtansine; ● ADP ribosylation factor 6 inhibitors, including but not limited to NAV-2729; ● adrenocorticotrophic hormone ligands, including but not limited to corticotropin and Mallinckrodt; ● aggrecanase-2/TNF gene inhibitors, including but not limited to GIBH-R-001-2; ● albumin modulators, including but not limited to ONS-1210; ● albumin modulators/IL-6 antagonists, including but not limited to ALX-0061 (vobarilizumab); ● albumin modulators/TNF alpha ligand inhibitors, including but not limited to HOT-3010; ● AP1 transcription factor/nuclear factor kappa B inhibitors, including but not limited to tarenflurbil and SP-100030; ● anti-TNF steroid antibody-drug conjugates (anti-TNF-GRM), including but not limited to ABBV-3373 and ABBV-154; ● basigin inhibitors/branched amino acid aminotransferase 1/metalloprotease-9 inhibitors/metalloprotease-2 inhibitors, including but not limited to ERG-240; ● BET inhibitors, including but not limited to GSK-3358699; ● bispecific anti-CD86/IL-10, including but not limited to APVO-210; ● bispecific humanized monoclonal antibody targeted against BAFF and IL-17A, including but not limited to tibulizumab; ● bispecific antibody-peptide conjugate (BAFF/ICOSL), including but not limited to AMG-570; ● B-lymphocyte antigen CD19 inhibitors, including but not limited to MDX-1342; ● B-lymphocyte antigen CD19 inhibitors/immunoglobulin gamma Fc receptor IIB antagonists, including but not limited to XmAb-5871; ● B-lymphocyte antigen CD20 inhibitors, including but not limited to ocrelizumab, ofatumumab, rituximab, ABP-798, Maball, Mabtas, Reditux, Zytux, veltuzumab, ocaratuzumab, BLX-301, IDEC-102, ABP-798, GP-2013, MK-8808, HLX-01, CT-P10, TL-011, PF-05280586, IBPM-001RX, IBI-301, AME-133v, BCD-020, BT-D004, SAIT- 101, and JHL-1101; ● B-lymphocyte antigen CD20 modulators, including but not limited to SBI-087, TRU- 015, DXL-625, and MabionCD20; ● B-lymphocyte cell adhesion molecule inhibitor, including but not limited to SM-06; ● B-lymphocyte stimulator ligand inhibitors, including but not limited to belimumab, RCT-18, blisibimod, tabalumab, and briobacept; ● B-lymphocyte stimulator ligand/Tumor necrosis factor ligand 13 inhibitors, including but not limited to atacicept; ● bradykinin receptor modulators/histone deacetylase inhibitors/P2X7 purinoceptor agonists, including but not limited to givinostat; ● BRAF gene/MEK protein kinase/PERK gene inhibitors, including but not limited to binimetinib; ● Bromodomain containing protein inhibitors, including but not limited to RVX-297, ZEN-003694 ● Btk tyrosine kinase inhibitors, including but not limited to AC-0058, acalabrutinib, HM- 71224, spebrutinib, BMS-986142, TAK-020, tirabrutinib (ONO-4059), TAS-5315, ABBV-105, GDC-0834, EBI-1459, BMS-986195, evobrutinib, fenebrutinib, SIMM-016, and YZJ-3058; ● Btk tyrosine kinase inhibitors/Syk tyrosine kinase inhibitors/ VEGF-2 receptor antagonists, including but not limited to CG-026806; ● Btk tyrosine kinase inhibitors/IL-6 antagonists, including but not limited to RN-486; ● Btk tyrosine kinase/Jak1 tyrosine kinase inhibitors, including but not limited to upadacitinib + ABBV-105; ● Btk tyrosine kinase/Jak3 tyrosine kinase inhibitors, including but not limited to AC- 0025; ● cadherin-11 antagonists, including but not limited to RG-6125; ● calcineurin inhibitors, including but not limited to ciclosporin; ● calcineurin inhibitors/opioid receptor delta antagonists, including but not limited to HS- 378; ● calcium channel inhibitors, including but not limited to RP-3128; ● calreticulin inhibitor, including but not limited to ALB-001 and ZYBK-2; ● carbonic anhydrase/cyclooxygenase 2 inhibitors, including but not limited to polmacoxib; ● cathepsin K inhibitors, including but not limited to CRA-013783 and VEL-0230; ● cathepsin K/cathepsin S inhibitors, including but not limited to AM-3876 and NPI-2019; ● cathepsin S inhibitors, including but not limited to MIV-247 and RWJ-445380; ● CCR1 chemokine antagonists, including but not limited to BX-471, BMS-817399, BI- 638683, CCX-354, MLN-3701, MLN-3897, CP-481715, and PS-375179; ● CCR2 chemokine antagonists, including but not limited to MK-0812 and AZD-6942; ● CCR3 gene modulators/eotaxin 2 ligand inhibitors, including but not limited to CM-102; ● CCR5 chemokine antagonists, including but not limited to OHR-118, NIBR-6465, AZD- 5672, and AZD-8566; ● CD29 modulators/interleukin-10 ligands, including but not limited to PF-06687234; ● CD3 modulators, including but not limited to otelixizumab; ● CD39/CD73 agonists, including but not limited to AAV5-CD39/CD73 (rheumatoid arthritis), and Arthrogen; ● CCR5 chemokine antagonists/CD4 agonists/HIV-1 gp120 protein inhibitors, including but not limited to maraviroc; ● CD4 antagonists, including but not limited to zanolimumab, MTRX-1011A, BW- 4162W94, EP-1645, clenoliximab, and DerG-PG275Cit; ● CD40 ligand inhibitors, including but not limited to dapirolizumab pegol, and TNX- 1500; ● CD40 ligand receptor antagonists, including but not limited to BI-655064, anti-CD40- XTEN, teneliximab, VIB-4920, and iscalimab; ● CD40 ligand receptor modulators/immunoglobulin G1 modulators, including but not limited to CFZ-533; ● CD52 antagonists/clusterin stimulators, including but not limited to alemtuzumab; ● bispecific CD32B/CD79B antibody, including but not limited to PRV-3279 (MGD-010); ● CD80 antagonists, including but not limited to abatacept biobetter; ● CD80 antagonists/T cell surface glycoprotein CD28 inhibitors, including but not limited to RhuDex; ● CD80 antagonists/CD86 antagonists, including but not limited to XENP-9523 and ASP- 2408; ● CD86 antagonists, including but not limited to abatacept biosuperior; ● CD86 antagonists/cytotoxic T-lymphocyte protein-4 modulators, including but not limited to ES-210; ● CD95 antagonists, including but not limited to DE-098 and CS-9507; ● Cell adhesion molecule inhibitors, including but not limited to alicaforsen, NPC-17923, TK-280, and PD-144795; ● Chemokine receptor antagonists, including but not limited to PF-06835375; ● Complement C5 factor inhibitors, including but not limited to eculizumab, ● Complement C5 factor inhibitors/IL-1 antagonists, including but not limited to antisense oligonucleotides (rheumatoid arthritis) and Leiden University Medical Center Complement Factor stimulators, including but not limited to CM-101; ● C-reactive protein inhibitors, including but not limited to ISIS-353512; ● C-reactive protein inhibitors/cyclooxygenase 2 inhibitors/Nuclear factor kappa B inhibitors/ immunoglobulin M antagonists/IL-2 receptor antagonists/PGE2 antagonists: IB-RACSF-1 antagonists, including but not limited to masitinib, FPA-008, JNJ- 27301937, JNJ-40346527, PLX-5622, CT-1578, PD-360324, and JNJ-28312141; ● CSF-1 antagonists/Fyn tyrosine kinase inhibitors/Kit tyrosine kinase inhibitors/Lyn tyrosine kinase inhibitors/NK cell receptor modulators/PDGF receptor antagonists, including but not limited to masitinib; ● CXC10 chemokine ligand inhibitors, including but not limited to 946414-98-8 and BMS- 936557; ● CXCR4 chemokine antagonists, including but not limited to plerixafor; ● CDK-2/7/9 inhibitors/MCL1 gene inhibitors, including but not limited to seliciclib; ● CDK-1/2/5/7/9 inhibitors, including but not limited to BP-14; ● Chaperonin modulator, including but not limited to IRL-201805; ● cyclooxygenase 2 inhibitors, including but not limited to celecoxib, etoricoxib, meloxicam, and lumiracoxib; ● cyclooxygenase 2/oxidoreductase inhibitors, including but not limited to etodolac; ● cyclooxygenase 2 modulators, including but not limited to DRGT-46; ● cyclooxygenase inhibitors, including but not limited to aceclofenac, diclofenac, naproxcinod, naproxen etemesil, nabumetone, Aleve, pelubiprofen, LY-210073, NS-398, bromfenac, L-746483, LY-255283, ibuprofen, flurbiprofen, SC-57666, and bermoprofen; ● cyclooxygenase inhibitors/H+ K+ ATPase inhibitors, including but not limited to naproxen + esomeprazole strontium; ● cyclooxygenase inhibitors/PGE1 agonists, including but not limited to misoprostol + diclofenac; ● cyclooxygenase inhibitors/oxidoreductase inhibitors, including but not limited to imidazole salicylate; ● cytosolic phospholipase A2 inhibitors/phospholipase A2 inhibitors, including but not limited to AVX-002; ● cytotoxic T-lymphocyte protein-4 stimulators/ T cell surface glycoprotein CD28 inhibitors, including but not limited to abatacept, BMS-188667, and belatacept; ● deoxyribonuclease gamma stimulator, including but not limited to NTR-441; ● DHFR inhibitors, including but not limited to MPI-2505, Jylamvo, and ZeNEO- Methotrexate; ● DHFR inhibitors/folate antagonists/transferrin modulators, including but not limited to methotrexate; ● diamine acetyltransferase inhibitors, including but not limited to diminazene aceturate; ● dihydroorotate dehydrogenase inhibitors, including but not limited to ASLAN-003, HWA-486, and ABR-224050; ● dihydroorotate dehydrogenase/protein tyrosine kinase inhibitors, including but not limited to leflunomide; ● DYRK-1 alpha protein kinase inhibitor, including but not limited to VRN-02; ● elongation factor 2 inhibitors/ interleukin-2 ligands/ NAD ADP ribosyltransferase stimulators, including but not limited to denileukin diftitox; ● enolase 1 inhibitor, including but not limited to HuL-001; ● EP4 prostanoid receptor antagonists, including but not limited to CR-6086; ● erythropoietin receptor agonists, including but not limited to cibinetide; ● Fas ligands, including but not limited to AP-300; ● FGF-2 ligand inhibitors, including but not limited to RBM-007; ● FK506 binding protein-12 modulators/mTOR inhibitors, including but not limited to temsirolimus; ● folate antagonists/ transferrin modulators/ DHFR inhibitors, including but not limited to MBP-Y003; ● folate receptor modulators, including but not limited to technetium (99mTc) etarfolatide; ● fractalkine ligand inhibitors, including but not limited to E-6011; ● Fyn tyrosine kinase inhibitors/ GABA A receptor modulators/ cyclooxygenase 2 inhibitors/ dihydroorotate dehydrogenase inhibitors, including but not limited to laflunimus; ● glucocorticoid agonists, including but not limited to prednisone, prednisolone, and fosdagrocorat; ● glucocorticoid antagonists, including but not limited to REC-200; ● glucocorticoid induced leucine zipper stimulators, including but not limited to ART-G01; ● GM-CSF ligand inhibitors, including but not limited to namilumab, gimsilumab (MORAb-022), and TJM-2; ● GM-CSF receptor antagonists, including but not limited to mavrilimumab; ● GM-CSF receptor modulators, including but not limited to GSK-3196165 and otilimab; ● growth regulated protein alpha ligand inhibitors/AP1 transcription factor inhibitors/ IL-6 antagonists/ interleukin-1 beta ligand inhibitors/cathepsin K inhibitors/ NFAT gene inhibitors, including but not limited to T-5224; ● H+ K+ ATPase inhibitors, including but not limited to naproxen + esomeprazole, ketoprofen + omeprazole, KEO-25001, HC-1004, and PN-40020; ● histamine H4 receptor antagonists, including but not limited to toreforant and GD-48; ● histone deacetylase inhibitors, including but not limited to CHR-5154 (GSK-3117391) and NIPEP-CARE; ● histone deacetylase-6 inhibitors, including but not limited to CKD-506; ● HLA class II antigen DQ-2 alpha modulators, including but not limited to NexVax2; ● HLA class II antigen inhibitors, including but not limited to HLA-DR1/DR4 inhibitors (rheumatoid arthritis) and Provid; ● HLA class II antigen modulators, including but not limited to recombinant T-cell receptor ligand (rheumatoid arthritis) and Artielle; ● Hsp 70 family inhibitors, including but not limited to gusperimus trihydrochloride; ● hypoxia inducible factor-1 inhibitors/ VEGF receptor antagonists, including but not limited to 2-methoxyestradiol; ● IFNB gene stimulators, including but not limited to ART-102; ● I-kappa B kinase beta inhibitors, including but not limited to IMD-2560; ● I-kappa B kinase beta inhibitors/Nuclear factor kappa B inhibitors, including but not limited to IMD-0560; ● I-kappa B kinase inhibitors/ NFE2L2 gene stimulators/ Nuclear factor kappa B inhibitors/ STAT3 gene inhibitors, including but not limited to bardoxolone methyl; ● IL-1 antagonists, including but not limited to recombinant human interleukin-1 receptor antagonist (rheumatoid arthritis), Shanghai Fudan-Zhangjiang Bio-Pharmaceutical; ● IL-1 antagonists/interleukin-1 beta ligand inhibitors, including but not limited to rilonacept; ● IL-10 agonists, including but not limited to peg-ilodecakin; ● IL-11 agonists/PDGF receptor agonists, including but not limited to oprelvekin; ● IL-12 antagonists/IL-23 antagonists, including but not limited to ustekinumab and briakinumab; ● IL-15 antagonists, including but not limited to AMG-714; ● IL-17 antagonists, including but not limited to ixekizumab and secukinumab; ● IL-17 receptor modulators, including but not limited to CNTO-6785; ● IL-2 receptor agonists, including but not limited to interleukin-2 follow-on biologic (IL- 2), Anteluke, and Interking; ● IL-2/IL-21/IL-15 antagonists, including but not limited to BNZ-132-2; ● IL-21 antagonists, including but not limited to NN-8828; ● IL-4 agonists, including but not limited to SER-130-AMI; ● IL-6 antagonists, including but not limited to BCD-089, olokizumab, clazakizumab, sirukumab, SA-237, FB-704A, OP-R003, peptide IL-6 antagonist, MEDI-5117, AMG- 220, FM-101, BLX-1025, esonarimod, TA-383, and sarilumab; ● IL-6 antagonists/interleukin-1 beta ligand inhibitors/ TNF alpha ligand inhibitors, including but not limited to K-832; ● IL-6 antagonists/insulin sensitizers/ interleukin-1 beta ligand inhibitors, including but not limited to BLX-1002; ● IL-6 receptor antagonists/modulators, including but not limited to tocilizumab, HS-628, and LusiNEX; ● IL-6 receptor modulators, including but not limited to BAT-1806 and RO-4877533; ● immunoglobulin antagonists, including but not limited to iguratimod; ● immunoglobulin G1 agonists, including but not limited to BX-2922 and HF-1020; ● immunoglobulin G1 agonists/interleukin-1 beta ligand inhibitors, including but not limited to canakinumab; ● immunoglobulin G1 agonists/TNF alpha ligand inhibitors, including but not limited to STI-002; ● immunoglobulin G1 antagonists/ TNF alpha ligand inhibitors, including but not limited to YHB-1411-2; ● immunoglobulin G1 modulators/ GM-CSF ligand inhibitors/ immunoglobulin kappa modulators, including but not limited to lenzilumab; ● immunoglobulin G2 antagonists/ NF kappa B inhibitor stimulators/ osteoclast differentiation factor antagonists/ osteoclast differentiation factor ligand inhibitors/ TNFSF11 gene inhibitors, including but not limited to denosumab; ● immunoglobulin gamma Fc receptor II modulators, including but not limited to MGD- 010; ● inducible nitric oxide synthase inhibitors/ cyclooxygenase 2 inhibitors/ MAP kinase modulators/ nuclear factor kappa B inhibitors, including but not limited to SKLB-023; ● inosine monophosphate dehydrogenase inhibitors, including but not limited to mizoribine; ● insulin sensitizers/ nuclear factor kappa B inhibitors/interleukin ligand inhibitors, including but not limited to HE-3286; ● integrin alpha-1/beta-1 antagonists, including but not limited to SAN-300; ● integrin alpha-4/beta-1 antagonists/cell adhesion molecule inhibitors, including but not limited to natalizumab; ● integrin alpha-9 antagonist, including but not limited to ASP-5094; ● integrin antagonists, including but not limited to PEG-HM-3 and CY-9652; ● interferon beta ligands, including but not limited to recombinant interferon beta-1a; ● interferon beta ligands/IL-6 antagonists, including but not limited to TA-383; ● interferon gamma ligands, including but not limited to Li Zhu Yin De Fu and Clongamma; ● interleukin 17A ligand inhibitors/tumor necrosis factor ligand inhibitors, including but not limited to ABT-122 and ABBV-257; ● interleukin 17F ligand inhibitors, including but not limited to bimekizumab; ● interleukin 18 ligand inhibitors, including but not limited to tadekinig alfa; ● interleukin 23A inhibitors, including but not limited to guselkumab; ● interleukin ligands/IL-1 antagonists, including but not limited to IBPB-007-IL; ● interleukin receptor 17A antagonists, including but not limited to brodalumab; ● interleukin-1 beta ligand inhibitors, including but not limited to gevokizumab, LY- 2189102, CDP-484, and AR-100; ● interleukin-1 beta ligand inhibitors/TNF alpha ligand inhibitors, including but not limited to PMI-001; ● interleukin-1 beta ligands/TNF alpha ligand modulators, including but not limited to PUR-0110; ● interleukin-2 ligands, including but not limited to recombinant interleukin-2 and CUG- 252; ● IL-2 modulators, including but not limited to AMG-592; ● interleukin-4 ligands/tenascin modulator, including but not limited to Tetravil; ● interleukin-6 ligand inhibitors, including but not limited to gerilimzumab and PF- 4236921; ● IRAK-4 protein kinase inhibitor, including but not limited to BAY-1830839, BAY- 1834845, PF-06650833, and KT-474; ● Itk tyrosine kinase inhibitors, including but not limited to JTE-051; ● Itk tyrosine kinase inhibitors/Jak3 tyrosine kinase inhibitors, including but not limited to ARN-4079; ● JAK tyrosine kinase inhibitors, including but not limited to deuterated tofacitinib analog, SD-900, and WXSH-0150; ● JAK tyrosine kinase inhibitors/Syk tyrosine kinase inhibitors, including but not limited to cerdulatinib and CVXL-0074; ● Jak1 tyrosine kinase inhibitors, including but not limited to ABT-494 (upadacitinib), ruxolitinib, filgotinib, itacitinib, NIP-585, YJC-50018, GLPG-0555, MRK-12, and SHR-0302; ● Jak1/3 tyrosine kinase inhibitors, including but not limited to tofacitinib, tofacitinib citrate, peficitinib, CKD-374, and CS-944X; ● JAK 1/3 inhibitor/ROCK1/2 inhibitor: CPL-409116 ● Jak1/2 tyrosine kinase inhibitors, including but not limited to baricitinib, ruxolitinib, LW-104, and TLL-018; ● Jak2 tyrosine kinase inhibitors/CSF-1 antagonists, including but not limited to CT-1578; ● JAK3 gene inhibitors, including but not limited to PF-06651600; ● Jak3 tyrosine kinase inhibitors, including but not limited to decernotinib, DNX-04042, MTF-003, and PS-020613; ● Jun N terminal kinase inhibitors, including but not limited to IQ-1S; ● KCNA voltage-gated potassium channel-3 modulators, including but not limited to MRAD-P1; ● Kelch like ECH associated protein 1 modulators/Nuclear erythroid 2-related factor 2 stimulators, including but not limited to dimethyl fumarate; ● LanC like protein 2 modulators, including but not limited to BT-11 and BT-104; ● LDL receptor related protein-1 stimulator, including but not limited to SP-16; ● leukotriene BLT receptor antagonists/complement C5 factor inhibitors, including but not limited to nomacopan; ● LITAF gene inhibitors/JAK3 gene inhibitors/ MAP3K2 gene inhibitors/ TNF antagonists, including but not limited to GBL-5b; ● Lymphocyte function antigen-3 receptor antagonists, including but not limited to alefacept; ● Macrophage mannose receptor 1 modulators, including but not limited to technetium Tc 99m tilmanocept; ● MAdCAM inhibitors/immunoglobulin G2 modulators, including but not limited to PF- 547659; ● MAPKAPK5 inhibitors/ matrix metalloprotease inhibitors, including but not limited to GLPG-0259; ● MEK protein kinase inhibitors, including but not limited to AD-GL0001; ● membrane copper amine oxidase inhibitors, including but not limited to BTT-1023, PRX-167700, and vepalimomab; ● metalloprotease-9 inhibitors, including but not limited to GS-5745; ● microbiome modulator, including but not limited to EDP-1815; ● midkine ligand inhibitors, including but not limited to CAB-102; ● mitochondrial 10 kDa heat shock protein stimulators, including but not limited to INV- 103; ● mTOR inhibitors, including but not limited to everolimus; ● NAMPT gene inhibitors, including but not limited to ART-D01; ● Nicotinic acetylcholine receptor antagonists, including but not limited to RPI-78 and RPI-MN; ● NKG2 A B activating NK receptor antagonists, including but not limited to monalizumab; ● NKG2 D activating NK receptor antagonists, including but not limited to NNC-0142- 002; ● nuclear factor kappa B inhibitors, including but not limited to dehydroxymethylepoxyquinomicin, MP-42, VGX-1027, SP-650003, MG-132, SIM- 916, VGX-350, VGX-300, GIT-027, MLN-1145, and NVP-IKK-005; ● nuclear factor kappa B modulators/ nuclear factor kappa B p105 inhibitors / transcription factor RelB inhibitors/ transcription factor p65 inhibitors, including but not limited to REM-1086; ● osteoclast differentiation factor antagonists, including but not limited to cyclic peptidomimetics (rheumatoid arthritis/osteoporosis), University of Michigan; ● p38 MAP kinase alpha inhibitors, including but not limited to VX-745, BMS-582949, and BMS-751324; ● p38 MAP kinase inhibitors, including but not limited to BCT-197, losmapimod, and ARRY-797; ● PDE 4 inhibitors, including but not limited to apremilast; ● PDE 5 inhibitors, including but not limited to PDE5 inhibitors (rheumatoid arthritis), University of Rochester; ● PDGF-B ligand inhibitors/VEGF receptor antagonists, including but not limited to SL- 1026; ● phosphoinositide-3 kinase delta inhibitors, including but not limited to CT-732, INK- 007, and GNE-293; ● phosphoinositide-3 kinase delta/gamma inhibitors, including but not limited to duvelisib and RP-6503; ● phospholipase A2 inhibitors, including but not limited to AK-106, varespladib methyl, Ro-31-4493, BM-162353, Ro-23-9358, and YM-26734; ● platelet activating factor receptor antagonists, including but not limited to piperidone hydrochloridum; ● PPAR gamma agonists, including but not limited to rosiglitazone XR; ● PPAR gamma agonists/insulin sensitizers, including but not limited to rosiglitazone; ● programmed cell death protein 1 modulators, including but not limited to INSIX RA; ● prostaglandin D synthase stimulators, including but not limited to HF-0220; ● protein tyrosine kinase inhibitors, including but not limited to tairuimide; ● PurH purine biosynthesis protein inhibitors/ inosine monophosphate dehydrogenase inhibitors, including but not limited to mycophenolate mofetil; ● Rev protein modulators, including but not limited to ABX-464; ● RIP-1 kinase inhibitors, including but not limited to GSK-2982772 and VRN-04; ● IL-17 antagonist/ rho associated protein kinase 2 inhibitor, including but not limited to KD-025; ● signal transducer CD24 modulators, including but not limited to CD24-IgFc; ● sodium glucose transporter-2 inhibitors/PPAR gamma agonists/ insulin sensitizers, including but not limited to THR-0921; ● STAT3 gene inhibitors, including but not limited to vidofludimus; ● STAT-3 inhibitors, including but not limited to HL-237; ● Superoxide dismutase stimulators, including but not limited to imisopasem manganese; ● SYK family tyrosine kinase inhibitors/Zap70 tyrosine kinase inhibitors, including but not limited to MK-8457; ● Syk tyrosine kinase inhibitors, including but not limited to fostamatinib, entospletinib, KDDF-201110-06, HMPL-523, AB-8779, GS-9876, PRT-2607, CG-103065, and SKI- O-703; ● T cell receptor antagonists, including but not limited to TCR inhibiting SCHOOL peptides (systemic/topical, rheumatoid arthritis/dermatitis/scleroderma), SignaBlok and CII modified peptide (rheumatoid arthritis), Peking University; ● T cell receptor modulators/ HLA class II antigen modulators, including but not limited to ARG-301; ● T cell surface glycoprotein CD28 stimulators, including but not limited to TAB-08 and theralizumab; ● TAK1 binding protein modulators, including but not limited to epigallocatechin 3- gallate; ● Talin modulators, including but not limited to short-form talin regulators (rheumatoid arthritis), KayteeBio; ● T-cell differentiation antigen CD6 inhibitors, including but not limited to itolizumab; ● T-cell surface glycoprotein CD8 inhibitors/TGF beta agonists/CD4 antagonists, including but not limited to tregalizumab; ● thymulin agonists, including but not limited to Syn-1002; ● TLR-2/TLR-4 antagonists, including but not limited to VB-201; ● TLR-4 antagonists, including but not limited to NI-0101; ● TLR-2/4/9 antagonists, including but not limited to P-13; ● TNF agonists/TNF antagonists/type II TNF receptor modulators, including but not limited to Lifmior; ● TNF alpha ligand inhibitors , including but not limited to Adfrar, FKB-327, Exemptia, Cinnora, Mabura, adalimumab, infliximab, Flixabi, PF-06438179, hadlima, recombinant humanized anti-TNF-alpha monoclonal antibody, CMAB-008, CT-P13, GB-242, golimumab (CNTO-148), ozoralizumab, AT-132, , ISIS-104838, ISU-202, CT-P17, MB- 612, Debio-0512, anti-TNF alpha human monoclonal antibody, UB-721, KN-002, DA- 3113, BX-2922, R-TPR-015, BOW-050, PF-06410293, CKD-760, CHS-1420, GS-071, ABP-710, BOW-015, HLX-03, BI-695501, MYL-1401A, ABP-501, BAX-2923, SCH- 215596, ABT-D2E7, BAT-1406, XPro-1595, Atsttrin, SSS-07, golimumab biosimilar, TA-101, , BLX-1002, ABX-0401, TAQ-588, TeHL-1, placulumab, CYT-007-TNFQb, SSR-150106, PassTNF, Verigen, DOM-0200, DOM-0215, AME-527, anti-TNF-alpha mAb, GENZ-38167, BLX-1028, CYT-020-TNFQb, CC-1080, CC-1069, LBAL, GP- 2017, Idacio, IBI-303, HS-016, TNF-2, and IA-14069; ● TNF alpha ligand inhibitors/ TNF antagonists/ type II TNF receptor modulators, including but not limited to BAX-2200; ● TNF alpha ligand inhibitors/Type II TNF receptor modulators, including but not limited to Eucept, TNF alpha ligand modulators: MM-A01-01, CDP-571, camobucol, and JNJ- 63823539; ● TNF antagonists, including but not limited to DNX-114, TNF antagonist + IL-12 antagonist (rheumatoid arthritis), University of Oxford, BN-006, pegsunercept, ACE- 772, onercept, DE-096, PN-0615, lenercept, ITF-1779, MDL-201112, HD-203, Qiangke, and TNF a Fc; ● TNF antagonists/type II TNF receptor modulators, including but not limited to Altebrel, Intacept, QL-0902, etanercept, Erelzi, opinercept, YISAIPU, Anbainuo, Benepali, YLB- 113, SCB-808, DA-3853, and SCB-131; ● TNF antagonists/TNF alpha ligand inhibitors, including but not limited to certolizumab pegol; ● TNF receptor modulators, including but not limited to recombinant TNF receptor 2-Fc fusion protein mutant, T-0001; ● TNF receptor modulators/TNF alpha ligand inhibitors, including but not limited to tgAAV-TNFR:Fc; ● tumor necrosis factor 13C receptor antagonists, including but not limited to VAY-736; ● tumor necrosis factor 15 ligand inhibitors, including but not limited to anti-TL1A antibodies (rheumatoid arthritis/inflammatory bowel disease), NIAMS; ● tumor necrosis factor ligand inhibitors, including but not limited to etanercept biosimilar; ● type I IL-1 receptor antagonists, including but not limited to anakinra, IL-1 Ra, anakinra follow-on biologic, and AXXO; ● type I TNF receptor antagonists, including but not limited to NM-940 and EN-2001; ● type II TNF receptor modulators, including but not limited to LBEC-0101, DMB-3853, DWP-422, and BT-D001; ● unspecified GPCR agonists, including but not limited to NCP-70X; ● VEGF receptor antagonists, including but not limited to NSC-650853; ● VEGF-2 receptor modulators, including but not limited to VEGFR2 neutralizing antibody (rheumatoid arthritis), University of Rochester; ● VEGF-B ligand inhibitors, including but not limited to CSL-346; ● X-linked inhibitor of apoptosis protein inhibitors, including but not limited to IAP inhibitors (oral), Pharmascience; and ● Zap70 tyrosine kinase inhibitors, including but not limited to CT-5332. Inflammatory Bowel Disease Combination Therapy [0602] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one or more additional therapeutic agents that treat or ameliorate inflammatory bowel disease (IBD). [0603] The term “inflammatory bowel disease” or “IBD” as used herein is a collective term describing inflammatory disorders of the gastrointestinal tract, the most common forms of which are ulcerative colitis and Crohn’s disease. Other forms of IBD that can be treated with the compounds provided herein, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions provided herein include, but are not limited to, diversion colitis, ischemic colitis, infectious colitis, chemical colitis, microscopic colitis (including collagenous colitis and lymphocytic colitis), atypical colitis, pseudomembranous colitis, fulminant colitis, autistic enterocolitis, indeterminate colitis, Behçet's disease, gastroduodenal CD, jejunoileitis, ileitis, ileocolitis, Crohn’s (granulomatous) colitis, irritable bowel syndrome, mucositis, radiation induced enteritis, short bowel syndrome, celiac disease, stomach ulcers, diverticulitis, pouchitis, proctitis, and chronic diarrhea. [0604] Treating or preventing IBD also includes ameliorating or reducing one or more symptoms of IBD. As used herein, the term “symptoms of IBD” refers to detected symptoms such as abdominal pain, diarrhea, rectal bleeding, weight loss, fever, loss of appetite, and other more serious complications, such as dehydration, anemia and malnutrition. A number of such symptoms are subject to quantitative analysis (e.g. weight loss, fever, anemia, etc.). Some symptoms are readily determined from a blood test (e.g. anemia) or a test that detects the presence of blood (e.g. rectal bleeding). The term “wherein said symptoms are reduced” refers to a qualitative or quantitative reduction in detectable symptoms, including but not limited to a detectable impact on the rate of recovery from disease (e.g. rate of weight gain). The diagnosis is typically determined by way of an endoscopic observation of the mucosa, and pathologic examination of endoscopic biopsy specimens. [0605] The course of IBD varies and is often associated with intermittent periods of disease remission and disease exacerbation. Various methods have been described for characterizing disease activity and severity of IBD as well as response to treatment in subjects having IBD. Treatment according to the present methods and uses is generally applicable to a subject having IBD of any level or degree of disease activity. [0606] The methods and uses provided herein can also be applied at any point in the course of the disease. In some embodiments, the methods and uses are applied to a subject having IBD during a time period of remission (i.e., inactive disease). In some embodiments, the present methods and uses provided herein provide benefit by extending the time period of remission (e.g., extending the period of inactive disease) or by preventing, reducing, or delaying the onset of active disease. In some embodiments, the methods and uses provided herein may be applied to a subject having IBD during a period of active disease. In some embodiments, the methods and uses provided herein provide benefit by reducing the duration of the period of active disease, reducing or ameliorating one or more symptoms of IBD, or treating IBD. [0607] Measures for determining efficacy of treatment of IBD in clinical practice have been described and include, for example, the following: symptom control; fistula closure; extent of corticosteroid therapy required; and improvement in quality of life. Heath-related quality of life (HRQL) can be assessed using the Inflammatory Bowel Disease Questionnaire (IBDQ), which is extensively used in clinical practice to assess quality of life in a subject with IBD. (See Guyatt et al. (1989) Gastroenterology 96:804-810.) [0608] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one or more additional therapeutic agents that treat or ameliorate IBD. Non-limiting examples of therapeutic agents that treat or ameliorate IBC include allogeneic bone marrow-derived MSC therapy, AMP activated protein kinase stimulator, aryl hydrocarbon receptor agonist and T cell receptor modulator, ASK1 inhibitors, beta adrenoceptor antagonists, BTK inhibitors, beta-catenin stimulator, beta-glucuronidase inhibitors, bradykinin receptor modulators, calcineurin inhibitors, calcium channel inhibitors, cathepsin S inhibitors, CCR3 chemokine antagonists, CD40 ligand receptor antagonists, chemokine CXC ligand inhibitors, CHST15 gene inhibitors, collagen modulators, CXCR3 chemokine antagonist, CSF-1 antagonists, cyclooxygenase inhibitors, cytochrome P4503A4 inhibitors, DYRK-1 alpha protein kinase inhibitor, endothelial dysfunction and vascular leakage blocker, enolase 1 inhibitor, eotaxin ligand inhibitors, EP4 prostanoid receptor agonists, erythropoietin receptor agonists, exportin 1 inhibitor, fractalkine ligand inhibitors, free fatty acid receptor 2 antagonists, GATA 3 transcription factor inhibitors, glucagon-like peptide 2 agonists, glucocorticoid agonists, guanylate cyclase receptor agonists, histone deacetylase inhibitors, HLA class II antigen modulators, IL-12 antagonists, IL-13 antagonists, Interleukin-2 ligand, IL- 23 antagonists, IL-6 antagonists, IL-6 receptor modulators, interleukin-7 receptor modulators, IL-7 antagonists, IL-8 antagonists, integrin alpha-4/beta-1 antagonists, integrin alpha-4/beta-7 antagonists, integrin alpha-E antagonists, integrin antagonists, integrin beta-7 antagonists, interleukin ligand inhibitors, Interleukin-10 ligand, interleukin receptor 17A antagonists, Interleukin 23A inhibitor, interleukin-1 beta ligands, interleukin-1 beta ligand modulators, IRAK4 inhibitors, JAK tyrosine kinase inhibitors, Jak1 tyrosine kinase inhibitors, Jak3 tyrosine kinase inhibitors, LanC like protein 2 modulators, lipoxygenase modulators, acrophage mannose receptor 1 modulator, MAdCAM inhibitors, matrix metalloprotease inhibitors, melanocortin agonists, metalloprotease-9 inhibitors, NADPH oxidase inhibitor, natriuretic peptide receptor C agonists, NC-301, next-generation intestinal microbiota therapy, neuregulin-4 ligands, NKG2 D activating NK receptor antagonists, Non receptor tyrosine kinase TYK2 antagonist, opioid receptor antagonists, opioid receptor delta antagonists, oxidoreductase inhibitors, P2X7 purinoceptor agonists, PDE 4 inhibitors, phagocytosis stimulating peptide modulators, potassium channel inhibitors, PPAR alpha agonists, PPAR delta agonists, PPAR gamma agonists, protein fimH inhibitors, P-selectin glycoprotein ligand-1 inhibitors, RNA polymerase inhibitors, sphingosine 1 phosphate phosphatase 1 stimulators, sphingosine 1 phosphate phosphatase modulators, sphingosine-1-phosphate receptor-1 agonists, sphingosine-1-phosphate receptor-1 antagonists, sphingosine-1-phosphate receptor-1 modulators, sphingosine-1- phosphate receptor-5 modulators, STAT3 gene inhibitors, stem cell antigen-1 inhibitors, superoxide dismutase modulators, superoxide dismutase stimulators, SYK inhibitors, TGF beta 1 ligand inhibitors, thymulin agonists, TLR antagonists, TNF alpha ligand inhibitors, TNF antagonists, tumor necrosis factor 14 ligand modulators, type II TNF receptor modulators, Tpl 2 inhibitors, X box binding protein 1 stimulator, and Zonulin inhibitors. [0609] In some embodiments, the compounds provided herein, or pharmaceutically acceptable salts thereof, may be combined with one or more additional therapeutic agents selected from ABX-464, adalimumab; ALLO-ASC-CD, AMG-966, AMT-101, anakinra, apremilast; Alequel; ALV-304, AMG-139; amiselimod, anti-CXCR3 mAb, ASD-003, ASP- 3291, AX-1505, balsalazide; beclomethasone dipropionate; BI-655130, BMC-321, BMC-322, BMS-986184; BT-051, budesonide; CBX-111, CEQ-508; certolizumab; cibinetide, Clostridium butyricum; ChAdOx2-HAV, CU-06, CUG-252 dexamethasone sodium phosphate, DNVX-078, EB-7020, EM-101, etanercept; ENERGI-F704, ETX-201, golimumab; GS-4997, GS-5718, GS- 9876, GS-4875, GS-4059, infliximab; IMS-001, mesalazine, HLD-400, IBI-112, IMM-H013, KB-295, LFS-829, LYC-30937 EC; IONIS-JBI1-2.5Rx, JNJ-64304500, JNJ-66525433, JNJ- 4447, mesalamine, MET-642, MVA-HAV, naltrexone; natalizumab; neihulizumab, olsalazine; NOS-1244, NTG-A-009, PH-46-A, propionyl-L-carnitine; PTG-100; remestemcel-L; tacrolimus; teduglutide; tofacitinib; ASP-1002; ustekinumab; vedolizumab; AVX-470; INN- 108; SGM-1019; PF-06480605; PF-06651600; PR-600; RBX-8225, R-2187, RG-6287, SER- 287; TOP-1288; VBY-129; 99mTc-annexin V-128; bertilimumab; DLX-105; dolcanatide; quetmolimab (E-6011); FFP-104; filgotinib; foralumab; GED-0507-34-Levo; givinostat; GLPG- 0974; iberogast; ICP-330, JNJ-40346527; K(D)PT; KAG-308; KHK-4083; KRP-203; larazotide acetate; LY-3074828, midismase; olokizumab; OvaSave; P-28-GST; PF-547659; prednisolone; QBECO; RG-7835; RBX-2660, RO7049665, JKB-122; SYGN-313, SB-012; STNM-01; SZN- 1326, TJC-0434, Debio-0512; TRK-170; ABT-494; Ampion; BI-655066; carotegast methyl; cobitolimod; elafibranor; etrolizumab; GS-5745; HMPL-004; LP-02, ozanimod; peficitinib; QX- 004-N, RHB-104; SEFA-1024, tildrakizumab; TOP-1890, tralokinumab; brodalumab; laquinimod; and plecanatide; or a pharmaceutically acceptable salt of any of the foregoing; or any combination thereof. VII. Compound Preparation [0610] Some embodiments of the present disclosure are directed to processes and intermediates useful for preparing the compounds provided herein or pharmaceutically acceptable salts thereof. [0611] Compounds described herein can be purified by any of the means known in the art, including chromatographic means, such as high performance liquid chromatography (HPLC), preparative thin layer chromatography, flash column chromatography and ion exchange chromatography. Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins. Most typically the disclosed compounds are purified via silica gel and/or alumina chromatography. [0612] During any of the processes for preparation of the compounds provided herein, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups as described in standard works, such as T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis,” 4 th ed., Wiley, New York 2006. The protecting groups may be removed at a convenient subsequent stage using methods known from the art. [0613] Exemplary chemical entities useful in methods of the embodiments will now be described by reference to illustrative synthetic schemes for their general preparation herein and the specific examples that follow. Artisans will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be necessary or desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Furthermore, one of skill in the art will recognize that the transformations shown in the schemes below may be performed in any order that is compatible with the functionality of the particular pendant groups. Each of the reactions depicted in the general schemes is preferably run at a temperature from about 0 °C to the reflux temperature of the organic solvent used. [0614] The methods of the present disclosure generally provide a specific enantiomer or diastereomer as the desired product, although the stereochemistry of the enantiomer or diastereomer was not determined in all cases. When the stereochemistry of the specific stereocenter in the enantiomer or diastereomer is not determined, the compound is drawn without showing any stereochemistry at that specific stereocenter even though the compound can be substantially enantiomerically or disatereomerically pure. [0615] Representative syntheses of compounds of the present disclosure are described in the schemes below, and the particular examples that follow. [0616] Certain abbreviations and acronyms are used in describing the experimental details. Although a person of ordinary skill in the art will readily recognize and understand most of the abbreviations and acronyms, the below list provides many of the meanings of the abbreviations and acronyms. List of Abbreviations and Acronyms

General Synthetic Schemes [0617] General Reaction Schemes 1-14 are provided as further embodiments of the present disclosure and illustrate general methods which were used to prepare certain compounds of the present disclosure and which can be used to prepare additional compounds of the present disclosure. Each of the variables (e.g. R 1 , R 2 , R 3 , R 4 ) of the compounds disclosed in General Reaction Schemes I-V are as defined herein. [0618] The compounds of the present disclosure may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent to a skilled artisan given the disclosure herein and methods well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of typical compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aldrich or other chemical suppliers. In general, compounds described herein are typically stable and isolatable at room temperature and pressure. [0619] Typical embodiments of compounds disclosed herein may be synthesized using the general reaction schemes described below. It will be apparent to a skilled artisan given the description herein that the general schemes may be altered by substitution of the starting materials with other materials having similar structures to result in products that are correspondingly different. Descriptions of syntheses follow to provide numerous examples of how the starting materials may vary to provide corresponding products. Given a desired product for which the substituent groups are defined, the necessary starting materials generally may be determined by inspection. Starting materials are typically obtained from commercial sources or synthesized using published methods. For synthesizing compounds which are embodiments disclosed in the present disclosure, inspection of the structure of the compound to be synthesized will provide the identity of each substituent group. The identity of the final product will generally render apparent the identity of the necessary starting materials by a simple process of inspection, given the examples herein. [0620] The terms “solvent”, “inert organic solvent”, or “inert solvent” refer to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran (“THF”), dimethylformamide (“DMF”), chloroform, methylene chloride (or dichloromethane), diethyl ether, methanol, and the like). Unless specified to the contrary, the solvents used in the reactions of the present disclosure are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen or argon. General Reaction Scheme 1 [0621] Compounds of formula 1.2 may be assembled by reacting intermediate 1.1 with a halogenating conditions (e.g. I2 and AgOTf). Intermediate 1.2 can then be coupled to a suitable metallated coupling partner R 1 -M (where R 1 is aryl or heteroaryl, and M is -B, -Sn, -Zn, -Si, or -Mg) using a suitable palladium catalyst to deliver intermediate 1.3. Intermediate 1.3 can then be coupled to a boronated fluoropyridine coupling partner using a suitable palladium catalyst to deliver intermediate 1.4. Alternatively, Intermediate 1.3 can then be coupled to a boronated chloropyridine coupling partner using a suitable palladium catalyst to deliver intermediate 1.5. General Reaction Scheme 2 [0622] Compounds of formula (I-a) can be assembled by the combination of intermediate 1.4 with a suitable primary or secondary amine NH(R)(R), in the presence of heat and a suitable base (e.g., N,N-diisopropylethylamine or triethylamine). If the compound of formula (I-a) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-a) that contains a primary or secondary amine. If the compound of formula (I-a) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas), to reveal a compound of formula (I-a) that contains a primary or secondary amine. General Reaction Scheme 3 [0623] Compounds of formula (I-b) can be assembled by the combination of intermediate 1.3 with a coupling partner Y-M (e.g., Y is aryl, heteroaryl, alkyl, or cycloalkyl, and -M is -B, -Zn, -Sn, -Mg, or -Si) in the presence of an appropriate catalyst (e.g., palladium). If the compound of formula (I-b) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-b) that contains a primary or secondary amine. If the compound of formula (I-b) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H2 gas), to reveal a compound of formula (I-b) that contains a primary or secondary amine. General Reaction Scheme 4 [0624] Compounds of formula 1.3 can also be assembled by the combination of intermediate 1.5 with alkyne intermediate 1.6 in the presence of a metal catalyst (e.g., Rh) and copper salt (e.g., Cu(OPiv) 2 ). General Reaction Scheme 5 [0625] Compounds of formula 5.1 can be assembled by the combination of intermediate 1.1 with a coupling partner Y-M (e.g., Y is aryl, heteroaryl, alkyl, or cycloalkyl, and -M is -B, - Zn, -Sn, -Mg, or -Si) in the presence of an appropriate catalyst (e.g., palladium). If intermediate 5.1 contains an alkene, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas). Intermediate 5.1 may then be reacted with a halogenating reagent (e.g., NBS) to produce Intermediate 5.2. Intermediate 5.2 may then be reacted with a suitable metalating agent (e.g., B2Pin2, HBPin) in the presence of a metal catalyst (e.g., Pd) to furnish intermediate 5.3. Alternatively, Intermediate 5.1 may be reacted with a suitable nitrogen- protecting reagent (e.g., Boc anyhydride) to furnish Intermediate 5.4. Intermediate 5.4 may then be reacted with a suitable metalating reagent (e.g., isopropoxy-BPin) in the presence of a strong base (e.g., LDA) to furnish intermediate 5.5. General Reaction Scheme 6 [0626] Compounds of formula 1.3 may be reacted with a suitable nitrogen-protecting reagent (e.g., Boc anyhydride) to furnish Intermediate 6.1. Intermediate 6.1 may then be reacted with a suitable metalating agent (e.g., B2Pin2, HBPin) in the presence of a metal catalyst (e.g., Pd) to furnish intermediate 6.2. Compounds of formula (I-b) may be assembled by reacting intermediate 6.2 with a suitable coupling partner Y-X (where X is -I, -Br, -Cl, -OTf) using a suitable palladium catalyst to deliver compounds of formula (I-b). If the compound of formula (I-b) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-b) that contains a primary or secondary amine. If the compound of formula (I-b) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas), to reveal a compound of formula (I-b) that contains a primary or secondary amine. General Reaction Scheme 7 [0627] Compounds of formula 7.2 may be assembled by coupling Intermediate 1.3 with intermediate 7.1 in the presence of a suitable palladium catalyst. Intermediate 7.2 may then be reacted with a suitable base (e.g., LiOH) to furnish Intermediate 7.3. General Reaction Scheme 8 [0628] Compounds of formula (I-b) may be assembled by reacting intermediate 5.3 with a suitable coupling partner R 1 -X (where R 1 is aryl or heteroaryl, and X is -I, -Br, -Cl, -OTf) in the presence of a suitable palladium catalyst to deliver compounds of formula (I-b). Alternatively, compounds of formula (I-b) may be assembled by reacting intermediate 5.5 with a suitable coupling partner R 1 -X (where R 1 is aryl or heteroaryl, and X is -I, -Br, -Cl, -OTf) using a suitable palladium catalyst to deliver compounds of formula (I-b). If the compound of formula (I-b) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-b) that contains a primary or secondary amine. If the compound of formula (I-b) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas), to reveal a compound of formula (I-b) that contains a primary or secondary amine. General Reaction Scheme 9 [0629] Compounds of formula (I-b) may be assembled by reacting intermediate 5.2 with a suitable metallated coupling partner R 1 -M (where R 1 is aryl or heteroaryl, and M is -B, -Sn, - Zn, -Si, or -Mg) using a suitable palladium catalyst to deliver compounds of formula (I-b). If the compound of formula (I-b) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-b) that contains a primary or secondary amine. If the compound of formula (I-b) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas) to reveal a compound of formula (I-b) that contains a primary or secondary amine. General Reaction Scheme 10 [0630] Compounds of formula (I-c) can be assembled by the combination of intermediate 7.3 with a suitable primary or secondary amine NH(R)(R), in the presence of a suitable peptide coupling reagent (e.g., HATU, TCFH, EDC) and a suitable base (e.g., N,N- diisopropylethylamine, triethylamine). If the compound of formula (I-c) contains a tert- butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-c) that contains a primary or secondary amine. If the compound of formula (I-c) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H2 gas) to reveal a compound of formula (I-c) that contains a primary or secondary amine. General Reaction Scheme 11 [0631] Compounds of formula (I-e) can be assembled by the combination of compounds of formula (I-d) with a suitable carboxylic acid 11.1, in the presence of a suitable peptide coupling reagent (e.g., HATU, TCFH, EDC) and a suitable base (e.g., N,N- diisopropylethylamine, triethylamine). If the compound of formula (I-e) contains a tert- butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-e) that contains a primary or secondary amine. If the compound of the formula (I-e) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H2 gas) to reveal a compound of formula (I-e) that contains a primary or secondary amine. General Reaction Scheme 12 [0632] Compounds of formula (I-f) can be assembled by the combination of compounds of formula (I-d) with a suitable aldehyde 12.1, in the presence of a suitable reducing reagent (e.g., NaBH4, Na(OAc) 3 BH, Na(CN) 3 BH). If the compound of formula (I-f) contains a tert- butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-f) that contains a primary or secondary amine. If the compound of the formula (I-f) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas) to reveal a compound of formula (I-f) that contains a primary or secondary amine. General Reaction Scheme 13 [0633] Compounds of formula (I-g) can be assembled by the combination of compounds of formula (I-d) with an intermediate 13.1, where X is a leaving group (e.g. -Cl, -Br, -I, OTs, - OMs), in the presence of a base (e.g., N,N-diisopropylethylamine, triethylamine, K 2 CO 3 , CsCO3). If the compound of the formula (I-g) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-g) that contains a primary or secondary amine. If the compound of the formula (I-g) contains a benzyl-amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H2 gas) to reveal a compound of formula (I-g) that contains a primary or secondary amine. General Reaction Scheme 14 [0634] Compounds of formula (I-h) can be assembled by the combination of intermediate 1.5 with a suitable metallated coupling partner R-M (where R is aryl, heteroaryl, alkenyl, and M is -B, -Sn, -Zn, -Si, or -Mg) using a suitable palladium catalyst to deliver compounds of formula (I-h). If the compound of formula (I-h) contains an alkene, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas). If the compound of formula (I-h) contains a tert-butylcarbamate functional group, this can be subsequently removed by treatment with acid (e.g., trifluoroacetic acid or hydrochloric acid) to reveal a compound of formula (I-h) that contains a primary or secondary amine. If the compound of formula (I-h) contains a benzyl- amine functional group, this can be subsequently removed (e.g., using a metal catalyst and H 2 gas) to reveal a compound of formula (I-h) that contains a primary or secondary amine. VIII. Examples [0635] Exemplary chemical entities of the present disclosure are provided in the specific examples that follow. Those skilled in the art will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be necessary or desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Furthermore, one of skill in the art will recognize that the transformations shown in the schemes below may be performed in any order that is compatible with the functionality of the particular pendant groups. [0636] The Examples provided herein describe the synthesis of compounds disclosed herein as well as intermediates used to prepare the compounds. It is to be understood that individual steps described herein may be combined. It is also to be understood that separate batches of a compound may be combined and then carried forth in the next synthetic step. [0637] In the following description of the Examples, specific embodiments are described. These embodiments are described in sufficient detail to enable those skilled in the art to practice certain embodiments of the present disclosure. Other embodiments may be utilized and logical and other changes may be made without departing from the scope of the disclosure. The following description is, therefore, not intended to limit the scope of the present disclosure. Intermediates Preparation of Intermediate I-1 [0638] 6-bromo-2-iodo-3-methyl-1H-indole: To a solution of 6-bromo-3-methyl-1H- indole (2.00 g, 9.52 mmol) in THF (40 mL) was added iodine (2.42 g, 9.52 mmol) and silver trifluoromethanesulfonate (2.94 g, 11.4 mmol) and the reaction mixture was stirred for 30 min at room temperature. The reaction was quenched by the addition of 10% aqueous thiosulfate and the mixture was extracted with EtOAc (3x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude residue was purified by column chromatography (0-30% EtOAc in hexane) to give I-1 which was carried forward to the next step. ES/MS: 337.1 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 7.93 (s, 1H), 7.46 (d, J = 1.7 Hz, 1H), 7.37 (d, J = 8.4 Hz, 1H), 7.22 (dd, J = 8.4, 1.7 Hz, 1H), 2.28 (s, 3H). Preparation of Intermediate I-2 [0639] 6-bromo-2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indole: To a vial was added 6-bromo-2-iodo-3-methyl-1H-indole (I-1) (500 mg, 1.49 mmol), 2,6-dimethyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (0.382 g, 1.64 mmol) and (1,1'- bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (0.0842 g, 0.119 mmol) followed by acetonitrile (10.00 mL) and 1.0 M potassium acetate in water (1.00 M, 3.72 mL, 3.72 mmol). Argon was bubbled through the mixture for 3 min and the reaction was heated to 100 °C in a microwave for 20 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The mixture was azeotroped with water and Toluene (1x). The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give I-2. ES/MS: 317.1 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 8.32 (s, 1H), 7.54 (d, J = 1.6 Hz, 1H), 7.49 (d, J = 8.4 Hz, 1H), 7.30 – 7.24 (m, 1H), 7.17 (s, 2H), 2.61 (s, 6H), 2.50 (s, 3H). Preparation of Intermediate I-3 [0640] 2-(2,6-dimethylpyridin-4-yl)-6-(6-fluoropyridin-3-yl)-3-meth yl-1H-indole: To a vial with 6-bromo-2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indole (I-2) (70 mg, 0.22 mmol), 2-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri dine (99 mg, 0.44 mmol), and [1,1′- Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (16.5 mg, 0.022 mmol) was added DME (1 mL) and 2M aqueous sodium carbonate (0.22 mL, 0.44 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 100 °C for 2 hours. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product I-3. ES/MS: 332.2 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 8.51 (d, J = 2.6 Hz, 1H), 8.49 – 8.37 (m, 1H), 8.09 – 8.03 (m, 1H), 7.73 (d, J = 8.3 Hz, 1H), 7.56 (d, J = 1.4 Hz, 1H), 7.36 (dd, J = 8.3, 1.6 Hz, 1H), 7.24 (s, 2H), 7.04 (dt, J = 8.4, 4.2 Hz, 1H), 2.65 (s, 6H), 2.57 (s, 3H). Preparation of Intermediate I-4 [0641] 6-bromo-2-iodo-3-ethyl-1H-indole (I-4): Prepared analogously to I-1 starting with 6-bromo-3-ethyl-1H-indole to give the title compound. ES/MS: 349.1 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 7.89 (s, 1H), 7.45 (d, J = 1.7 Hz, 1H), 7.40 (dd, J = 8.5, 0.7 Hz, 1H), 7.19 (dd, J = 8.4, 1.7 Hz, 1H), 2.69 (q, J = 7.5 Hz, 2H), 1.20 (t, J = 7.6 Hz, 4H). Preparation of Intermediate I-5 [0642] 6-bromo-2-iodo-3-isopropyl-1H-indole (I-5): Prepared analogously to I-1 starting with 6-bromo-3-isopropyl-1H-indole to give the title compound. ES/MS: 365.0 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 7.88 (s, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.47 (d, J = 1.7 Hz, 1H), 7.18 (dd, J = 8.5, 1.8 Hz, 1H), 3.12 (hept, J = 7.1 Hz, 1H), 1.42 (d, J = 7.1 Hz, 6H). Preparation of Intermediate I-6 [0643] 2-(2,6-dimethylpyridin-4-yl)-6-(2-fluoropyridin-4-yl)-3-meth yl-1H-indole (I- 6): To a vial was added 6-bromo-2-iodo-3-methyl-1H-indole (I-2) (205 mg, 0.61 mmol), 2,6- dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri dine (156 mg, 0.67 mmol) and (1,1'-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (34.5 mg, 0.049 mmol) followed by acetonitrile (8.0 mL) and 1.0 M potassium acetate in water (1.53 mL, 1.53 mmol). Argon was bubbled through the mixture for 4 min and the reaction was heated to 100 °C in a microwave for 20 min. 2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri dine (163 mg, 0.73 mmol), Pd(AmPhos) 2 Cl2 (43.2 mg, 0.061 mmol) and sodium carbonate (0.114 g, 1.83 mmol) were added, nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 332.3 (M+H + ). Preparation of Intermediate I-7 [0644] 4-(cyclopropylethynyl)-2,6-dimethylpyridine (I-7): To a vial was added 4- bromo-2,6-dimethyl-pyridine (300 mg, 1.61 mmol), bis(triphenylphosphine)palladium chloride (113 mg, 0.16 mmol) and copper(I)iodide (31 mg, 0.16 mmol). The vial was sparged with argon and acetonitrile (20 mL) was added followed by ethynylcyclopropane (0.68 mL, 8.1 mmol) and triethylamine (1.1 mL, 8.1 mmol) and the reaction mixture was stirred for 1 h at room temperature then heated to 60 °C for 1 h. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 172.2 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 6.98 (s, 2H), 2.56 (s, 6H), 1.53 – 1.39 (m, 1H), 0.99 – 0.90 (m, 2H), 0.90 – 0.78 (m, 2H). Preparation of Intermediate I-8 [0645] (E)-1-((3-bromophenyl)diazenyl)pyrrolidine (I-8): To a solution of 4- bromoaniline (989 mg, 5.75 mmol) in acetonitrile (5.0 mL) and water (2.5 mL), cooled to 0 °C, was added hydrochloric acid (11.7 mol/L, 2.0 mL, 23 mmol) dropwise. A solution of sodium nitrite (595 mg, 8.63 mmol) in water (7.5 mL) was then added dropwise and then the reaction was stirred at 0 °C for 30 min. This solution was then added dropwise to a stirred solution of pyrrolidine (1.2 mL, 14.4 mmol) and potassium carbonate (4.13 g, 30 mmol) in acetonitrile (20 mL) and water (10 mL) which was also cooled to 0 °C via cannula. The reaction mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was extracted with EtOAc (3x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-10% EtOAc in hexane) to give the title compound. ES/MS: 256.1 (M+H + ). Preparation of Intermediate I-9 [0646] 6-bromo-3-cyclopropyl-2-(2,6-dimethylpyridin-4-yl)-1H-indole (I-9): To a vial was added (E)-1-((3-bromophenyl)diazenyl)pyrrolidine (76.2 mg, 0.30 mmol), [RhCp*(CH 3 CN) 3 ](SbF6) 2 (50 mg, 0.060 mmol) and copper(II) pivalate (159 mg, 0.60 mmol). 4-(2-cyclopropylethynyl)-2,6-dimethyl-pyridine (56.5 mg, 0.33 mmol) was then added followed by methanol (1.50 mL) and tert-amyl alcohol (1.50 mL) and the solution was heated to 80 °C for 16 h. The reaction was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The residue was then diluted with saturated aqueous sodium bicarbonate and the mixture was extracted with DCM (3x). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound (major isomer). ES/MS: 341.3 (M+H + ). Preparation of Intermediate I-10 [0647] tert-butyl 4-(5-(3-methyl-1H-indol-6-yl)pyridin-2-yl)piperazine-1- carboxylate: To a vial was added 6-bromo-3-methyl-1H-indole (400 mg, 1.90 mmol), tert-butyl 4-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl ]piperazine-1-carboxylate (890 mg, 2.28 mmol) and Pd(AmPhos) 2 Cl 2 (0.108 g, 0.15 mmol) followed by acetonitrile (2.0 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (4.8 mL, 4.8 mmol / 7.2 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 130 °C in a microwave for 30 min. The reaction mixture was filtered through celite, eluting with DCM, water was added to the filtrate, the layers were separated, and the aqueous layer was back extracted with DCM (2x). The combined organic layers were combined, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 393.3 (M+H + ). [0648] tert-butyl 4-(5-(2-bromo-3-methyl-1H-indol-6-yl)pyridin-2-yl)piperazine -1- carboxylate (I-10): To a solution of tert-butyl 4-(5-(3-methyl-1H-indol-6-yl)pyridin-2- yl)piperazine-1-carboxylate (522 mg, 1.33 mmol) in THF (3.0 mL) and DCM (3.0mL) cooled to 0 °C was added pyridinium tribromide (468 mg, 1.46 mmol) and the reaction mixture was stirred for 10 min at 0 °C. The reaction was diluted with DCM and quenched by the addition of 10% aqueous thiosulfate and the layers separated. The aqueous layer was extracted with DCM (2x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-50% EtOAc in hexane) to give the title compound. ES/MS: 471.1 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.61 (s, 1H), 8.47 – 8.43 (m, 1H), 7.91 – 7.84 (m, 1H), 7.54 – 7.48 (m, 1H), 7.43 (s, 1H), 7.31 – 7.25 (m, 1H), 7.01 – 6.84 (m, 1H), 3.58 – 3.49 (m, 4H), 3.49 – 3.40 (m, 4H), 2.20 (s, 3H), 1.44 (s, 9H). Preparation of Intermediate I-11 [0649] tert-butyl 4-(5-(3-methyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)-1H- indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate (I-11): To a vial was added tert-butyl 4-(5- (3-methyl-1H-indol-6-yl)pyridin-2-yl)piperazine-1-carboxylat e (I-10) (200 mg, 0.424 mmol), Bis(pinacolato)diboron (140 mg, 0.55 mmol), PdCl 2 (dppf) (25.2 mg, 0.034 mmol) and potassium propionate (143 mg, 1.27 mmol). 1,4-Dioxane (3.6 mL) was added, nitrogen was bubbled through the reaction mixture for 4 min, and the mixture was heated to 100 °C for 1 h. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give crude I-11. ES/MS: 519.3 (M+H + ). Preparation of Intermediate I-12 [0650] tert-butyl 4-(5-(3-cyano-1H-indol-6-yl)pyridin-2-yl)piperazine-1-carbox ylate (I-12): To a vial was added 6-bromo-1H-indole-3-carbonitrile (421 mg, 1.90 mmol), tert-butyl 4-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl ]piperazine-1-carboxylate (890 mg, 2.28 mmol) and Pd(AmPhos) 2 Cl 2 (108 mg, 0.15 mmol) followed by acetonitrile (12 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (4.8 mL, 4.8 mmol / 7.1 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 130 °C in a microwave for 30 min. The reaction mixture was diluted with water to precipitate product. The crude mixture was filtered and the precipitate was washed with water, dissolved in DCM, and filtered through celite. The filtrate was concentrated in vacuo to give the crude product which was used directly without further purification. ES/MS: 404.3 (M+H + ). Preparation of Intermediate I-13 [0651] tert-butyl 4-(5-(3-acetyl-1H-indol-6-yl)pyridin-2-yl)piperazine-1-carbo xylate (I-13): Prepared analogously to I-12 starting with 1-(6-bromo-1H-indol-3-yl)ethan-1-one to give the title compound. ES/MS: 421.2 (M+H + ). 1 H NMR (400 MHz, Chloroform-d) δ 7.88 (s, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.47 (d, J = 1.7 Hz, 1H), 7.18 (dd, J = 8.5, 1.8 Hz, 1H), 3.12 (hept, J = 7.1 Hz, 1H), 1.42 (d, J = 7.1 Hz, 6H). Preparation of Intermediate I-14 [0652] tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin- 2-yl)piperazine-1-carboxylate (I-14): To a vial was added 6-bromo-2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indole (750 mg, 2.38 mmol), tert-butyl 4-[5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2-pyridyl]piperazine-1-carboxylate (1.11 g, 2.86 mmol) and Pd(AmPhos) 2 Cl2 (168 mg, 0.24 mmol) followed by acetonitrile (12.0 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (6.0 mL, 6.0 mmol / 9.0 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 20 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 498.3 (M+H + ). Preparation of Intermediate I-15 [0653] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(6-(piperazin-1-yl)p yridin-3-yl)-1H- indole hydrocloride (I-15): To a solution of tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3- methyl-1H-indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate (I-14) (300 mg, 0.60 mmol) in DCM (5.0 mL) was added 4.0 N HCl in dioxane (1.5 mL, 6.03 mmol) and the reaction mixture was stirred for 30 min at room temperature. The reaction mixture was concentrated in vacuo to give the title compound, which was used as crude in subsequent reactions. ES/MS: 389.2 (M+H + ). Preparation of Intermediate I-16 [0654] tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3-met hyl- 1H-indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate: To a solution of tert-butyl 4-[5-[2-(2,6- dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-pyridyl]pipera zine-1-carboxylate (I-14) (300 mg, 0.60 mmol) in DMF (5 mL) cooled to 0 °C was added 60% sodium hydride in mineral oil (26.5 mg, 0.66 mmol) and the reaction was stirred at 0 °C for 5 min. 4-Methoxybenzylchloride (0.090 mL, 0.66 mmol) was then added and the reaction mixture was stirred at 0 °C for 10 min and then warmed to room temperature for 1 h. The reaction was quenched by the addition of water and the mixture was extracted with EtOAc. The organic layer was washed with water, 10% aqueous LiCl, and brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 618.4 (M+H + ). [0655] 2-(2,6-dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3-methyl-6- (6-(piperazin- 1-yl)pyridin-3-yl)-1H-indole hydrochloride (I-16): To a solution of tert-butyl 4-(5-(2-(2,6- dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3-methyl-1H-indol- 6-yl)pyridin-2-yl)piperazine-1- carboxylate (200 mg, 0.32 mmol) in DCM (5.0 mL) was added 4.0 N HCl in dioxane (0.81 mL, 3.24 mmol) and the reaction mixture was stirred for 30 min at room temperature. The reaction mixture was concentrated in vacuo to give the title compound, used as crude in subsequent reactions. ES/MS: 418.2 (M+H + ). Preparation of Intermediate I-17 [0656] methyl 3-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)benzo ate: To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (200 mg, 0.635 mmol), methyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoate (200 mg, 0.76 mmol) and bis(di-tert-butyl(4-dimethylaminophenyl)phosphine)dichloropa lladium(II) (67.4 mg, 0.095 mmol) followed by acetonitrile (10 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (1.6 mL, 1.6 mmol / 2.4 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 120 °C in a microwave for 30 min. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 371.2 (M+H + ). [0657] 3-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)benzo ic acid (I-17): To a solution of methyl 3-[6-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-2-yl]benzoat e (169 mg, 0.46 mmol) in acetonitrile (1.00 mL) and water (0.330 mL) was added lithium hydroxide monohydrate (57.4 mg, 1.37 mmol) and the reaction mixture was stirred for 90 min at 80 °C. The reaction was quenched by the addition of several drops of concentrated HCl, neutralized by stirring with MP-carbonate, filtered and the filtrate was concentrated in vacuo to give the crude product which was used directly in subsequent reactions. ES/MS: 357.2 (M+H + ). Preparation of Intermediate I-18 [0658] methyl 4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)benzo ate: To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (200 mg, 0.635 mmol), methyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoate (0.200 g, 0.761 mmol) and bis(di-tert-butyl(4-dimethylaminophenyl)phosphine)dichloropa lladium(II) (0.0674 g, 0.0952 mmol) followed by acetonitrile (10 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (1.6 mL, 1.6 mmol / 2.4 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 120 °C in a microwave for 30 min. LCMS showed conversion to desired product. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 371.2 (M+H + ). [0659] 4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)benzo ic acid (I-18): To a solution of methyl 4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)benzo ate (157 mg, 0.42 mmol) in acetonitrile (3.00 mL) and water (0.330 mL) was added lithium hydroxide monohydrate (53.4 mg, 1.27 mmol) and the reaction mixture was stirred for 90 min at 50 °C. The reaction was quenched by the addition of several drops of concentrated HCl, neutralized by stirring with MP-carbonate, filtered and the filtrate was concentrated in vacuo to give the crude product which was used directly in subsequent reactions. ES/MS: 357.2 (M+H + ). Preparation of Intermediate I-19 [0660] 3-cyclopropyl-2-(2,6-dimethylpyridin-4-yl)-6-(6-fluoropyridi n-3-yl)-1H- indole (I-19): To a vial was added 6-bromo-3-cyclopropyl-2-(2,6-dimethyl-4-pyridyl)-1H- indole (56.7 mg, 0.17 mmol), 6-fluoro-3-pyridyl)boronic acid (28.1 mg, 0.20 mmol) and bis(di- tert-butyl(4-dimethylaminophenyl)phosphine)dichloropalladium (II) (11.8 mg, 0.017 mmol) followed by acetonitrile (1.0 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.11 mL, 0.11 mmol / 0.16 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 20 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 358.2 (M+H + ). Preparation of Intermediate I-20 [0661] methyl 5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)picol inate: To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (100 mg, 0.32 mmol), methyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-ca rboxylate (100 mg, 0.38 mmol) and bis(di-tert-butyl(4-dimethylaminophenyl)phosphine)dichloropa lladium(II) (33.7 mg, 0.048 mmol) followed by acetonitrile (3.5 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.8 mL, 0.8 mmol / 1.2 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 120 °C in a microwave for 30 min. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 372.2 (M+H + ). [0662] 5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)picol inic acid (I-20): To a solution of methyl 5-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]pyridin e-2- carboxylate (20.3 mg, 0.055 mmol) in acetonitrile (3.00 mL) and water (0.330 mL) was added lithium hydroxide monohydrate (6.9 mg, 0.16 mmol) and the reaction mixture was stirred for 90 min at 50 °C. The reaction was quenched by the addition of several drops of concentrated HCl, neutralized with MP-Carbonate, filtered and the filtrate was concentrated in vacuo to give the crude product which was used directly in subsequent reactions. ES/MS: 358.2 (M+H + ). Preparation of Intermediate I-21 [0663] tert-butyl (R)-(1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6- yl)pyridin-2-yl)pyrrolidin-3-yl)carbamate: To a solution of 2-(2,6-dimethyl-4-pyridyl)-6-(6- fluoro-3-pyridyl)-3-methyl-1H-indole (I-3) (30.0 mg, 0.091 mmol) in 2-methoxyethanol (1.0 mL) was added N,N-diisopropylethylamine (0.16 mL, 0.915 mmol) and tert-butyl N-[(3R)- pyrrolidin-3-yl]carbamate (0.169 g, 0.91 mmol) and the reaction mixture was heated in a microwave to 150 °C for 4 h. The reaction mixture was concentrated in vacuo and the crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 498.3 (M+H + ). [0664] (R)-1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-y l)pyridin-2- yl)pyrrolidin-3-amine hydrochloride (I-21): To a solution of tert-butyl (R)-(1-(5-(2-(2,6- dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl)py rrolidin-3-yl)carbamate (24.8 mg, 0.050 mmol) in DCM (2.0 mL) was added 4.0 N HCl in dioxane (0.13 mL, 0.50 mmol) and the reaction mixture was stirred for 2 h at room temperature. LCMS showed complete conversion to desired product. The reaction mixture was concentrated in vacuo to give the title compound as an HCl salt. ES/MS: 398.2 (M+H + ). Preparation of Intermediate I-22 [0665] tert-butyl (1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6- yl)pyridin-2-yl)piperidin-4-yl)carbamate: To a solution of 2-(2,6-dimethyl-4-pyridyl)-6-(6- fluoro-3-pyridyl)-3-methyl-1H-indole (I-3) (30.0 mg, 0.091 mmol) in 2-methoxyethanol (1.0 mL) was added N,N-diisopropylethylamine (0.158 mL, 0.91 mmol) and tert-butyl N-(4- piperidyl)carbamate (0.181 g, 0.91 mmol) and the reaction mixture was heated in a microwave to 165 °C for 5 h. The reaction mixture was concentrated in vacuo and the crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 512.4 (M+H + ). [0666] 1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin-2- yl)piperidin-4-amine hydrochloride (I-22): To a solution of tert-butyl (1-(5-(2-(2,6- dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl)pi peridin-4-yl)carbamate (22.4 mg, 0.044 mmol) in DCM (2.0 mL) was added 4.0 N HCl in dioxane (0.11 mL, 0.44 mmol) and the reaction mixture was stirred for 2 h at room temperature. The reaction mixture was concentrated in vacuo to give the title compound as an HCl salt. ES/MS: 412.2 (M+H + ). Preparation of Intermediate I-23 [0667] tert-butyl 4-(5-(3-methyl-1H-indol-6-yl)pyridin-2-yl)piperazine-1- carboxylate: To a vial was added 6-bromo-3-methyl-1H-indole (2.00 g, 9.52 mmol), tert-butyl 4-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl ]piperazine-1-carboxylate (4.08 g, 10.5 mmol) and Pd(AmPhos) 2 Cl 2 (0.539 g, 0.76 mmol) followed by acetonitrile (60 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (24 mL, 24 mmol / 36 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 100 °C in for 1 h. The reaction was diluted with water to precipitate product. The crude mixture was filtered, the precipitate was washed with water followed by acetonitrile (3x), dissolved in DCM, and filtered through celite. The filtrate was concentrated in vacuo to give the crude product which was used directly in subsequent reactions. ES/MS: 393.3 (M+H + ). [0668] tert-butyl 6-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)pyridin-3-yl)-3- methyl-1H-indole-1-carboxylate: To a solution of tert-butyl 4-[5-(3-methyl-1H-indol-6-yl)-2- pyridyl]piperazine-1-carboxylate (1.00 g, 2.55 mmol) and 4-dimethylaminopyridine (6.2 mg, 0.51 mmol) in THF (10 mL) was added di-tert-butyl dicarbonate (834 mg, 3.82 mmol) followed by triethylamine (1.1 mL, 7.64 mmol) and the reaction mixture was stirred at room temperature for 30 min. The reaction was quenched by the addition of saturated aqueous ammonium chloride and the mixture was extracted with DCM (3x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-50% EtOAc in hexane) to give the title compound. ES/MS: 493.2 (M+H + ). [0669] tert-butyl 6-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)pyridin-3-yl)-3- methyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-in dole-1-carboxylate (I-23): To a solution of tert-butyl 6-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)pyridin-3-yl)-3- methyl-1H- indole-1-carboxylate (500 mg, 1.02 mmol) and 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (0.31 mL, 1.52 mmol) in THF (10.0 mL) cooled to -78 °C was added a freshly prepared solution of LDA cooled to -78 °C [made from diisopropylamine (0.17 mL, 1.22 mmol) and N-butyllithium (2.5 M in hexanes) (0.49 mL, 1.22 mmol) in THF (3 mL)] dropwise via cannula and the reaction mixture was stirred at -78 °C for 30 min, then 0 °C for 30 min. The reaction was quenched by the addition of saturated aqueous ammonium chloride and the mixture was extracted with EtOAc (3x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 537.9 (M+H + ). Preparation of Intermediate I-24 [0670] tert-butyl 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-indole-1-carboxy late (I-24): To a 100 mL flask was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (1.50 g, 4.76 mmol), triethylamine (1.33 ml, 9.52 mmol), tert-butoxycarbonyl tert-butyl carbonate (1.56 g, 7.14 mmol), and 4-dimethylaminopyridine (0.70 g, 5.71 mmol), and the mixture was dissolved in THF (10 mL). The solution was stirred overnight at room temperature. The reaction mixture was concentrated under reduced pressure. The crude residue was dissolved in dichloromethane (50 mL) and washed with sat. aq. sodium bicarbonate (20 mL). The phases were separated, and the aqueous layer was extracted once with dichloromethane (20 mL). The combined organic layers were washed with sat. aq. NH 4 Cl (15 mL), brine (15 mL), and then the organic layer was dried over MgSO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 415.3 (M+H + ). Preparation of Intermediate I-25 [0671] tert-butyl 2-(2,6-dimethyl-4-pyridyl)-3-methyl-6-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)indole-1-carboxylate (I-25): To a 100 mL flask was added tert-butyl 6- bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-indole-1-carboxyla te (I-24) (800 mg, 1.93 mmol), Bis(pinacolato)diboron (1.23 g, 4.82 mmol), [1,1′- Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (286 mg, 0.39 mmol), and potassium propanoate (1,3 g, 11.6 mmol), and the mixture was dissolved in dioxane (40 mL). The solution was heated at 100 °C for 3 hours. The reaction mixture was concentrated under reduced pressure. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 463.3 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 8.63 (t, J = 0.9 Hz, 1H), 7.68 (dd, J = 7.8, 1.0 Hz, 1H), 7.58 (dd, J = 7.8, 0.8 Hz, 1H), 7.13 (s, 2H), 2.59 (s, 6H), 2.17 (s, 3H), 1.40 (s, 12H), 1.30 (s, 9H), 1.26 (s, 7H), 1.21 (s, 8H). Preparation of Intermediate I-26 [0672] 1-methyl-2-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)pyridine-3- carbonitrile (I-26): To a 50 mL flask was added 5-bromo-1-methyl-2-oxo-pyridine-3- carbonitrile (60 mg, 0.282 mmol), bis(pinacolato)diboron (78.7 g, 0.310 mmol), [1,1′- bis(diphenylphosphino)ferrocene]dichloropalladium(II) (20.9 mg, 0.0282 mmol), and potassium propanoate (79 mg, 11.6 mmol), and the mixture was dissolved in dioxane (3 mL). The solution was heated at 100 °C for 3 hours. The reaction mixture was concentrated under reduced pressure. The reaction mixture was used directly in the next step. ES/MS: 266.6 (M+H + ). Preparation of Intermediate I-27 [0673] 6-(6-chloropyridin-3-yl)-2-(2,6-dimethylpyridin-4-yl)-3-meth yl-1H-indole (I- 27): To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (250 mg, 0.79 mmol), 2-chloro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri dine (199 mg, 0.83 mmol) and (1,1'-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (44.9 mg, 0.064 mmol) followed by acetonitrile (10.0 mL) and 1.0 M potassium acetate in water (2.0 mL, 2.0 mmol / 3.0 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 120 °C in a microwave for 30 min. The reaction mixture was diluted with DCM, dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 348.2 (M+H + ). Preparation of Intermediate I-28 [0674] tert-butyl 5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-3',6 '- dihydro-[2,4'-bipyridine]-1'(2'H)-carboxylate (I-28): To a vial was added 6-(6- chloropyridin-3-yl)-2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H -indole (I-27) (100 mg, 0.29 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro- 2H-pyridine-1- carboxylate (107 mg, 0.35 mmol) and bis(di-tert-butyl(4- dimethylaminophenyl)phosphine)dichloropalladium(II) (30.5 mg, 0.043 mmol) followed by acetonitrile (3.0 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.72 mL, 0.72 mmol / 1.1 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 120 °C in a microwave for 30 min. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 495.3 (M+H + ). Preparation of Intermediate I-29: [0675] 5-(cyclopropylethynyl)-1,3-dimethylpyridin-2(1H)-one (I-29): Prepared analogously to I-7, substituting 4-bromo-2,6-dimethyl-pyridine with 5-bromo-1,3- dimethylpyridin-2(1H)-one. ES/MS: 188.2 (M+H + ). Preparation of Intermediate I-30: [0676] 6- 5-(6-bromo-3-cyclopropyl-1H-indol-2-yl)-1,3-dimethylpyridin- 2(1H)-one (I-30): Prepared analogously to I-9, substituting I-7 with I-29. ES/MS: 357.1 (M+H + ). Preparation of Intermediate I-31: [0677] (6-bromo-3-cyclopropyl-1H-indol-2-yl)-trimethyl-silane: To a round-bottomed flask was added 5-bromo-2-iodo-aniline (4.00 g, 13.4 mmol), Bis(triphenylphosphine)palladium dichloride (0.942 g, 1.34 mmol) and sodium carbonate (4.27 g, 40.3 mmol) followed by DMF (60 mL) and argon was bubbled through the solution for 4 minutes, before 2- cyclopropylethynyl(trimethyl)silane (3.71 mL, 22.8 mmol) was added and the reaction mixture was heated to 80 °C for 16 h. The reaction mixture was filtered through celite, rinsing with EtOAc, and the filtrate was washed with brine (3x), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude residue was purified by column chromatography (eluent: EtOAc in hexane) to give the title compound. ES/MS: 309.1 (M+H + ). 1H NMR (400 MHz, DMSO-d6) δ 10.65 (s, 1H), 7.52 – 7.46 (m, 2H), 7.06 (dd, J = 8.4, 1.8 Hz, 1H), 1.94 – 1.82 (m, 1H), 0.96 – 0.88 (m, 2H), 0.68 – 0.61 (m, 2H), 0.39 (s, 9H). [0678] 6-bromo-3-cyclopropyl-1H-indole (I-31): To a solution of (6-bromo-3- cyclopropyl-1H-indol-2-yl)-trimethyl-silane (3.28 g, 10.6 mmol) in THF (32.0 mL) was added tetrabutylammonium fluoride (TBAF) (1M in THF, 32 mL, 32.0 mmol) and the reaction mixture was heated to 70 °C for 3 h. The reaction mixture was concentrated under reduced pressure and the crude residue was purified by column chromatography (eluent: EtOAc in hexane) to give the title compound. ES/MS: 237.2 (M+H + ) Preparation of Intermediate I-32: [0679] 6-bromo-3-cyclopropyl-2-iodo-1H-indole (I-32): Prepared analogously to I-1, substituting 6-bromo-3-methyl-1H-indole with 6-bromo-3-cyclopropyl-1H-indole (I-31). Preparation of Intermediate I-33: [0680] 6-(6-bromo-3-methyl-1H-indol-2-yl)-8-methyl-[1,2,4]triazolo[ 1,5-a]pyridine (I-33): Prepared analogously to I-2, substituting 2,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridine with 8-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- [1,2,4]triazolo[1,5-a]pyridine. ES/MS: 341.1 (M + ). Preparation of Intermediate I-34: [0681] methyl 2-bromo-3-methyl-1H-indole-6-carboxylate: To a solution of methyl 3-methyl-1H-indole-6-carboxylate (2.5g, 13.4 mmol) in AcOH (26.5 ml) was added NBS (2.5 g, 13.4 mmol) and the resulting mixture stirred at room temperature for 1 h. Water (50 ml) was added and the resulting mixture neutralized through the dropwise addition of aq. NaOH (1.0 M). The precipitate that formed was filtered and dried over air to provide the product. ES/MS: 269.2 (M+H + ). [0682] methyl 2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole-6-carboxylate: To a vial was added methyl 2-bromo-3-methyl-1H-indole-6-carboxylate (2.1 g, 7.7 mmol), 2,6- dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri dine (2.1 g, 9.2 mmol), XPhos Pd G2 (300 mg, 0.38 mmol), cesium carbonate (6.2 g, 19.1 mmol), and 10:1 mixture of 1,4-dioxane and water (240 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc/ Hex) to provide the desired product. ES/MS: 295.2 (M+H + ). [0683] 2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole-6-carboxylic acid (I-34): To a solution of methyl 2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole-6-carboxylate (1300 mg, 4.4 mmol) in THF (12 mL), MeOH (12 ml), and water (12 mL) was added lithium hydroxide monohydrate (317 mg, 13.2 mmol) and the reaction mixture was stirred for 120 min at 80 °C. The crude mixture was concentrated under reduced pressure, and 1N HCl (4 mL) was added to the resulting slurry. The precipitate was filtered, washed with water, and dried under vacuum to provide the product. ES/MS: 281.3 (M+H + ). Preparation of Intermediate I-35: [0684] 3-methyl-2-(8-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)-1H- indole-6- carboxylic acid (I-35): Prepared analogously to I-34, substituting 2,6-dimethyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine with 8-methyl-6-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-[1,2,4]triazolo[1,5-a]pyridine. ES/MS: 307.1 (M+H + ). Final Procedures Procedure 1: Example 1: [0685] tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin- 2-yl)piperazine-1-carboxylate: To a vial was added 6-bromo-2-(2,6-dimethylpyridin-4-yl)-3- methyl-1H-indole (I-2) (50 mg, 0.16 mmol), tert-butyl 4-(5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)piperazine-1-carboxylate (93 mg, 0.24 mmol), 1,4-dioxane (1 mL), 2M aqueous sodium carbonate (0.16 mL, 0.37 mmol), and [1,1′- bis(diphenylphosphino)ferrocene]dichloropalladium(II) (11.8 mg, 0.016 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 85 °C for 30 minutes. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 498.6 (M+H + ). [0686] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(6-(piperazin-1-yl)p yridin-3-yl)-1H- indole (Example 1): To a solution of tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H- indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate (40 mg, 0.080 mmol) in MeCN (1 mL) was added trifluoroacetic acid (0.2 mL). The reaction mixture was heated at 45 °C for 45 minutes. To the cooled reaction mixture was added water (0.1 mL), and the mixture was purified by RP- HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 1 as a bis- trifluoroacetate salt. ES/MS: 398.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.78 (s, 1H), 8.76 (s, 2H), 8.57 (d, J = 2.6 Hz, 1H), 8.01 (dd, J = 8.8, 2.6 Hz, 1H), 7.88 (s, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 1.4 Hz, 1H), 7.41 (dd, J = 8.4, 1.5 Hz, 1H), 7.06 (d, J = 8.9 Hz, 1H), 3.78 (t, J = 5.3 Hz, 4H), 3.27 – 3.22 (m, 4H), 2.71 (s, 6H), 2.63 (s, 3H). [0687] The following Examples were made in an analogous fashion according to Procedure 1 and are shown below in Table 1. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 1 and are noted in the last column of Table 1 – “Changes to Procedure 1: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 1 were replaced with the different reagents/starting materials noted below. Table 1. Examples 2 to 20 Procedure 2: Example 21: [0688] tert-butyl (1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6- yl)pyridin-2-yl)piperidin-4-yl)carbamate: To a microwave vial with 2-(2,6-dimethylpyridin-4- yl)-6-(6-fluoropyridin-3-yl)-3-methyl-1H-indole (I-3) (40 mg, 0.12 mmol), tert-butyl piperidin- 4-ylcarbamate (97 mg, 0.48 mmol) and DMSO (0.5 mL) was added triethylamine (0.85 mL, 0.60 mmol). The solution was heated under microwave conditions at 150 °C for 2 hours. The crude mixture was directly added to a silica loading column, and was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 512.3 (M+H + ). [0689] 1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin-2- yl)piperidin-4-amine (Example 21): To a solution of tert-butyl (1-(5-(2-(2,6-dimethylpyridin- 4-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl)piperidin-4-yl)car bamate (48 mg, 0.094 mmol) in MeCN (1 mL) was added trifluoroacetic acid (0.1 mL). The reaction mixture was heated at 45 °C overnight. To the cooled reaction mixture was added water (0.1 mL), and the mixture was purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 21 as a bis-trifluoroacetate salt. ES/MS: 412.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.80 (s, 1H), 8.52 (d, J = 2.5 Hz, 1H), 8.00 – 7.73 (m, 7H), 7.59 (d, J = 1.4 Hz, 1H), 7.40 (dd, J = 8.4, 1.6 Hz, 1H), 7.03 (d, J = 9.0 Hz, 1H), 4.41 (d, J = 13.2 Hz, 2H), 3.45 – 3.28 (m, 1H), 3.09 – 2.88 (m, 2H), 2.72 (s, 6H), 2.64 (s, 3H), 1.96 (d, J = 12.1 Hz, 2H), 1.50 (dd, J = 12.0, 4.0 Hz, 2H). [0690] The following Examples were made in an analogous fashion according to Procedure 2 and are shown below in Table 2. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 2 and are noted in the last column of Table 2 – “Changes to Procedure 2: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 2 were replaced with the different reagents/starting materials noted below. Table 2. Example 22 to 36

Procedure 3: Example 37: [0691] 2-(4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl )pyridin-2- yl)piperazin-1-yl)ethan-1-ol (Example 37): To a microwave vial with 2-(2,6-dimethylpyridin- 4-yl)-6-(6-fluoropyridin-3-yl)-3-methyl-1H-indole (I-3) (80 mg, 0.24 mmol), 2-piperazin-1- ylethanol (126 mg, 0.97 mmol) and DMSO (0.5 mL) was added N,N-diisopropylethylamine (DIPEA) (0.21 mL, 1.21 mmol). The solution was heated under microwave conditions at 150 °C for 2 hours. To the crude mixture was added TFA (0.2 mL), water (0.1 mL), and acetonitrile (0.3 mL), and the mixture was purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 37 as a bis-trifluoroacetate salt. ES/MS: 442.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.83 (s, 1H), 9.71 (s, 1H), 8.57 (d, J = 2.6 Hz, 1H), 8.02 (dd, J = 8.9, 2.6 Hz, 1H), 7.91 (s, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.42 (dd, J = 8.5, 1.6 Hz, 1H), 7.09 (d, J = 8.9 Hz, 1H), 4.45 (d, J = 13.8 Hz, 2H), 3.79 (dd, J = 6.2, 4.2 Hz, 2H), 3.34 – 3.10 (m, 6H), 2.72 (s, 6H), 2.64 (s, 3H). [0692] The following Examples were made in an analogous fashion according to Procedure 3 and are shown below in Table 3. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 3 and are noted in the last column of Table 3 – “Changes to Procedure 3: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 3 were replaced with the different reagents/starting materials noted below. Table 3. Examples 38 to 40 Procedure 4: Example 41: [0693] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(6-(4-methylpiperazi n-1-yl)pyridin- 3-yl)-1H-indole (Example 41): To a vial was added 6-bromo-2-(2,6-dimethylpyridin-4-yl)-3- methyl-1H-indole (I-2) (35 mg, 0.11 mmol), 1-methyl-4-(5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)piperazine (50.5 mg, 0.167 mmol), 1,4-dioxane (1 mL), 2M aqueous sodium carbonate (0.11 mL, 0.22 mmol), and [1,1′- Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (8.2 mg, 0.01 mmol). The solution was degassed by bubbling argon for 30 seconds, and then heated in a sealed tube at 85 °C for 2 hours. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The crude residue was purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 41 as a bis-trifluoroacetate salt. ES/MS: 412.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.95 – 11.62 (m, 1H), 9.77 – 9.56 (m, 1H), 8.57 (d, J = 2.5 Hz, 1H), 8.01 (dd, J = 8.9, 2.6 Hz, 1H), 7.97 – 7.71 (m, 3H), 7.60 (s, 1H), 7.44 – 7.32 (m, 1H), 7.09 (d, J = 8.9 Hz, 1H), 4.57 – 4.44 (m, 2H), 3.54 (d, J = 11.1 Hz, 2H), 3.15 (p, J = 11.8 Hz, 4H), 2.88 (d, J = 4.4 Hz, 3H), 2.75 – 2.65 (m, 6H), 2.62 (s, 3H). [0694] The following Examples were made in an analogous fashion according to Procedure 4 and are shown below in Table 4. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 4 and are noted in the last column of Table 4 – “Changes to Procedure 4: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 4 were replaced with the different reagents/starting materials noted below. Table 4. Examples 42 to 47

Procedure 5: Example 48: [0695] tert-butyl 4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-3,6- dihydropyridine-1(2H)-carboxylate: To a vial was added 6-bromo-2-iodo-3-methyl-1H-indole (I-1) (150 mg, 0.45 mmol), 2,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridine (114 mg, 0.49 mmol) and (1,1'-bis(diphenylphosphino)ferrocene)- dichloropalladium(II) (0.025 g, 0.036 mmol) followed by acetonitrile (8.00 mL) and 1.0 M potassium acetate in water (1.0 mol/L, 1.1 mL, 1.1 mmol). Argon was bubbled through the mixture for 3 min and the reaction was heated to 100 °C in a microwave for 20 min. Tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro- 2H-pyridine-1-carboxylate (166 mg, 0.54 mmol), Pd(AmPhos) 2 Cl2 (32 mg, 0.045 mmol) and sodium carbonate (831 mg, 1.34 mmol) were added. Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. Complete conversion to desired product was assumed and the crude product was carried through directly to next step. ES/MS: 418.3 (M+H + ) [0696] tert-butyl 4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)piper idine- 1-carboxylate: To a solution of tert-butyl 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6- yl]-3,6-dihydro-2H-pyridine-1-carboxylate (60 mg, 0.14 mmol) in ethanol (5.00 mL) under nitrogen was added palladium on carbon 10 wt. % (dry basis), (15.3 mg, 0.014 mmol) and the reaction mixture was stirred under an atmosphere of hydrogen for 16 h. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. Complete conversion to desired product was assumed and the crude product was carried through directly to next step. ES/MS: 420.3 (M+H + ) [0697] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(piperidin-4-yl)-1H- indole (Example 48): To a mixture of tert-butyl 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]piperid ine- 1-carboxylate (60 mg, 0.14 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.750 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10- 27% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18 110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 320.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 8.61 (s, 1H), 8.39 (d, J = 11.8 Hz, 1H), 7.86 (s, 3H), 7.67 (d, J = 8.4 Hz, 1H), 7.24 (s, 1H), 7.01 (dd, J = 8.4, 1.4 Hz, 1H), 3.82 (s, 1H), 3.48 – 3.35 (m, 2H), 3.14 – 2.90 (m, 2H), 2.71 (s, 6H), 2.60 (s, 3H), 1.99 (d, J = 13.6 Hz, 2H), 1.93 – 1.78 (m, 2H). Example 49: [0698] 2-(2,6-dimethylpyridin-4-yl)-3-ethyl-6-(piperidin-4-yl)-1H-i ndole (Example 49): Prepared using Procedure 5 starting from I-4 to give the title compound as a bis- trifluoroacetate salt. ES/MS: 334.4 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.60 (s, 1H), 8.67 – 8.53 (m, 1H), 8.48 – 8.27 (m, 1H), 7.80 – 7.71 (m, 2H), 7.67 (d, J = 8.3 Hz, 1H), 7.24 (d, J = 1.3 Hz, 1H), 7.00 (dd, J = 8.4, 1.4 Hz, 1H), 3.13 – 2.82 (m, 5H), 2.70 (s, 6H), 2.04 – 1.93 (m, 2H), 1.93 – 1.72 (m, 2H), 1.27 (t, J = 7.5 Hz, 3H). Example 50: [0699] 2-(2,6-dimethylpyridin-4-yl)-3-isopropyl-6-(piperidin-4-yl)- 1H-indole (Example 50): Prepared using Procedure 5 starting from I-5 to give the title compound as a bis-trifluoroacetate salt. ES/MS: 348.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.51 (s, 1H), 8.66 – 8.54 (m, 1H), 8.48 – 8.30 (m, 1H), 7.83 (d, J = 8.4 Hz, 1H), 7.66 (s, 2H), 7.24 (d, J = 1.5 Hz, 1H), 6.95 (dd, J = 8.5, 1.6 Hz, 1H), 3.51 – 3.33 (m, 3H), 3.12 – 2.89 (m, 3H), 2.71 (s, 6H), 2.03 – 1.93 (m, 2H), 1.93 – 1.78 (m, 2H), 1.46 (d, J = 7.0 Hz, 6H). [0700] The following Examples were made in an analogous fashion according to Procedure 5 and are shown below in Table 5. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 5 and are noted in the last column of Table 5 – “Changes to Procedure 5: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 5 were replaced with the different reagents/starting materials noted below. Table 5. Examples 51 to 55 Procedure 6: Example 56: [0701] 2-(2,6-dimethylpyridin-4-yl)-6-(2-methoxypyridin-4-yl)-3-met hyl-1H-indole (Example 56): To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (40 mg, 0.13 mmol), 2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyr idine (0.0358 g, 0.152 mmol) and Pd(AmPhos) 2 Cl 2 (9.0 mg, 0.013 mmol) followed by acetonitrile (2.5 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.317 mL, 0.317 mmol/0.476 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by RP-HPLC (15-35.1% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a mono-trifluoroacetate salt. ES/MS: 344.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.98 (s, 1H), 8.26 (d, J = 5.4 Hz, 1H), 7.95 (s, 2H), 7.84 (d, J = 8.5 Hz, 1H), 7.77 (d, J = 1.4 Hz, 1H), 7.53 (dd, J = 8.5, 1.6 Hz, 1H), 7.38 (dd, J = 5.5, 1.6 Hz, 1H), 7.15 (d, J = 1.5 Hz, 1H), 3.92 (s, 3H), 2.74 (s, 6H), 2.65 (s, 3H). [0702] The following Examples were made in an analogous fashion according to Procedure 6 and are shown below in Table 6. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 6 and are noted in the last column of Table 6 – “Changes to Procedure 6: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 6 were replaced with the different reagents/starting materials noted below. Table 6. Examples 57 and 58 Procedure 7: Example 59: Example 59 [0703] 4-(4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin-2- yl)morpholine (Example 59): To a solution of 2-(2,6-dimethyl-4-pyridyl)-6-(2-fluoro-4- pyridyl)-3-methyl-1H-indole (I-6) (27.0 mg, 0.082 mmol) in NMP (1.00 mL) was added morpholine (0.0703 mL, 0.815 mmol) and the reaction mixture was heated to 225 °C for 3 h under microwave irradiation. The reaction mixture was diluted with water and the mixture was extracted with DCM (3x). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo, azeotroping with toluene 2x. The crude residue was purified by RP-HPLC (10-30% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to provide the title compound as a mono-trifluoroacetate salt. ES/MS: 399.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 12.13 (s, 1H), 8.18 (d, J = 6.0 Hz, 1H), 7.97 (s, 2H), 7.89 – 7.86 (m, 2H), 7.63 (dd, J = 8.6, 1.5 Hz, 1H), 7.42 (s, 1H), 7.27 (dd, J = 6.1, 1.4 Hz, 1H), 3.83 – 3.75 (m, 4H), 3.72 – 3.65 (m, 4H), 2.75 (s, 6H), 2.66 (s, 3H). Procedure 8: Example 60: [0704] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(2-(4-methylpiperazi n-1-yl)pyridin- 4-yl)-1H-indole (Example 60): To a vial was added 6-bromo-2-iodo-3-methyl-1H-indole (I-1) (300 mg, 0.89 mmol), 2,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) pyridine (229 mg, 0.98 mmol) and (1,1'-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (50.5 mg, 0.071 mmol) followed by acetonitrile (10.0 mL) and 1.0 M potassium acetate in water (2.23 mL, 2.23 mmol). Argon was bubbled through the mixture for 4 min and the reaction was heated to 100 °C in a microwave for 20 min. Tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-3,6-dihydro-2H-pyridine-1-carboxylate (331 mg, 1.07 mmol), bis(di-tert-butyl(4- dimethylaminophenyl)phosphine)dichloropalladium(II) (63.2 mg, 0.089 mmol) and sodium carbonate (166 mg, 2.68 mmol) were added. Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite and sodium sulfate, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by RP-HPLC (10-25% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 412.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 9.73 (s, 1H), 8.25 (d, J = 5.3 Hz, 1H), 7.89 (s, 2H), 7.83 (d, J = 8.4 Hz, 1H), 7.77 (d, J = 1.5 Hz, 1H), 7.56 (dd, J = 8.5, 1.6 Hz, 1H), 7.26 (s, 1H), 7.14 (dd, J = 5.3, 1.3 Hz, 1H), 4.65 – 4.52 (m, 2H), 3.61 – 3.51 (m, 2H), 3.24 – 3.03 (m, 4H), 2.92 – 2.83 (m, 3H), 2.72 (s, 6H), 2.64 (s, 3H). [0705] The following Examples were made in an analogous fashion according to Procedure 8 and are shown below in Table 7. To prepare the below Example, different reagents/starting materials were used than some of those described in Procedure 8 and are noted in the last column of Table 7 – “Changes to Procedure 8: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 8 were replaced with the different reagents/starting materials noted below. Table 7. Example 61

Procedure 9: Example 62: [0706] tert-butyl 5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-3,4- dihydroisoquinoline-2(1H)-carboxylate: To a vial was added 6-bromo-2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indole (35.0 mg, 0.11 mmol), tert-butyl 5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3,4-dihydro-1H-isoquinoline-2-carboxylate (47.9 mg, 0.13 mmol) and Pd(AmPhos) 2 Cl 2 (7.9 mg, 0.011 mmol) followed by acetonitrile (2.2 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.28 mL, 0.28 mmol / 0.42 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 468.4 (M+H + ). [0707] 5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-1,2, 3,4- tetrahydroisoquinoline (Example 62): To a mixture of tert-butyl 5-(2-(2,6-dimethylpyridin-4- yl)-3-methyl-1H-indol-6-yl)-3,4-dihydroisoquinoline-2(1H)-ca rboxylate (45.4 mg, 0.097 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.250 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (15-34% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 368.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.85 – 11.59 (m, 1H), 8.97 (s, 2H), 7.87 – 7.66 (m, 3H), 7.39 (t, J = 7.6 Hz, 1H), 7.32 – 7.24 (m, 3H), 7.01 (d, J = 8.3 Hz, 1H), 4.41 – 4.34 (m, 2H), 3.30 – 3.27 (m, 2H), 2.84 (t, J = 6.2 Hz, 2H), 2.67 (s, 6H), 2.62 (s, 3H). Example 63: [0708] 3-cyclopropyl-2-(2,6-dimethylpyridin-4-yl)-6-(6-(piperazin-1 -yl)pyridin-3- yl)-1H-indole (Example 63): Prepared using Procedure 9 starting from I-9 and tert-butyl 4-(5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)pi perazine-1-carboxylate to give the title compound as a bis-trifluoroacetate salt. ES/MS: 424.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.82 (s, 2H), 8.55 (d, J = 2.5 Hz, 1H), 8.11 (s, 2H), 7.99 (dd, J = 8.9, 2.6 Hz, 1H), 7.86 (d, J = 8.4 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.41 (dd, J = 8.5, 1.6 Hz, 1H), 7.07 (d, J = 8.9 Hz, 1H), 3.82 – 3.75 (m, 4H), 3.28 – 3.18 (m, 4H), 2.74 (s, 6H), 2.27 – 2.18 (m, 1H), 1.22 – 1.16 (m, 2H), 0.60 – 0.51 (m, 2H). [0709] The following Examples were made in an analogous fashion according to Procedure 9 and are shown below in Table 8. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 9 and are noted in the last column of Table 8 – “Changes to Procedure 9: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 9 were replaced with the different reagents/starting materials noted below. Table 8. Examples 64 to 69

Procedure 10: Example 70: [0710] tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-ethyl-1H-indol-6-yl)pyr idin-2- yl)piperazine-1-carboxylate: To a vial was added 6-bromo-3-ethyl-2-iodo-1H-indole (I-4) (60 mg, 0.17 mmol), 2,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) pyridine (44 mg, 0.19 mmol) and (1,1'-bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (9.7 mg, 0.014 mmol) followed by acetonitrile (2.3 mL) and 1.0 M potassium acetate in water (0.43 mL, 0.43 mmol). Argon was bubbled through the mixture for 4 min and the reaction was heated to 100 °C in a microwave for 20 min. Tert-butyl 4-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2- pyridyl]piperazine-1-carboxylate (80 mg, 0.21 mmol), Pd(AmPhos) 2 Cl2 (12.1 mg, 0.017 mmol) and sodium carbonate (32 mg, 0.51 mmol) were added. Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 512.2 (M+H + ). [0711] 2-(2,6-dimethylpyridin-4-yl)-3-ethyl-6-(6-(piperazin-1-yl)py ridin-3-yl)-1H- indole (Example 70): To a mixture of tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-ethyl-1H- indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate (44.3 mg, 0.087 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (15-28% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 412.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.80 (s, 1H), 8.83 (s, 2H), 8.56 (d, J = 2.5 Hz, 1H), 8.00 (dd, J = 8.9, 2.6 Hz, 1H), 7.85 (s, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.41 (dd, J = 8.5, 1.6 Hz, 1H), 7.07 (d, J = 8.9 Hz, 1H), 3.83 – 3.74 (m, 4H), 3.31 – 3.18 (m, 4H), 3.11 (q, J = 7.5 Hz, 2H), 1.29 (t, J = 7.5 Hz, 3H). [0712] The following Examples were made in an analogous fashion according to Procedure 10 and are shown below in Table 9. To prepare the below Example, different reagents/starting materials were used than some of those described in Procedure 10 and are noted in the last column of Table 9 – “Changes to Procedure 10: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 10 were replaced with the different reagents/starting materials noted below. Table 9. Example 71

[0713] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-((trimethylsilyl)eth ynyl)-1H-indole: To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (75.0 mg, 0.24 mmol), bis(triphenylphosphine)palladium chloride (16.7 mg, 0.024 mmol) and copper(I)iodide (4.5 mg, 0.0.024 mmol). The vial was sparged with argon and dioxane (1.5 mL) was added followed by trimethylsilylacetylene (84 µL, 0.60 mmol) and triethylamine (1.1 mL, 8.1 mmol) and the reaction mixture was heated to 80 °C and stirred for 1 h. An additional portion of trimethylsilylacetylene (84 µL, 0.60 mmol) was added and the reaction mixture was heated to 80 °C for 16 h. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 333.3 (M+H + ). [0714] 2-(2,6-dimethylpyridin-4-yl)-6-ethynyl-3-methyl-1H-indole: To a solution of 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-((trimethylsilyl)eth ynyl)-1H-indole (35.6 mg, 0.107 mmol) in THF (0.80 mL) and methanol (0.20 mL) was added potassium hydroxide (9.0 mg, 0.16 mmol) and the reaction mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated in vacuo and the crude residue was purified by column chromatography (0- 100% EtOAc in hexane) to give the title compound. ES/MS: 261.1 (M+H + ). [0715] tert-butyl 4-(4-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-1 H- 1,2,3-triazol-1-yl)piperidine-1-carboxylate: To a solution of 2-(2,6-dimethylpyridin-4-yl)-6- ethynyl-3-methyl-1H-indole (11.9 mg, 0.046 mmol) in methanol (1.00 mL) was added tert-butyl 4-azidopiperidine-1-carboxylate (8.7 µL, 0.050 mmol), copper(I)iodide (0.8 mg, 4.6 µmol) and N,N-diisopropylethylamine (0.012 mL, 69 µmol) and the reaction mixture was stirred at room temperature for 90 min. Another portion of tert-butyl 4-azidopiperidine-1-carboxylate (8.7 µL, 0.050 mmol) was added and the reaction mixture was stirred for an additional 90 min. The reaction mixture was concentrated in vacuo and the crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 487.3 (M+H + ). [0716] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(1-(piperidin-4-yl)- 1H-1,2,3-triazol- 4-yl)-1H-indole (Example 72): To a mixture of tert-butyl 4-(4-(2-(2,6-dimethylpyridin-4-yl)-3- methyl-1H-indol-6-yl)-1H-1,2,3-triazol-1-yl)piperidine-1-car boxylate (14.1 mg, 0.0290 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-30% 0.1% TFA-ACN in 0.1% TFA- Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 387.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.80 (s, 1H), 8.82 – 8.67 (m, 2H), 8.51 (s, 1H), 8.01 – 7.93 (m, 2H), 7.90 – 7.73 (m, 3H), 7.62 (d, J = 8.5 Hz, 1H), 4.96 – 4.79 (m, 1H), 3.47 (d, J = 16.1 Hz, 2H), 3.26 – 3.08 (m, 2H), 2.70 (s, 6H), 2.62 (s, 3H), 2.45 – 2.31 (m, 2H), 2.31 – 2.13 (m, 2H). Procedure 12: Example 73: [0717] tert-butyl 4-(5-(3-methyl-2-(2-methylpyridin-4-yl)-1H-indol-6-yl)pyridi n-2- yl)piperazine-1-carboxylate: To a vial was added tert-butyl 4-[5-(2-bromo-3-methyl-1H-indol- 6-yl)-2-pyridyl]piperazine-1-carboxylate (I-10) (22.0 mg, 0.047 mmol), 2-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (12.3 mg, 0.056 mmol) and Pd(AmPhos) 2 Cl2 (2.6 mg, 0.0037 mmol) followed by acetonitrile (0.75 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (1.00 mol/L, 0.117 mL, 0.117 mmol/0.176 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 130 °C in a microwave for 30 min. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 484.2 (M+H + ). [0718] 3-methyl-2-(2-methylpyridin-4-yl)-6-(6-(piperazin-1-yl)pyrid in-3-yl)-1H- indole (Example 73): To a mixture of tert-butyl 4-(5-(3-methyl-2-(2-methylpyridin-4-yl)-1H- indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate (18.1 mg, 0.037 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-35% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 384.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.86 (s, 1H), 8.84 (s, 2H), 8.79 – 8.71 (m, 1H), 8.57 (s, 1H), 8.09 – 7.97 (m, 3H), 7.79 (d, J = 8.5 Hz, 1H), 7.62 (s, 1H), 7.42 (d, J = 8.6 Hz, 1H), 7.07 (d, J = 9.0 Hz, 1H), 3.84 – 3.74 (m, 5H), 3.29 – 3.18 (m, 4H), 2.74 (s, 3H), 2.65 (s, 3H). [0719] The following Examples were made in an analogous fashion according to Procedure 12 and are shown below in Table 10. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 12 and are noted in the last column of Table 10 – “Changes to Procedure 12: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 12 were replaced with the different reagents/starting materials noted below. Table 10. Examples 74 to 93 Procedure 13: Example 94: [0720] tert-butyl 4-(5-(2-(1,5-dimethyl-6-oxo-1,6-dihydropyridin-3-yl)-3-methy l-1H- indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate: To a vial was added tert-butyl 4-(5-(3- methyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-in dol-6-yl)pyridin-2-yl)piperazine- 1-carboxylate (I-11) (31.4 mg, 0.061 mmol), 5-bromo-1,3-dimethyl-pyridin-2-one (14.7 mg, 0.073 mmol) and Pd(AmPhos) 2 Cl2 (3.4 mg, 0.0049 mmol) followed by acetonitrile (0.75 mL) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.15 mL, 0.15 mmol/0.23 mmol). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 130 °C in a microwave for 20 min. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 514.3 (M+H + ) [0721] 1,3-dimethyl-5-(3-methyl-6-(6-(piperazin-1-yl)pyridin-3-yl)- 1H-indol-2- yl)pyridin-2(1H)-one (Example 94): To a mixture of tert-butyl 4-(5-(2-(1,5-dimethyl-6-oxo- 1,6-dihydropyridin-3-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl )piperazine-1-carboxylate (17.8 mg, 0.035 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.250 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (15-31% 0.1% TFA- ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 414.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H), 8.76 (s, 2H), 8.50 (d, J = 2.5 Hz, 1H), 7.96 (dd, J = 8.9, 2.5 Hz, 1H), 7.86 (d, J = 2.6 Hz, 1H), 7.69 – 7.64 (m, 1H), 7.55 (d, J = 8.2 Hz, 1H), 7.48 (d, J = 1.6 Hz, 1H), 7.29 (dd, J = 8.2, 1.6 Hz, 1H), 7.04 (d, J = 8.9 Hz, 1H), 3.80 – 3.74 (m, 4H), 3.56 (s, 3H), 3.28 – 3.18 (m, 4H), 2.36 (s, 3H), 2.12 (s, 3H). Example 95: [0722] 1-methyl-5-(3-methyl-6-(6-(piperazin-1-yl)pyridin-3-yl)-1H-i ndol-2- yl)pyridin-2(1H)-one (Example 95): Prepared using Procedure 13 starting from I-23 and 5- bromo-1-methylpyridin-2(1H)-one to give the title compound as a bis-trifluoroacetate salt. ES/MS: 400.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.11 (s, 1H), 8.74 (s, 2H), 8.51 (d, J = 2.5 Hz, 1H), 8.00 (d, J = 2.6 Hz, 1H), 7.96 (dd, J = 8.8, 2.6 Hz, 1H), 7.76 (dd, J = 9.4, 2.7 Hz, 1H), 7.56 (d, J = 8.2 Hz, 1H), 7.49 (d, J = 1.5 Hz, 1H), 7.29 (dd, J = 8.3, 1.6 Hz, 1H), 7.04 (d, J = 8.9 Hz, 1H), 6.56 (d, J = 9.4 Hz, 1H), 3.80 – 3.73 (m, 4H), 3.55 (s, 3H), 3.28 – 3.18 (m, 4H), 2.36 (s, 3H). [0723] The following Examples were made in an analogous fashion according to Procedure 13 and are shown below in Table 11. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 13 and are noted in the last column of Table 11 – “Changes to Procedure 13: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 13 were replaced with the different reagents/starting materials noted below.

Table 11. Examples 96 to 99

Procedure 14: Example 100: [0724] tert-butyl 4-((2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6- yl)ethynyl)piperidine-1-carboxylate: To a vial was added 6-bromo-2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indole (I-2) (40.0 mg, 0.127 mmol), tert-butyl 4-ethynylpiperidine-1- carboxylate (31.9 mg, 0.15 mmol), bis(triphenylphosphine)palladium chloride (8.9 mg, 0.013 mmol) and copper(I)iodide (2.4 mg, 0.013 mmol). The vial was sparged with argon and dioxane (3.0 mL) was added followed by triethylamine (0.89 mL, 8.1 mmol) and the reaction mixture was heated to 80 °C and stirred for 16 h. The reaction mixture was filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 444.3 (M+H + ). [0725] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(piperidin-4-ylethyn yl)-1H-indole (Example 100): To a mixture of tert-butyl 4-((2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol- 6-yl)ethynyl)piperidine-1-carboxylate (28.6 mg, 0.065 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-35% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 344.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 12.05 (s, 1H), 8.56 – 8.41 (m, 1H), 8.19 (d, J = 11.2 Hz, 1H), 8.04 (d, J = 1.3 Hz, 1H), 7.79 (d, J = 8.5 Hz, 2H), 7.74 (dd, J = 8.5, 1.5 Hz, 1H), 3.15 – 3.04 (m, 2H), 3.02 – 2.85 (m, 2H), 2.69 (s, 6H), 2.59 (s, 3H), 2.28 – 2.14 (m, 1H), 1.96 – 1.81 (m, 2H), 1.51 – 1.34 (m, 2H). [0726] tert-butyl 4-(5-(3-cyano-2-(2,6-dimethylpyridin-4-yl)-1H-indol-6-yl)pyr idin- 2-yl)piperazine-1-carboxylate: To a solution of tert-butyl 4-(5-(3-cyano-1H-indol-6- yl)pyridin-2-yl)piperazine-1-carboxylate (I-12) (101 mg, 0.25 mmol) in 1,1,1,3,3,3-hexafluoro- 2-propanol (1.0 mL) was added N-bromosuccinimide (66.7 mg, 0.375 mmol) and the reaction mixture was stirred for 5 min at room temperature. To the reaction mixture was added 2,6- dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri dine (81.9 mg, 0.35 mmol), Pd(AmPhos) 2 Cl 2 (26.6 mg, 0.038 mmol) and 1.0 M potassium acetate/1.5 M sodium carbonate in water (0.63 mL, 0.63 mmol) and the reaction mixture was heated to 120 °C in a microwave for 30 min. The reaction mixture was dried over sodium sulfate, filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 509.3 (M+H + ). [0727] 2-(2,6-dimethylpyridin-4-yl)-6-(6-(piperazin-1-yl)pyridin-3- yl)-1H-indole-3- carbonitrile (Example 101): To a mixture of tert-butyl 4-(5-(3-cyano-2-(2,6-dimethylpyridin- 4-yl)-1H-indol-6-yl)pyridin-2-yl)piperazine-1-carboxylate (51.5 mg, 0.10 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-35% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 409.2 (M+H + ). 1 H NMR (400 MHz, DMSO- d6) δ 12.36 (d, J = 3.0 Hz, 1H), 8.79 – 8.68 (m, 3H), 8.34 (d, J = 3.0 Hz, 1H), 8.14 (d, J = 2.4 Hz, 1H), 8.05 (s, 2H), 7.92 – 7.85 (m, 1H), 7.76 (d, J = 8.4 Hz, 1H), 7.65 (dd, J = 8.4, 1.6 Hz, 1H), 3.35 – 3.26 (m, 4H), 3.20 – 3.08 (m, 4H), 2.72 (s, 6H). Example 102: [0728] 1-(2-(2,6-dimethylpyridin-4-yl)-6-(6-(piperazin-1-yl)pyridin -3-yl)-1H-indol- 3-yl)ethan-1-one (Example 102): Prepared using Procedure 15 starting from I-13 to give the title compound as a bis-trifluoroacetate salt. ES/MS: 426.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 12.06 (d, J = 3.1 Hz, 1H), 8.74 (d, J = 2.4 Hz, 1H), 8.69 (s, 2H), 8.38 (d, J = 3.1 Hz, 1H), 8.27 (d, J = 8.3 Hz, 1H), 8.13 (d, J = 2.4 Hz, 1H), 8.04 (s, 2H), 7.79 (d, J = 1.5 Hz, 1H), 7.58 (dd, J = 8.4, 1.6 Hz, 1H), 3.37 – 3.23 (m, 4H), 3.22 – 3.05 (m, 4H), 2.71 (s, 6H), 2.48 (s, 3H).

Procedure 16: Example 103: [0729] tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin- 2-yl)-1,4-diazepane-1-carboxylate: To a vial containing 2-(2,6-dimethyl-4-pyridyl)-6-(6- fluoro-3-pyridyl)-3-methyl-1H-indole (I-3) (40.0 mg, 0.12 mmol) was added DMSO (0.5 mL), N,N-diisopropylethylamine (0.21 mL, 1.21 mmol) and tert-butyl 1,4-diazepane-1-carboxylate (0.095 mL, 0.48 mmol) and the reaction mixture was heated in a microwave to 150 °C for 4 h. The reaction mixture was diluted with water and extracted with DCM (2x). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 512.3 (M+H + ). [0730] 6-(6-(1,4-diazepan-1-yl)pyridin-3-yl)-2-(2,6-dimethylpyridin -4-yl)-3-methyl- 1H-indole (Example 103): To a mixture of tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3- methyl-1H-indol-6-yl)pyridin-2-yl)-1,4-diazepane-1-carboxyla te (17.2 mg, 0.034 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo crude reaction mixture was purified directly by RP-HPLC (10-35% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 412.2 (M+H + ). 1 H NMR (400 MHz, DMSO- d6) δ 11.79 (s, 1H), 8.68 (s, 2H), 8.52 (d, J = 2.5 Hz, 1H), 7.96 (dd, J = 8.9, 2.6 Hz, 1H), 7.90 (s, 2H), 7.77 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 1.9 Hz, 1H), 7.40 (dd, J = 8.5, 1.6 Hz, 1H), 6.89 (d, J = 9.0 Hz, 1H), 3.98 (t, J = 5.2 Hz, 2H), 3.73 (t, J = 6.1 Hz, 2H), 3.34 – 3.26 (m, 2H), 3.25 – 3.15 (m, 2H), 2.72 (s, 6H), 2.64 (s, 3H), 2.16 – 2.03 (m, 2H). Example 104: [0731] 1-(5-(3-cyclopropyl-2-(2,6-dimethylpyridin-4-yl)-1H-indol-6- yl)pyridin-2- yl)piperidin-4-amine (Example 104): Prepared using Procedure 16 starting from I-19 and tert- butyl piperidin-4-ylcarbamate to give the title compound as a bis-trifluoroacetate salt. ES/MS: 438.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 8.51 (d, J = 2.6 Hz, 1H), 8.11 (s, 2H), 7.95 (dd, J = 8.9, 2.6 Hz, 1H), 7.92 – 7.86 (m, 3H), 7.85 (d, J = 8.5 Hz, 1H), 7.60 (d, J = 1.5 Hz, 1H), 7.40 (dd, J = 8.5, 1.6 Hz, 1H), 7.04 (d, J = 9.0 Hz, 1H), 4.46 – 4.33 (m, 2H), 3.41 – 3.28 (m, 1H), 3.05 – 2.92 (m, 2H), 2.74 (s, 6H), 2.28 – 2.17 (m, 1H), 2.02 – 1.90 (m, 2H), 1.59 – 1.43 (m, 2H), 1.23 – 1.15 (m, 2H), 0.62 – 0.50 (m, 2H). [0732] The following Examples were made in an analogous fashion according to Procedure 16 and are shown below in Table 12. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 16 and are noted in the last column of Table 12 – “Changes to Procedure 16: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 16 were replaced with the different reagents/starting materials noted below. Table 12. Example 105

Procedure 17: Example 106: [0733] 2-(dimethylamino)-1-(4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-me thyl-1H-indol- 6-yl)pyridin-2-yl)piperazin-1-yl)ethan-1-one (Example 106): To a solution of 2-(2,6- dimethylpyridin-4-yl)-3-methyl-6-(6-(piperazin-1-yl)pyridin- 3-yl)-1H-indole hydrochloride (I- 15) (20.0 mg, 0.046 mmol) in DCM (0.75 mL) was added N,N-dimethylglycine (4.8 mg, 0.0461 mmol), 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (21.0 mg, 0.055 mmol) and N,N-diisopropylethylamine (0.040 mL, 0.23 mmol) and the reaction mixture was stirred for 1 hour. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate and the mixture was extracted with DCM (2x). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified directly by RP-HPLC (15-37% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 483.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.79 (s, 1H), 9.56 (s, 1H), 8.55 (d, J = 2.6 Hz, 1H), 7.98 (dd, J = 8.9, 2.6 Hz, 1H), 7.91 (s, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 1.5 Hz, 1H), 7.42 (dd, J = 8.5, 1.6 Hz, 1H), 7.03 (d, J = 8.9 Hz, 1H), 4.34 (d, J = 4.9 Hz, 2H), 3.73 – 3.61 (m, 8H), 2.84 (d, J = 4.6 Hz, 6H), 2.72 (s, 6H), 2.64 (s, 3H). [0734] The following Examples were made in an analogous fashion according to Procedure 17 and are shown below in Table 13. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 17 and are noted in the last column of Table 13 – “Changes to Procedure 17: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 17 were replaced with the different reagents/starting materials noted below. Table 13. Examples 107 to 112

Procedure 18: Example 113: [0735] 2-(4-(5-(2-(2,6-dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3- methyl-1H- indol-6-yl)pyridin-2-yl)piperazin-1-yl)-N-methylacetamide: To a solution of 2-(2,6- dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3-methyl-6-(6-(pip erazin-1-yl)pyridin-3-yl)-1H- indole hydrochloride (I-16) (20.0 mg, 0.036 mmol) and sodium iodide (1.1 mg, 0.0072 mmol) in DMF (0.75 mL) was added N,N-diisopropylethylamine (0.031 mL, 0.180 mmol) and 2-bromo- N-methyl-acetamide (0.011 mL, 0.108 mmol) and the reaction mixture was stirred at 120 °C for 10 min. The reaction mixture was concentrated in vacuo and used as crude in the following reaction. ES/MS: 589.3 (M+H + ). [0736] 2-(4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl )pyridin-2- yl)piperazin-1-yl)-N-methylacetamide (Example 113): To a solution of 2-(4-(5-(2-(2,6- dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3-methyl-1H-indol- 6-yl)pyridin-2-yl)piperazin-1- yl)-N-methylacetamide in DCM (1.0 mL) was added triethylsilane (0.058 mL, 0.36 mmol) followed by trifluoromethanesulfonic acid (0.0096 mL, 0.36 mmol) and the reaction mixture was stirred for 10 min at room temperature. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-38% 0.1% TFA-ACN in 0.1% TFA-Water, 15 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a mono-trifluoroacetate salt. ES/MS: 469.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 10.32 – 10.06 (m, 1H), 8.57 (d, J = 2.6 Hz, 1H), 8.48 (s, 1H), 8.02 (dd, J = 8.9, 2.6 Hz, 1H), 7.90 (s, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.42 (dd, J = 8.5, 1.6 Hz, 1H), 7.06 (d, J = 8.9 Hz, 1H), 4.54 – 4.33 (m, 2H), 3.98 (s, 2H), 3.27 – 3.12 (m, 6H), 2.73 – 2.69 (m, 9H), 2.64 (s, 3H). Example 114 and Example 115: [0737] 2-(4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl )pyridin-2- yl)piperazin-1-yl)-N,N-dimethylethan-1-amine (Example 114) and 1,1-bis(2- (dimethylamino)ethyl)-4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-m ethyl-1H-indol-6-yl)pyridin- 2-yl)piperazin-1-ium (Example 115): Prepared using Procedure 18 starting from I-16 and 2- chloro-N,N-dimethyl-ethanamine hydrochloride to give the title compounds as bis- trifluoroacetate and mono-trifluoroacetate salts respectively. Example 114: [0738] ES/MS: 469.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.78 (s, 1H), 8.53 (d, J = 2.6 Hz, 1H), 7.96 (d, J = 8.5 Hz, 1H), 7.89 (s, 2H), 7.78 (d, J = 8.5 Hz, 1H), 7.59 (d, J = 1.6 Hz, 1H), 7.41 (dd, J = 8.5, 1.6 Hz, 1H), 7.03 (d, J = 8.8 Hz, 1H), 3.79 – 3.09 (m, 12H), 2.84 (s, 6H), 2.72 (s, 6H), 2.64 (s, 3H). Example 115: [0739] ES/MS: 540.4 (M + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.85 (s, 1H), 8.53 (d, J = 2.5 Hz, 1H), 7.98 (dd, J = 8.9, 2.6 Hz, 1H), 7.92 (s, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 1.5 Hz, 1H), 7.41 (dd, J = 8.5, 1.6 Hz, 1H), 7.05 (d, J = 8.9 Hz, 1H), 4.06 (s, 2H), 3.83 – 3.74 (m, 2H), 3.70 – 3.56 (m, 8H), 3.20 (s, 6H), 3.06 – 2.92 (m, 2H), 2.86 (s, 6H), 2.80 – 2.72 (m, 2H), 2.73 (s, 6H), 2.65 (s, 3H). [0740] The following Examples were made in an analogous fashion according to Procedure 18 and are shown below in Table 14. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 18 and are noted in the last column of Table 14 – “Changes to Procedure 14: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 18 were replaced with the different reagents/starting materials noted below. Table 14. Examples 116 to 118

Procedure 19: Example 119: [0741] (S)-(3-aminopyrrolidin-1-yl)(3-(2-(2,6-dimethylpyridin-4-yl) -3-methyl-1H- indol-6-yl)phenyl)methanone (Example 119): To a solution of 3-[2-(2,6-dimethyl-4-pyridyl)- 3-methyl-1H-indol-6-yl]benzoic acid (I-17) (20.0 mg, 0.056 mmol) in DCM (0.75 mL) was added tert-butyl N-[(3S)-pyrrolidin-3-yl]carbamate (11.5 mg, 0.062 mmol), 2-(7-Aza-1H- benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (25.6 mg, 0.067 mmol) and N,N-diisopropylethylamine (0.049 mL, 0.28 mmol) and the reaction mixture was stirred for 1 hour. Trifluoroacetic acid (0.250 mL, 3.27 mmol) was added and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude residue was purified directly by RP-HPLC (10-38% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 425.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.84 (s, 1H), 8.08 (s, 2H), 7.96 (s, 1H), 7.91 – 7.76 (m, 5H), 7.67 (d, J = 1.5 Hz, 1H), 7.60 (t, J = 7.8 Hz, 1H), 7.55 – 7.48 (m, 1H), 7.45 (dd, J = 8.4, 1.6 Hz, 1H), 3.99 – 3.88 (m, 1H), 3.88 – 3.51 (m, 3H), 2.71 (s, 6H), 2.64 (s, 3H), 2.56 – 2.53 (m, 1H), 2.30 – 2.18 (m, 1H), 2.08 – 1.93 (m, 1H). Example 120: [0742] (S)-(3-aminopyrrolidin-1-yl)(4-(2-(2,6-dimethylpyridin-4-yl) -3-methyl-1H- indol-6-yl)phenyl)methanone (Example 120): Prepared using Procedure 19 starting from I-18 to give the title compound as a bis-trifluoroacetate salt. ES/MS: 425.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 8.09 – 8.00 (m, 2H), 7.98 – 7.89 (m, 2H), 7.86 – 7.75 (m, 3H), 7.71 – 7.62 (m, 3H), 7.47 (d, J = 8.4 Hz, 1H), 3.99 – 3.43 (m, 3H), 2.68 (s, 6H), 2.62 (s, 3H), 2.31 – 2.17 (m, 2H), 2.08 – 1.93 (m, 2H). [0743] The following Examples were made in an analogous fashion according to Procedure 19 and are shown below in Table 15. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 19 and are noted in the last column of Table 15 – “Changes to Procedure 19: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 19 were replaced with the different reagents/starting materials noted below. Table 15. Examples 121 to 128

Procedure 20: Example 129: [0744] 1-(4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl )pyridin-2- yl)piperazin-1-yl)-2-methylpropan-2-ol (Example 129): To a suspension of 2-(2,6- dimethylpyridin-4-yl)-1-(4-methoxybenzyl)-3-methyl-6-(6-(pip erazin-1-yl)pyridin-3-yl)-1H- indole hydrochloride (I-16) (40.0 mg, 0.072 mmol) in acetonitrile (0.75 mL) was added N,N- diisopropylethylamine (0.0466 g, 0.63 mmol) and isobutylene oxide (0.013 mL, 0.144 mmol) and the reaction mixture was stirred at 100 °C for 16 h. An additional portion of isobutylene oxide (0.013 mL, 0.144 mmol) was added and the reaction mixture was stirred at 120 °C for 3 h. An additional portion of isobutylene oxide (0.026 mL, 0.29 mmol) was added and the reaction mixture was stirred at 100 °C for 16 h. An additional portion of isobutylene oxide (13 uL, 0.144 mmol) was added and the reaction mixture was stirred at 100 °C for 2 h. An additional portion of isobutylene oxide (26 uL, 0.29 mmol) was added and the reaction mixture was stirred at 100 °C for 3 h. The reaction mixture was concentrated in vacuo, trifluoroacetic acid (0.250 mL, 3.27 mmol) was added and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was made basic by the dropwise addition of 2 N aqueous sodium carbonate, re- acidified with TFA (0.25 mL) then concentrated in vacuo. The crude residue was purified directly by RP-HPLC (10-38% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis- trifluoroacetate salt. ES/MS: 470.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 9.20 (s, 1H), 8.57 (d, J = 2.5 Hz, 1H), 8.02 (dd, J = 8.8, 2.6 Hz, 1H), 7.90 (s, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.42 (dd, J = 8.5, 1.5 Hz, 1H), 7.07 (d, J = 8.9 Hz, 1H), 5.32 (s, 1H), 4.35 – 4.21 (m, 2H), 3.72 – 3.60 (m, 4H), 3.31 – 3.16 (m, 4H), 2.72 (s, 6H), 2.64 (s, 3H), 1.30 (s, 6H). Procedure 21: Example 130: [0745] (R)-1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-y l)pyridin-2-yl)- N-(2-((trimethylsilyl)oxy)ethyl)pyrrolidin-3-amine: To a solution of (R)-1-(5-(2-(2,6- dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl)py rrolidin-3-amine hydrochloride (I-21) (21.6 mg, 0.050 mmol) in DCE (2.mL) was added 2-[tert- butyl(dimethyl)silyl]oxyacetaldehyde (0.0083 mL, 0.044 mmol) and one drop of acetic acid and the reaction mixture was stirred for 16 h at room temperature. Sodium borohydride (3.8 mg, 0.10 mmol) was then added followed by ethanol (1 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate and the mixture was extracted with DCM (3x). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-10% MeOH in DCM) to give the title compound which was used directly in the next step. ES/MS: 556.4 (M+H + ). [0746] (R)-2-((1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol -6-yl)pyridin-2- yl)pyrrolidin-3-yl)amino)ethan-1-ol (Example 130): To a mixture of (R)-1-(5-(2-(2,6- dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl)-N -(2- ((trimethylsilyl)oxy)ethyl)pyrrolidin-3-amine (20.8 mg, 0.0374 mmol) in methanol (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-35% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 442.3 (M+H + ). 1 H NMR (400 MHz, DMSO- d6) δ 11.79 (s, 1H), 8.96 – 8.77 (m, 2H), 8.51 (d, J = 2.5 Hz, 1H), 8.00 (d, J = 8.8 Hz, 1H), 7.90 (s, 2H), 7.78 (d, J = 8.5 Hz, 1H), 7.59 (s, 1H), 7.41 (d, J = 8.4 Hz, 1H), 6.71 (d, J = 8.8 Hz, 1H), 5.35 (s, 1H), 4.08 – 3.95 (m, 1H), 3.93 – 3.79 (m, 1H), 3.74 – 3.67 (m, 2H), 3.67 – 3.61 (m, 3H), 3.21 – 3.08 (m, 2H), 2.72 (s, 6H), 2.64 (s, 3H), 2.45 – 2.38 (m, 1H), 2.30 – 2.17 (m, 1H). Example 131: [0747] 2-((1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-y l)pyridin-2- yl)piperidin-4-yl)amino)ethan-1-ol (Example 131): Prepared using Procedure 21 starting from I-22 to give the title compound as a bis-trifluoroacetate salt. ES/MS: 456.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 8.53 (d, J = 2.4 Hz, 1H), 8.48 (s, 1H), 7.96 (dd, J = 9.0, 2.4 Hz, 1H), 7.91 (s, 2H), 7.89 – 7.83 (m, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.59 (s, 1H), 7.41 (d, J = 8.5 Hz, 1H), 7.09 – 7.00 (m, 1H), 5.33 (s, 1H), 4.60 – 4.43 (m, 2H), 3.82 – 3.74 (m, 2H), 3.41 – 3.17 (m, 2H), 3.07 (s, 1H), 2.97 – 2.86 (m, 2H), 2.72 (s, 6H), 2.64 (s, 3H), 2.16 – 2.05 (m, 2H), 1.79 – 1.44 (m, 2H). Procedure 22: Example 132: [0748] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(1',2',3',6'-tetrahy dro-[2,4'- bipyridin]-5-yl)-1H-indole (Example 132): To a mixture of tert-butyl 4-[5-[2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indol-6-yl]-2-pyridyl]-3,6-dihydro-2H-p yridine-1-carboxylate (I-28) (14.5 mg, 0.029 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10- 38% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18 110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 395.2 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.92 (s, 1H), 8.98 (d, J = 2.4 Hz, 1H), 8.87 (s, 2H), 8.19 (dd, J = 8.3, 2.5 Hz, 1H), 7.91 (s, 2H), 7.85 (d, J = 8.4 Hz, 1H), 7.78 – 7.71 (m, 2H), 7.52 (dd, J = 8.4, 1.5 Hz, 1H), 6.80 (d, J = 4.1 Hz, 1H), 3.92 – 3.83 (m, 2H), 3.41 – 3.33 (m, 2H), 2.87 – 2.79 (m, 2H), 2.72 (s, 6H), 2.65 (s, 3H). Procedure 23: Example 133: [0749] tert-butyl 4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin- 2-yl)piperidine-1-carboxylate: To a solution of tert-butyl 4-[5-[2-(2,6-dimethyl-4-pyridyl)-3- methyl-1H-indol-6-yl]-2-pyridyl]-3,6-dihydro-2H-pyridine-1-c arboxylate (I-28) (79.3 mg, 0.16 mmol) in ethanol (2.5 mL) was added ammonium formate (202 mg, 3.21 mmol). Nitrogen was bubbled through the solution for 4 min then palladium on carbon 10 wt. % (34.1 mg, 0.032 mmol) was added and the reaction mixture was heated to 85 °C for 1 h. The reaction mixture was diluted with DCM and filtered through celite, eluting with DCM and the filtrate was concentrated in vacuo to give the crude product which was used directly in the next step. ES/MS: 497.2 (M+H + ). [0750] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(1',2',3',6'-tetrahy dro-[2,4'- bipyridin]-5-yl)-1H-indole (Example 133): To a mixture of tert-butyl 4-[5-[2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indol-6-yl]-2-pyridyl]piperidine-1-carb oxylate (15.5 mg, 0.0312 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude reaction mixture was purified directly by RP-HPLC (10-35% 0.1% TFA-ACN in 0.1% TFA- Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 397.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 8.91 (d, J = 2.4 Hz, 1H), 8.61 (d, J = 11.7 Hz, 1H), 8.36 (d, J = 11.3 Hz, 1H), 8.12 (dd, J = 8.1, 2.5 Hz, 1H), 7.92 (s, 2H), 7.84 (d, J = 8.4 Hz, 1H), 7.70 (d, J = 1.4 Hz, 1H), 7.47 (dd, J = 8.5, 1.5 Hz, 1H), 7.42 (d, J = 8.2 Hz, 1H), 3.47 – 3.33 (m, 2H), 3.15 – 2.99 (m, 3H), 2.73 (s, 6H), 2.65 (s, 3H), 2.12 – 2.03 (m, 2H), 2.03 – 1.86 (m, 2H). Procedure 24: Example 134: [0751] (R)-2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(6-(4-prolylpipe razin-1- yl)pyridin-3-yl)-1H-indole (Example 134): To a solution of 2-(2,6-dimethylpyridin-4-yl)-3- methyl-6-(6-(piperazin-1-yl)pyridin-3-yl)-1H-indole hydrochloride (I-15) (20.0 mg, 0.046 mmol) in DCM (0.75 mL) was added N-Boc-D-Proline (9.9 mg, 0.046 mmol), 2-(7-Aza-1H- benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (21.0 mg, 0.055 mmol) and N,N-diisopropylethylamine (0.040 mL, 0.23 mmol) and the reaction mixture was stirred for 30 min. Trifluoroacetic acid (0.25 mL) was added and the reaction mixture was stirred for 10 min at room temperature. The reaction mixture was concentrated in vacuo and the crude residue was purified directly by RP-HPLC (10-40% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 495.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 9.40 – 9.22 (m, 1H), 8.63 – 8.47 (m, 2H), 7.99 (dd, J = 8.9, 2.6 Hz, 1H), 7.91 (s, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 1.4 Hz, 1H), 7.42 (dd, J = 8.4, 1.6 Hz, 1H), 7.04 (d, J = 8.9 Hz, 1H), 4.74 – 4.62 (m, 1H), 3.77 – 3.59 (m, 8H), 3.34 – 3.25 (m, 1H), 3.25 – 3.13 (m, 1H), 2.72 (s, 6H), 2.64 (s, 3H), 2.48 – 2.36 (m, 1H), 2.02 – 1.78 (m, 3H). [0752] The following Examples were made in an analogous fashion according to Procedure 24 and are shown below in Table 16. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 24 and are noted in the last column of Table 16 – “Changes to Procedure 24: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 24 were replaced with the different reagents/starting materials noted below. Table 16. Examples 135 to 140 Procedure 25: Example 141: [0753] 6-(6-(4-(2,2-difluoroethyl)piperazin-1-yl)pyridin-3-yl)-2-(2 ,6- dimethylpyridin-4-yl)-3-methyl-1H-indole (Example 141): To a solution of 2-(2,6-dimethyl- 4-pyridyl)-6-(6-fluoro-3-pyridyl)-3-methyl-1H-indole (30.0 mg, 0.091 mmol) (I-3) in 2- methoxyethanol (0.5 mL) was added N,N-diisopropylethylamine (0.158 mL, 0.91 mmol) and 1- (2,2-difluoroethyl)piperazine (0.13 mL, 0.91 mmol) and the reaction mixture was heated in a microwave to 150 °C for 8 h. Trifluoroacetic acid (0.2 mL) was added and the reaction mixture was purified directly by RP-HPLC (10-48% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a mono-trifluoroacetate salt. ES/MS: 462.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 8.53 (d, J = 2.5 Hz, 1H), 7.99 (dd, J = 8.9, 2.6 Hz, 1H), 7.92 (s, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 1.4 Hz, 1H), 7.41 (dd, J = 8.4, 1.6 Hz, 1H), 7.04 (d, J = 9.0 Hz, 1H), 6.35 (t, J = 55.0 Hz, 1H), 3.22 – 2.85 (m, 10H), 2.72 (s, 6H), 2.65 (s, 3H). [0754] The following Examples were made in an analogous fashion according to Procedure 25 and are shown below in Table 17. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 25 and are noted in the last column of Table 17 – “Changes to Procedure 25: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 25 were replaced with the different reagents/starting materials noted below. Table 17. Examples 142 to 144 Procedure 26: Example 145: [0755] tert-butyl (1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6- yl)picolinoyl)piperidin-4-yl)carbamate: To a solution of 5-(2-(2,6-dimethylpyridin-4-yl)-3- methyl-1H-indol-6-yl)picolinic acid (I-18) (19.5mg, 0.055 mmol) in DMF (0.75 mL) was added tert-butyl N-(4-piperidyl)carbamate (12.0 mg, 0.060 mmol) and chloro-N,N,N',N'- tetramethylformamidinium hexafluorophosphate (TCFH) (23.0 mg, 0.082 mmol) followed by 1- methylimidazole (0.015 mL, 0.19 mmol) and the reaction mixture was stirred for 16 h at room temperature. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate and the mixture was extracted with DCM (3x). The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography (0-100% EtOAc in hexane) to give the title compound. ES/MS: 540.4 (M+H + ). [0756] (4-aminopiperidin-1-yl)(5-(2-(2,6-dimethylpyridin-4-yl)-3-me thyl-1H-indol- 6-yl)pyridin-2-yl)methanone (Example 145): To a solution of tert-butyl (1-(5-(2-(2,6- dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)picolinoyl)pipe ridin-4-yl)carbamate (16.0 mg, 0.030 mmol) in DCM (0.75 mL) was added trifluoroacetic acid (0.25 mL) and the reaction mixture was stirred at room temperature for 10 min. The reaction mixture was concentrated in vacuo and the crude residue was purified directly by RP-HPLC (10-38% 0.1% TFA-ACN in 0.1% TFA-Water, 10 min gradient, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound as a bis-trifluoroacetate salt. ES/MS: 440.3 (M+H + ). 1 H NMR (400 MHz, DMSO-d6) δ 11.96 – 11.82 (m, 1H), 8.96 (d, J = 2.3 Hz, 1H), 8.28 (dd, J = 8.2, 2.4 Hz, 1H), 7.88 (s, 4H), 7.75 (s, 1H), 7.69 (d, J = 8.1 Hz, 1H), 7.52 (d, J = 8.7 Hz, 1H), 4.62 – 4.50 (m, 1H), 4.00 – 3.90 (m, 1H), 3.23 – 3.12 (m, 1H), 2.99 – 2.86 (m, 1H), 2.68 (s, 6H), 2.62 (s, 3H), 2.46 – 2.37 (m, 1H), 2.07 – 2.00 (m, 1H), 1.94 – 1.86 (m, 1H), 1.62 – 1.44 (m, 2H). Procedure 27: Example 146: [0757] tert-butyl 6-[2-(4-tert-butoxycarbonylpiperazin-1-yl)thiazol-4-yl]-2-(2 ,6- dimethyl-4-pyridyl)-3-methyl-indole-1-carboxylate: To a vial was added tert-butyl 2-(2,6- dimethyl-4-pyridyl)-3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)indole-1- carboxylate (I-25) (30 mg, 0.064 mmol), tert-butyl 4-(4-bromothiazol-2-yl)piperazine-1- carboxylate (34 mg, 0.097 mmol), XPhos Pd G2 (7.7 mg, 0.00097 mmol), cesium carbonate (63 mg, 0.19 mmol), 10:11,4-dioxane and water (1 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc/ Hex) to provide the desired product. ES/MS: 604.2 (M+H + ). [0758] 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-pipe razin-1-yl- thiazole (Example 146): To a solution of tert-butyl 6-[2-(4-tert-butoxycarbonylpiperazin-1- yl)thiazol-4-yl]-2-(2,6-dimethyl-4-pyridyl)-3-methyl-indole- 1-carboxylate (20 mg, 0.033 mmol) in DCM (1 mL) was added trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temp for 1 hr. The crude was concentrated and purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 146 as a bis-trifluoroacetate salt. ES/MS: 404.2 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 7.98 (d, J = 1.4 Hz, 1H), 7.89 (s, 2H), 7.72 – 7.66 (m, 2H), 7.21 (s, 1H), 3.86 (q, J = 4.2, 3.1 Hz, 4H), 3.45 (dd, J = 6.4, 4.5 Hz, 4H), 2.79 (s, 6H), 2.70 (s, 3H). Procedure 28: Example 147: [0759] 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-nitr o-aniline: To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (350 mg, 1.11 mmol), (4-amino-3-nitro-phenyl)boronic acid (242 mg, 1.33 mmol), XPhos Pd G2 (46 mg, 0.056 mmol), cesium carbonate (1085 mg, 3.33 mmol), 10:11,4-dioxane and water (5 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, the crude was diluted with ethyl acetate, washed with organic layer with water. The organic layer filtered through Celite (eluent: EtOAc), and concentrated to provide the crude product, which was used directly on the next step. ES/MS: 373.2 (M+H + ). [0760] tert-butyl 4-[5-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-1H- benzimidazol-2-yl]piperidine-1-carboxylate.4-[2-(2,6-dimethy l-4-pyridyl)-3-methyl-1H- indol-6-yl]-2-nitro-aniline, (50 mg, 0.13 mmol) and 4-formyl-piperidine-1-carboxylic acid-tert- butyl ester (34.4 g, 0.16 mmol) were charged in a vial. The mixture was dissolved in 2 mL of EtOH/H 2 O (2:1) and sodium dithionite (82.5 mg, 0.41 mmol) was added in one portion. The flask was immediately sealed and heated to 80 °C for 16 hr. The LCMS was taken to check the full conversion of the starting materials. The crude residue was dissolved in ethyl acetate and washed with water. The phases were separated, and the aqueous layer was extracted once with ethyl acetate. The combined organic layers were washed with sat. aq. NH 4 Cl (15 mL), brine (15 mL), and then the organic layer was dried over MgSO4, filtered, and concentrated. The crude was used directly for the next step. ES/MS: 536.4 (M+H + ). [0761] 5-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-(4-p iperidyl)-1H- benzimidazole (WC-2): To a solution of tert-butyl 6-[2-(4-tert-butoxycarbonylpiperazin-1- yl)thiazol-4-yl]-2-(2,6-dimethyl-4-pyridyl)-3-methyl-indole- 1-carboxylate (20 mg, 0.033 mmol) in DCM (1 mL) was added trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temp for 1 hr. The crude was conc and purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 147 as a bis-trifluoroacetate salt. ES/MS: 436.2 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 7.96 (s, 1H), 7.92 (s, 2H), 7.85 – 7.72 (m, 5H), 7.51 (dd, J = 8.6, 1.6 Hz, 1H), 3.63 (d, J = 13.3 Hz, 3H), 2.80 (s, 6H), 2.74 (s, 3H), 2.48 (d, J = 14.2 Hz, 3H), 2.21 (d, J = 13.4 Hz, 3H). Procedure 29: Example 148: [0762] 2-(2,6-dimethyl-4-pyridyl)-6-(4-fluoro-3-nitro-phenyl)-3-met hyl-1H-indole: To a vial was added 6-bromo-2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole (I-2) (350 mg, 1.11 mmol), (4-fluoro-3-nitro-phenyl)boronic acid (246 mg, 1.33 mmol), XPhos Pd G2 (46 mg, 0.056 mmol), cesium carbonate (1085 mg, 3.33 mmol), 10:11,4-dioxane and water (5 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, the crude was diluted with ethyl acetate, washed with organic layer with water. The organic layer was filtered through Celite (eluent: EtOAc) and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc/ Hex) to provide the desired product. ES/MS: 376.2 (M+H + ) [0763] tert-butyl 4-[4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-n itro- anilino]piperidine-1-carboxylate.2-(2,6-dimethyl-4-pyridyl)- 6-(4-fluoro-3-nitro-phenyl)-3- methyl-1H-indole (138 mg, 0.37 mmol), tert-butyl 4-aminopiperidine-1-carboxylate (221 mg, 1.10 mmol) and THF (2 mL) was added triethylamine (0.16 mL, 1.10 mmol). The solution was heated at 80 °C for 16 hours. The crude mixture was concentrated and was purified by silica gel column chromatography (eluent: EtOAc/ Hex) to provide the desired product. ES/MS: 556.3 (M+H + ). [0764] tert-butyl 4-[5-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6- yl]benzimidazol-1-yl]piperidine-1-carboxylate. A 40 mL glass vial was charged with tert- butyl 4-[4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-n itro-anilino]piperidine-1- carboxylate (50 mg, 0.09 mmol), iron powder 325 mesh (50.3 mg, 0.090 mmol), NH 4 Cl (50 mg, 0.09 mmol), and 2-PrOH (0.45 ml) and formic acid (0.45 ml) were added. The reaction mixture was stirred at 80 ˚C for 1 h at which time LC-MS analysis indicated that conversion into the product was complete. The reaction mixture was diluted with 2-PrOH (10 mL) and filtered to remove insoluble materials. The filtrate was concentrated to dryness and used directly for the next step. ES/MS: 536.4 (M+H + ). [0765] 5-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-1-(4- piperidyl)benzimidazole (Example 148): To a solution of tert-butyl 4-[5-[2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indol-6-yl]benzimidazol-1-yl]piperidine -1-carboxylate (40 mg, 0.075 mmol) in DCM (2 mL) was added trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temp for 1 hr. The crude was concentrated and purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 148 as a bis-trifluoroacetate salt. ES/MS: 436.6 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 9.11 (s, 1H), 8.98 (s, 1H), 8.09 (d, J = 5.3 Hz, 1H), 8.02 (s, 1H), 7.96 (s, 1H), 7.93 (s, 2H), 7.85 (d, J = 8.5 Hz, 1H), 7.76 (s, 1H), 7.53 (dd, J = 8.5, 1.6 Hz, 1H), 3.72 (d, J = 13.1 Hz, 2H), 3.40 (d, J = 12.6 Hz, 2H), 2.81 (s, 6H), 2.74 (s, 3H), 2.58 (d, J = 13.7 Hz, 2H), 2.45 (d, J = 13.9 Hz, 2H). Procedure 30: Example 149: [0766] tert-butyl 6-bromo-3-methyl-pyrrolo[3,2-b]pyridine-1-carboxylate: To a 100 mL flask was added 6-bromo-3-methyl-1H-pyrrolo[3,2-b]pyridine (900 mg, 4.26 mmol), tert- butoxycarbonyl tert-butyl carbonate (1.12 mg, 5.21 mmol), 4-dimethylaminopyridine (625 mg, 5.12 mmol), and triethylamine (0.89 ml, 6.40 mmol) and the mixture was dissolved in THF (20 mL). The solution was stirred overnight at room temperature. The reaction mixture was concentrated under reduced pressure. The crude residue was concentrated under reduced pressure and was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 312.7 (M+H + ). [0767] tert-butyl 4-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2- pyridyl]piperazine-1-carboxylate: To vial was added tert-butyl 6-bromo-3-methyl-pyrrolo[3,2- b]pyridine-1-carboxylate (309 mg, 0.99 mmol), tert-butyl 4-(5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)piperazine-1-carboxylate (425 mg, 1.09 mmol), XPhos Pd G2 (39 mg, 0.050 mmol), 10: 1 Dioxanes/water (30 mL), and cesium carbonate (971 mg, 2.98 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated under microwave conditions at 110 °C for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 493.9 (M+H + ). [0768] tert-butyl 2-bromo-6-[6-(4-tert-butoxycarbonylpiperazin-1-yl)-3-pyridyl ]-3- methyl-pyrrolo[3,2-b]pyridine-1-carboxylate: To a 100 mL flask was added tert-butyl 6-[6- (4-tert-butoxycarbonylpiperazin-1-yl)-3-pyridyl]-3-methyl-py rrolo[3,2-b]pyridine-1-carboxylate (420 mg, 0.85 mmol) and carbon tetrachloride (5 mL). The solution was cooled to 0 °C, and N- bromosuccinimide (151 mg, 0.85 mmol) dissolved in 5 mL acetonitrile was added to the solution dropwise. The solution was refluxed for 1 hr. The reaction mixture was dry-loaded onto silica, and was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 573.8 (M+H + ). [0769] tert-butyl 6-[6-(4-tert-butoxycarbonylpiperazin-1-yl)-3-pyridyl]-2-(2,6 - dimethyl-4-pyridyl)-3-methyl-pyrrolo[3,2-b]pyridine-1-carbox ylate: To a vial was added tert-butyl 2-bromo-6-[6-(4-tert-butoxycarbonylpiperazin-1-yl)-3-pyridyl ]-3-methyl-pyrrolo[3,2- b]pyridine-1-carboxylate (150 mg, 0.26 mmol), (2,6-dimethyl-4-pyridyl)boronic acid (43.5 mg, 0.29 mmol), XPhos Pd G2 (23 mg, 0.026 mmol), 10: 1 Dioxanes/water (3 mL), and cesium carbonate (971 mg, 2.98 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated under microwave conditions at 110 °C for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 599.2 (M+H + ). [0770] 2-(2,6-dimethyl-4-pyridyl)-3-methyl-6-(6-piperazin-1-yl-3-py ridyl)-1H- pyrrolo[3,2-b]pyridine (Example 149): To a solution of tert-butyl 6-[6-(4-tert- butoxycarbonylpiperazin-1-yl)-3-pyridyl]-2-(2,6-dimethyl-4-p yridyl)-3-methyl-pyrrolo[3,2- b]pyridine-1-carboxylate (43 mg, 0.07 mmol) in MeCN (1 mL) was added trifluoroacetic acid (0.1 mL). The reaction mixture was heated at 40 °C for overnight. To the cooled reaction mixture was added water (0.1 mL), and the mixture was purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 149 as a bis-trifluoroacetate salt. ES/MS: 399.2 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 8.55 (d, J = 8.2 Hz, 1H), 8.50 (d, J = 1.4 Hz, 1H), 8.17 (dd, J = 8.6, 1.4 Hz, 1H), 7.91 – 7.84 (m, 2H), 7.76 (dd, J = 10.5, 8.4 Hz, 2H), 7.59 (dd, J = 7.4, 1.7 Hz, 1H), 7.35 (dd, J = 11.2, 6.0 Hz, 1H), 6.98 (d, J = 8.2 Hz, 1H), 5.68 (s, 2H), 4.77 (t, J = 4.9 Hz, 2H), 4.70 (d, J = 7.6 Hz, 1H), 3.86 – 3.81 (m, 2H), 2.86 (s, 3H), 2.77 (s, 3H).

Procedure 31: Example 150: [0771] 1-(benzenesulfonyl)-6-chloro-2-(2,6-dimethyl-4-pyridyl)pyrro lo[2,3- b]pyridine: To a vial was added 1-(benzenesulfonyl)-6-chloro-2-iodo-pyrrolo[2,3-b]pyridine (960 mg, 2.29 mmol), (2,6-dimethyl-4-pyridyl)boronic acid (381 mg, 2.52 mmol), XPhos Pd G2 (91.4 mg, 0.12 mmol), 10: 1 Dioxanes/water (10 mL), and cesium carbonate (2.24 g, 6.88 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated under microwave conditions at 110 °C for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product.ES/MS: 398.1 (M+H + ). [0772] tert-butyl 4-[5-[1-(benzenesulfonyl)-2-(2,6-dimethyl-4-pyridyl)pyrrolo[ 2,3- b]pyridin-6-yl]-2-pyridyl]piperazine-1-carboxylate: 1-(benzenesulfonyl)-6-chloro-2-(2,6- dimethyl-4-pyridyl)pyrrolo[2,3-b]pyridine (500 mg, 1.26 mmol), tert-butyl 4-(5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine- 1-carboxylate (538 mg, 1.38 mmol) 4-ditert-butylphosphanyl-N,N-dimethyl-aniline;dichloropallad ium (89 mg, 0.13 mmol) and sodium carbonate (308 mg, 3.25 mmol) were added in 10: 1 CAN/water (5 ml). Nitrogen was bubbled through the reaction mixture for 3 min and the reaction was heated to 140 °C in a microwave for 30 min. The reaction mixture was filtered through celite and sodium sulfate. The reaction mixture was dry-loaded onto silica, and was purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 493.9 (M+H + ). [0773] tert-butyl 4-[5-[1-(benzenesulfonyl)-3-bromo-2-(2,6-dimethyl-4- pyridyl)pyrrolo[2,3-b]pyridin-6-yl]-2-pyridyl]piperazine-1-c arboxylate: To a 50 mL flask was added tert-butyl 4-[5-[1-(benzenesulfonyl)-2-(2,6-dimethyl-4-pyridyl)pyrrolo[ 2,3-b]pyridin- 6-yl]-2-pyridyl]piperazine-1-carboxylate (400 mg, 0.64 mmol) and carbon tetrachloride (10 mL). The solution was cooled to 0 °C, and N-bromosuccinimide 125 mg, 0.70 mmol) dissolved in 5 mL acetonitrile was added to the solution dropwise. The solution was refluxed for 1 hr. The reaction mixture was concentrated and used directly on the next step. ES/MS: 703.2 (M+H + ). [0774] tert-butyl 4-[5-[1-(benzenesulfonyl)-2-(2,6-dimethyl-4-pyridyl)-3-methy l- pyrrolo[2,3-b]pyridin-6-yl]-2-pyridyl]piperazine-1-carboxyla te: To a vial was added tert- butyl 4-[5-[1-(benzenesulfonyl)-3-bromo-2-(2,6-dimethyl-4-pyridyl) pyrrolo[2,3-b]pyridin-6-yl]- 2-pyridyl]piperazine-1-carboxylate (230 mg, 0.33 mmol), bis(trimethylaluminum)-1,4- diazabicyclo[2.2.2]octane adduct (168 mg, 0.65 mmol), XPhos Pd G2 (23 mg, 0.026 mmol), and THF (3 mL). The solution was degassed by bubbling argon for 30 seconds, then heated at 80 °C for 1 hr. The crude was diluted with ethyl acetate, filtered and washed with excess EA. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 639.3 (M+H + ). [0775] 2-(2,6-dimethyl-4-pyridyl)-3-methyl-6-(6-piperazin-1-yl-3-py ridyl)-1H- pyrrolo[3,2-b]pyridine): To a solution of tert-butyl 4-[5-[1-(benzenesulfonyl)-2-(2,6-dimethyl- 4-pyridyl)-3-methyl-pyrrolo[2,3-b]pyridin-6-yl]-2-pyridyl]pi perazine-1-carboxylate (140 mg, 0.22 mmol) in THF (1 mL) was added tetra-n-butylammonium fluoride (287 mg.1.1 mmol). The reaction mixture was stirred at room temp for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 499.2 (M+H + ). [0776] 2-(2,6-dimethyl-4-pyridyl)-3-methyl-6-(6-piperazin-1-yl-3-py ridyl)-1H- pyrrolo[3,2-b]pyridine (Example 150): To a solution of 2-(2,6-dimethyl-4-pyridyl)-3-methyl- 6-(6-piperazin-1-yl-3-pyridyl)-1H-pyrrolo[2,3-b]pyridine (70 mg, 0.14 mmol) in DCM (1 mL) was added trifluoroacetic acid (0.1 mL). The reaction mixture was heated at 40 °C for 1 hr. To the cooled reaction mixture was added water (0.1 mL), and the mixture was purified by RP- HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 150 as a bis- trifluoroacetate salt. ES/MS: 399.2 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 8.95 (d, J = 2.4 Hz, 1H), 8.38 (dd, J = 2.4, 0.8 Hz, 1H), 8.20 (d, J = 8.4 Hz, 1H), 8.09 (s, 2H), 7.77 (d, J = 8.4 Hz, 1H), 7.57 (s, 1H), 3.51 (dd, J = 6.8, 3.7 Hz, 4H), 3.47 – 3.43 (m, 4H), 2.79 (s, 6H), 2.47 (s, 3H). [0777] The following Example was made in an analogous fashion according to Procedure 31 and is shown below in Table 18. To prepare the below Example, different reagents/starting materials were used than some of those described in Procedure 31 and are noted in the last column of Table 18 – “Changes to Procedure 31: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 31 were replaced with the different reagents/starting materials noted below. Table 18. Example 151 Procedure 32: Example 152: [0778] Tert-butyl 4-(1H-pyrrolo[2,3-b]pyridin-6-yl)-3,6-dihydro-2H-pyridine-1- carboxylate: To 500 ml RBF was added 6-bromo-1H-pyrrolo[2,3-b]pyridine (2000 mg, 10.2 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro- 2H-pyridine-1- carboxylate (3453 mg, 11.2 mmol), XPhos Pd G2 (399 mg, 0.51 mmol), 10: 1 Dioxanes/water (364 mL), and cesium carbonate (8268 g, 25.4 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated under microwave conditions at 110 °C for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 299 (M+H + ). [0779] Tert-butyl 4-(1H-pyrrolo[2,3-b]pyridin-6-yl)piperidine-1-carboxylate: tert- butyl 4-(1H-pyrrolo[2,3-b]pyridin-6-yl)-3,6-dihydro-2H-pyridine-1- carboxylate (2800 mg, 9.35 mmol), Pd/C and H2 in THF (50 ml) were added to a flask and stirred at room temperature overnight. The reaction mixture was filtered through celite and used directly for the next step. ES/MS: 302.2 (M+H + ). [0780] Tert-butyl 4-(3-bromo-1H-pyrrolo[2,3-b]pyridin-6-yl)piperidine-1- carboxylate: To a 50 mL flask was added tert-butyl 4-(1H-pyrrolo[2,3-b]pyridin-6- yl)piperidine-1-carboxylate (2600 mg, 8.63 mmol) and THF (300 mL). The solution was cooled to 0 °C, and N-bromosuccinimide (1748 mg, 9.82 mmol) dissolved in 30 mL THF was added to the solution dropwise. The solution was stirred at RT for 1 hr. The reaction mixture was concentrated and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 379.9 (M+H + ). [0781] Tert-butyl 4-[1-(benzenesulfonyl)-3-bromo-pyrrolo[2,3-b]pyridin-6- yl]piperidine-1-carboxylate: To 500 ml RBF was added tert-butyl 4-(3-bromo-1H-pyrrolo[2,3- b]pyridin-6-yl)piperidine-1-carboxylate (2700 mg, 7.10 mmol) and THF (200 ml), and then sodium hydride (544 mg, 14.2 mmol) was added portions wise to the reaction mixture at 0 °C. The solution was warmed to room temp and further stirred for 30 mins. Then, benzenesulfonyl chloride (1.1 ml, 8.52 mmol) was added and the reaction was stirred at room temp overnight. The crude was diluted with ethyl acetate and washed with brine. The organic layer was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 519.6 (M+H + ). [0782] Tert-butyl 4-[5-[1-(benzenesulfonyl)-2-(2,6-dimethyl-4-pyridyl)-3-methy l- pyrrolo[2,3-b]pyridin-6-yl]-2-pyridyl]piperazine-1-carboxyla te: To 250 ml RBF was added tert-butyl 4-[1-(benzenesulfonyl)-3-bromo-pyrrolo[2,3-b]pyridin-6-yl]pi peridine-1-carboxylate (2.20 g, 4.23 mmol), bis(trimethylaluminum)-1,4-diazabicyclo[2.2.2]octane adduct (2.20, 8.45 mmol), XPhos Pd G2 (166 mg, 0.21 mmol), and THF (44 mL). The solution was degassed by bubbling argon for 30 seconds, and then heated at 80 °C for 1 hr. The crude was diluted with ethyl acetate, filtered and washed with excess EA. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 455.8 (M+H + ). [0783] Tert-butyl 4-[1-(benzenesulfonyl)-2-bromo-3-methyl-pyrrolo[2,3-b]pyridi n- 6-yl]piperidine-1-carboxylate: To a 200 mL flask was added tert-butyl 4-[1-(benzenesulfonyl)- 3-methyl-pyrrolo[2,3-b]pyridin-6-yl]piperidine-1-carboxylate (1830 mg, 4.02 mmol) and THF (100 mL). The solution was cooled to 0 °C, and N-bromosuccinimide (858 mg, 4.82 mmol) dissolved in 30 mL THF was added to the solution dropwise. The solution was refluxed for 1 hr. The reaction mixture was concentrated and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 535.7 (M+H + ). [0784] Tert-butyl 4-(2-bromo-3-methyl-1H-pyrrolo[2,3-b]pyridin-6-yl)piperidine -1- carboxylate: To a solution of tert-butyl 4-[1-(benzenesulfonyl)-2-bromo-3-methyl-pyrrolo[2,3- b]pyridin-6-yl]piperidine-1-carboxylate (700 mg, 1.31 mmol) in THF (5 mL) was added tetra-n- butylammonium fluoride (2.05 g.7.86 mmol). The reaction mixture was stirred at room temp for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 395.8 (M+H + ). [0785] 1-(benzenesulfonyl)-6-chloro-2-(2,6-dimethyl-4-pyridyl)pyrro lo[2,3- b]pyridine: To a vial was added 1-(benzenesulfonyl)-6-chloro-2-iodo-pyrrolo[2,3-b]pyridine (960 mg, 2.29 mmol), (2,6-dimethyl-4-pyridyl)boronic acid (381 mg, 2.52 mmol), XPhos Pd G2 (91.4 mg, 0.12 mmol), 10: 1 Dioxanes/water (10 mL), and cesium carbonate (2.24 g, 6.88 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated under microwave conditions at 110 °C for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 499.2 (M+H + ). [0786] 1-(benzenesulfonyl)-6-chloro-2-(2,6-dimethyl-4-pyridyl)pyrro lo[2,3- b]pyridine: To a vial was added tert-butyl 4-(2-bromo-3-methyl-1H-pyrrolo[2,3-b]pyridin-6- yl)piperidine-1-carboxylate (140 mg, 0.36 mmol), (2,6-dimethyl-4-pyridyl)boronic acid (59.0mg, 0.39 mmol), XPhos Pd G2 (28 mg, 0.036 mmol), 10: 1 Dioxanes/water (1 mL), and cesium carbonate (347 g, 1.1 mmol). The solution was degassed by bubbling argon for 30 seconds, then heated under microwave conditions at 110 °C for 1 hr. The crude reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc / Hex) to provide the desired product. ES/MS: 421.2 (M+H + ). [0787] 2-(2,6-dimethyl-4-pyridyl)-3-methyl-6-(4-piperidyl)-1H-pyrro lo[2,3- b]pyridine (Example 152): To a solution of tert-butyl 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl- 1H-pyrrolo[2,3-b]pyridin-6-yl]piperidine-1-carboxylate (100 mg, 0.24 mmol) in DCM (1 mL) was added trifluoroacetic acid (0.1 mL). The reaction mixture was heated at 40 °C for 1 hr. To the cooled reaction mixture was added water (0.1 mL), and the mixture was purified by RP- HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 152 as a bis- trifluoroacetate salt. ES/MS: 321.2 (M+H + ). 1 H NMR (400 MHz, MeOD) δ 8.11 (d, J = 8.2 Hz, 1H), 7.93 (s, 2H), 7.18 (d, J = 8.3 Hz, 1H), 3.56 (dt, J = 13.5, 3.5 Hz, 2H), 3.26 – 3.16 (m, 3H), 2.80 (s, 6H), 2.68 (s, 3H), 2.28 – 2.13 (m, 4H). [0788] The following Examples were made in an analogous fashion according to Procedure 32 and is shown below in Table 19. To prepare the below Example, different reagents/starting materials were used than some of those described in Procedure 32 and are noted in the last column of Table 19 – “Changes to Procedure 32: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 32 were replaced with the different reagents/starting materials noted below. Table 19. Example 153 Procedure 33: Example 154: [0789] 3-cyclopropyl-2-(2,6-dimethylpyridin-4-yl)-6-(6-(6-(methylsu lfonyl)-2,6- diazaspiro[3.3]heptan-2-yl)pyridin-3-yl)-1H-indole (Example 154): To a solution of 3- cyclopropyl-2-(2,6-dimethylpyridin-4-yl)-6-(6-fluoropyridin- 3-yl)-1H-indole (I-19) (40.0 mg, 0.112 mmol) in DMSO (0.5 mL) is added triethylamine (0.078 mL, 0.56 mmol) and 2- methylsulfonyl-2,6-diazaspiro[3.3]heptane (79 mg, 0.448 mmol) and the reaction mixture is heated in a microwave to 150 °C for 4 h. Trifluoroacetic acid (0.2 mL) is added and the reaction mixture is purified directly by RP-HPLC (0.1% TFA-ACN in 0.1% TFA-Water) to give the title compound. Procedure 34: Example 155: [0790] N-(1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl )pyridin-2- yl)piperidin-4-yl)-1-(oxetan-3-yl)piperidine-4-carboxamide (Example 155): To a solution of 1-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)py ridin-2-yl)piperidin-4-amine (Example 2) (40 mg, 0.097 mmol) in DCM (0.75 mL) is added 1-(oxetan-3-yl)piperidine-4- carboxylic acid (27 mg, 0.146 mmol), 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate (HATU) (44 mg, 0.117 mmol) and N,N- diisopropylethylamine (0.085 mL, 0.49 mmol) and the reaction mixture is stirred for 30 min. Trifluoroacetic acid (0.25 mL) is added, and the reaction mixture is concentrated in vacuo and the crude residue is purified directly by RP-HPLC (0.1% TFA-ACN in 0.1% TFA-Water) to give the title compound. Procedure 35: Example 156: [0791] (7-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)p yridin-2-yl)-4,7- diazaspiro[2.5]octan-4-yl)(1H-thieno[2,3-d]imidazol-5-yl)met hanone (Example 156): To a solution of 6-(6-(4,7-diazaspiro[2.5]octan-7-yl)pyridin-3-yl)-2-(2,6-dim ethylpyridin-4-yl)-3- methyl-1H-indole (Example 22) (20 mg, 0.047 mmol) in DCM (0.75 mL) is added 1H- thieno[2,3-d]imidazole-5-carboxylic acid (12 mg, 0.071 mmol), 2-(7-Aza-1H-benzotriazole-1- yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (21.5 mg, 0.057 mmol) and N,N- diisopropylethylamine (0.041 mL, 0.236 mmol) and the reaction mixture is stirred for 30 min. Trifluoroacetic acid (0.25 mL) is added, and the reaction mixture is concentrated in vacuo and the crude residue is purified directly by RP-HPLC (0.1% TFA-ACN in 0.1% TFA-Water) to give the title compound. Procedure 36: Example 157: [0792] tert-butyl 3-((4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6- yl)pyridin-2-yl)piperazin-1-yl)sulfonyl)pyrrolidine-1-carbox ylate: To a solution of 2-(2,6- dimethylpyridin-4-yl)-3-methyl-6-(6-(piperazin-1-yl)pyridin- 3-yl)-1H-indole (Example 1) (40.0 mg, 0.10 mmol) in DCM (0.5 mL) is added N,N-diisopropylethylamine (0.07 mL, 0.4 mmol) and tert-butyl 3-(chlorosulfonyl)pyrrolidine-1-carboxylate (30 mg, 0.111 mmol) and the reaction mixture is stirred at room temperature for 4 h. The crude mixture is dry-loaded onto silica, and the crude material is purified by silica chromatography (Eluent: EtOAc / hexanes) to provide the desired product. [0793] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-6-(6-(4-(pyrrolidin-3- ylsulfonyl)piperazin-1-yl)pyridin-3-yl)-1H-indole: To a solution of tert-butyl 3-((4-(5-(2-(2,6- dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)pyridin-2-yl)pi perazin-1-yl)sulfonyl)pyrrolidine- 1-carboxylate (20 mg, 0.035 mmol) in DCM (1.0 mL) is added 4.0 N HCl in dioxane (0.11 mL, 0.44 mmol) and the reaction mixture is stirred for 2 h at room temperature. The reaction mixture is concentrated in vacuo to give the title compound as an HCl salt. [0794] 2-(3-((4-(5-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol- 6-yl)pyridin-2- yl)piperazin-1-yl)sulfonyl)pyrrolidin-1-yl)ethan-1-ol (Example 157): To a suspension of 2- (2,6-dimethylpyridin-4-yl)-3-methyl-6-(6-(4-(pyrrolidin-3-yl sulfonyl)piperazin-1-yl)pyridin-3- yl)-1H-indole hydrochloride (20 mg, 0.035 mmol) in THF (0.1 mL) is added N,N- diisopropylethylamine (0.06 mL, 0.35 mmol) and ethylene oxide (2M solution in THF, 0.17 mL, 0.35 mmol) and the reaction mixture is heated in a sealed vial at 50 °C for 48 h. The reaction mixture is concentrated in vacuo, acetonitrile (0.2 mL) is added, and trifluoroacetic acid (0.25 mL) is added, and the mixture is purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA-Water) to give the title compound. [0795] The following Examples were made in an analogous fashion according to the listed procedure and are shown below in Table 20. To prepare the below Examples, different reagents/starting materials were used than some of those described in the listed procedure and are noted in the last column of Table 20 – “Changes to Procedure: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of the listed procedure were replaced with the different reagents/starting materials noted below. Table 20. Examples 158 to 176

Procedure 37, Example 177 [0796] 2-(4-(5-(3-ethyl-2-(8-methyl-[1,2,4]triazolo[1,5-a]pyridin-6 -yl)-1H-indol-6- yl)pyridin-2-yl)piperazin-1-yl)acetamide (Example 177): To a solution of 6-(3-ethyl-6-(6- (piperazin-1-yl)pyridin-3-yl)-1H-indol-2-yl)-8-methyl-[1,2,4 ]triazolo[1,5-a]pyridine (bis-HCl salt) (Example 167) (25 mg, 0.049 mmol) in 1,2-dichloroethane (1 mL) was added 2- bromoacetamide (8.1 mg, 0.059 mmol) and 1,8-diazabicyclo[5.4.0]undec-7-ene (0.03 mL, 0.2 mmol). The mixture was stirred overnight at rt. To the mixture was added 0.5 mL TFA, and the DCE was subsequently removed under reduced pressure. Acetonitrile (0.5 mL) and water (0.2 mL) were added, and the mixture was filtered through an acrodisc before purification by RP- HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 177. ES/MS: 495.3 (M+H + ). 1H NMR (400 MHz, DMSO-d6) δ 11.40 (s, 1H), 10.19 – 9.93 (m, 1H), 8.94 (s, 1H), 8.56 – 8.51 (m, 2H), 8.02 – 7.94 (m, 2H), 7.75 (s, 2H), 7.68 (d, J = 8.3 Hz, 1H), 7.56 (s, 1H), 7.34 (d, J = 8.3 Hz, 1H), 7.06 (d, J = 8.9 Hz, 1H), 4.51 – 4.31 (m, 2H), 4.00 (s, 2H), 3.29 (d, J = 39.4 Hz, 6H), 2.93 (q, J = 7.5 Hz, 2H), 2.66 (s, 3H), 1.29 (t, J = 7.5 Hz, 3H). Procedure 38, Example 178: [0797] 2-(2,6-dimethylpyridin-4-yl)-3-methyl-N-(piperidin-4-yl)-1H- indole-6- carboxamide (Example 178): To a dram vial with 2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H- indole-6-carboxylic acid (I-34) (20 mg, 0.071 mmol) in dichloromethane (0.75 mL) was added tert-butyl 4-aminopiperidine-1-carboxylate (15.7 mg, 0.079 mmol), 2-(7-Aza-1H-benzotriazole- 1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (32.6 mg, 0.086 mmol), N,N- diisopropylethylamine (0.06 mL, 0.36 mmol). The mixture was stirred 0.5 hr at rt. To the mixture was added TFA (0.25 mL), and the mixture was stirred 10 minutes at rt. The crude mixture was concentrated under reduced pressure, and acetonitrile (0.7 mL) and water (0.15 mL) were added. The mixture was filtered through an acrodisc before purification by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 178. ES/MS: 363.2 (M+H + ). 1H NMR (400 MHz, DMSO-d6) δ 11.88 (s, 1H), 8.60 – 8.51 (m, 1H), 8.49 (d, J = 7.4 Hz, 1H), 8.37 – 8.21 (m, 1H), 7.95 (s, 1H), 7.87 – 7.77 (m, 1H), 7.74 (d, J = 8.4 Hz, 1H), 7.62 (dd, J = 8.5, 1.5 Hz, 1H), 4.18 – 4.02 (m, 1H), 3.36 – 3.31 (m, 2H), 3.13 – 2.98 (m, 2H), 2.68 (s, 6H), 2.59 (s, 3H), 2.07 – 1.96 (m, 2H), 1.85 – 1.68 (m, 2H). [0798] The following Examples were made in an analogous fashion according to Procedure 38 and are shown below in Table 21. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 38 and are noted in the last column of Table 21 – “Changes to Procedure 38: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 38 were replaced with the different reagents/starting materials noted below. Table 21. Examples 179 and 180 Procedure 39: Example 181: [0799] 3-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-5,6,7, 8-tetrahydro-1,7- naphthyridine: To a vial was added tert-butyl 2-(2,6-dimethyl-4-pyridyl)-3-methyl-6-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)indole-1-carboxylate (I-25) (50.1 mg, 0.11 mmol), 3- bromo-5,6,7,8-tetrahydro-1,7-naphthyridine;hydrochloride (43.2 mg, 0.17 mmol), XPhos Pd G2 (8.5 mg, 0.011 mmol), cesium carbonate (176 mg, 0.54 mmol), and a 10:1 mixture of 1,4- dioxane and water (2 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, the crude was diluted with ethyl acetate, washed with organic layer with water. The organic layer filtered through celite (rinsing with EtOAc) and concentrated under reduced pressure. The crude residue was dissolved in DCM (1 mL), and trifluoroacetic acid (0.5 mL) was added. The reaction mixture was heated at 60 °C for 1hr. To the cooled reaction mixture was added water (0.1 mL), and the mixture was purified by RP-HPLC (eluent: water / MeCN * 0.1% TFA) to provide the product Example 181. ES/MS: 369.2 (M+H + ). 1H NMR (400 MHz, MeOD) δ 8.83 (d, J = 2.2 Hz, 1H), 8.08 (d, J = 2.1 Hz, 1H), 7.94 (s, 2H), 7.86 (d, J = 8.5 Hz, 1H), 7.76 (d, J = 1.6 Hz, 1H), 7.49 (dd, J = 8.4, 1.6 Hz, 1H), 4.49 (s, 2H), 3.64 (t, J = 6.3 Hz, 2H), 3.29 (t, J = 6.3 Hz, 2H), 2.81 (s, 6H), 2.73 (d, J = 1.9 Hz, 3H). [0800] The following Examples were made in an analogous fashion according to Procedure 39 and are shown below in Table 22 To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 39 and are noted in the last column of Table 22 – “Changes to Procedure 39: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 39 were replaced with the different reagents/starting materials noted below. Table 22. Examples 182 to 184 Procedure 40: Example 185: [0801] tert-butyl 6-[2-(4-tert-butoxycarbonylpiperazin-1-yl)thiazol-4-yl]-2-(2 ,6- dimethyl-4-pyridyl)-3-methyl-indole-1-carboxylate: To a vial was added tert-butyl 2-(2,6- dimethyl-4-pyridyl)-3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)indole-1- carboxylate (I-25) (270 mg, 0.58 mmol), 2-bromo-4-chloro-thiazole (139 mg, 0.71 mmol), XPhos Pd G2 (45.9 mg, 0.058 mmol), cesium carbonate (571 mg, 1.75 mmol), and a 10:1 mixture of 1,4-dioxane and water (2 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, filtered through celite (rinsing with EtOAc), and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc/ Hex) to provide the desired product. ES/MS: 454.2 (M+H + ). [0802] 2-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-4-(p iperazin-1- yl)thiazole: To a vial was added tert-butyl 6-(4-chlorothiazol-2-yl)-2-(2,6-dimethyl-4-pyridyl)- 3-methyl-indole-1-carboxylate (54 mg, 0.12 mmol), tert-butyl piperazine-1-carboxylate (26.2 mg, 0.14 mmol), RuPhos Pd G3 (8.5 mg, 0.011 mmol), cesium carbonate (70.5 mg, 0.22 mmol), 1,4-dioxane (2 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 2 h. The reaction mixture was cooled, filtered through Celite (eluent: EtOAc), and concentrated. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc/ Hex). The isolated material was dissolved in DCM (1 mL) and trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temp for 1 hr. The crude mixture was concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 185. ES/MS: 404.2 (M+H + ). 1H NMR (400 MHz, MeOD) δ 8.03 (dd, J = 1.5, 0.8 Hz, 1H), 7.93 (s, 2H), 7.82 – 7.71 (m, 3H), 3.66 (dd, J = 6.6, 3.9 Hz, 4H), 3.44 (dd, J = 6.5, 4.0 Hz, 4H), 2.81 (s, 6H), 2.71 (s, 3H). Procedure 41: Example 186: [0803] tert-butyl 4-[2-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]thia zol-4- yl]-3,6-dihydro-2H-pyridine-1-carboxylate: To a vial was tert-butyl 6-(4-chlorothiazol-2-yl)- 2-(2,6-dimethyl-4-pyridyl)-3-methyl-indole-1-carboxylate (synthesized identically as in Procedure W-2) (50 mg, 0.11 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 3,6-dihydro-2H-pyridine-1-carboxylate (43 mg, 0.14 mmol), RuPhos Pd G2 (7.8 mg, 0.01 mmol), cesium carbonate (107 mg, 0.33 mmol), and a 10:1 mixture of 1,4-dioxane and water (2 mL). The solution was degassed by bubbling argon for 30 seconds, then heated in a sealed tube at 110 °C for 4 h. The reaction mixture was cooled, filtered through celite (rinsing with EtOAc), and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: EtOAc/ Hex) to provide the product. ES/MS: 601.2 (M+H + ). [0804] 2-(2-(2,6-dimethylpyridin-4-yl)-3-methyl-1H-indol-6-yl)-4-(p iperidin-4- yl)thiazole: To a solution of tert-butyl 6-[4-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4- yl)thiazol-2-yl]-2-(2,6-dimethyl-4-pyridyl)-3-methyl-indole- 1-carboxylate (24 mg, 0.005 mmol) in ethanol (1 mL) and ethyl acetate (1 mL), nitrogen was bubbled through the solution for 4 min, then palladium on carbon 10 wt. % (12 mg, 0.001 mmol) was added and the reaction mixture was stirred for 5 h under an atmosphere of hydrogen. The reaction mixture degassed with argon, diluted with EtOAc, and filtered through celite, rinsing with EtOAc. The filtrate was concentrated under reduced pressure to give the crude product, which was dissolved in DCM (1 mL) and trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temperature for 1 hr. The crude mixture was concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 186. ES/MS: 403.2 (M+H+). 1H NMR (400 MHz, MeOD) δ 8.06 (dd, J = 1.5, 0.8 Hz, 1H), 7.94 (s, 2H), 7.79 (qd, J = 8.5, 1.2 Hz, 2H), 7.31 (d, J = 0.8 Hz, 1H), 3.56 (d, J = 12.8 Hz, 2H), 3.26 – 3.18 (m, 3H), 2.81 (s, 6H), 2.72 (s, 3H), 2.38 (d, J = 12.7 Hz, 2H), 2.12 – 2.01 (m, 2H). [0805] tert-butyl 4-[2-amino-4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6 - yl]anilino]piperidine-1-carboxylate: To a solution of tert-butyl 4-[4-[2-(2,6-dimethyl-4- pyridyl)-3-methyl-1H-indol-6-yl]-2-nitro-anilino]piperidine- 1-carboxylate (synthesized identically as in Procedure 29) (130 mg, 0.23 mmol) in ethanol (10 mL) and ethyl acetate (2 mL), nitrogen was bubbled through the solution for 4 minutes, then palladium on carbon (10 wt.%, 24.9 mg, 0.23 mmol) was added and the reaction mixture was stirred for 5 h under an atmosphere of hydrogen. The reaction mixture degassed with argon, diluted with EtOAc, and filtered through celite, rinsing with EtOAc. The filtrate was concentrated under reduced pressure to give the product which was used directly in the next step. ES/MS: 526.3 (M+H + ) [0806] 6-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-3-(4-p iperidyl)-1H- benzimidazol-2-one: To a solution of tert-butyl 4-[2-amino-4-[2-(2,6-dimethyl-4-pyridyl)-3- methyl-1H-indol-6-yl]anilino]piperidine-1-carboxylate (48.4 mg, 0.091 mmol) and CDI (37.3, 0.23 mmol) was added dioxane (1 mL), and the reaction mixture was stirred for 20 h at 130 °C. The reaction mixture was diluted with EtOAc, and filtered through celite, rinsing with EtOAc. The filtrate was concentrated under reduced pressure, and the crude residue was dissolved in DCM (1 mL) and trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temperature for 1 hr. The crude mixture was concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 187. ES/MS: 452.3 (M+H + ). 1H NMR (400 MHz, MeOD) δ 8.16 – 7.97 (m, 1H), 7.90 (s, 1H), 7.78 (d, J = 8.5 Hz, 1H), 7.67 – 7.63 (m, 1H), 7.54 – 7.36 (m, 4H), 4.62 (tt, J = 12.1, 4.0 Hz, 1H), 3.63 (d, J = 13.0 Hz, 2H), 3.31 – 3.21 (m, 2H), 2.80 (d, J = 5.6 Hz, 7H), 2.73 (s, 3H), 2.18 – 2.10 (m, 2H), 1.33 (s, 2H). Procedure 43: Example 188: [0807] 5-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-3-pipe razin-1-yl-1,2,4- oxadiazole: To a solution of 2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole-6-carboxylic acid (I-34) (66 mg, 0.23 mmol), and CDI (37.3, 0.23 mmol) in dioxane (1 mL) and the reaction mixture was stirred for 2 h at 100 °C. Then, tert-butyl 4-[(E)-N'- hydroxycarbamimidoyl]piperazine-1-carboxylate (86.3 mg, 0.35 mmol) added to the reaction mixture and stirred for 4 h at 100 C The reaction mixture was diluted with EtOAc, and filtered through celite, eluting with EtOAc and the filtrate was concentrated under reduced pressure. The crude residue was dissolved in DCM (1 mL) and trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temperature for 1 hr. The crude mixture was concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP- HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 188. ES/MS: 389.3 (M+H + ). 1H NMR (400 MHz, MeOD) δ 8.21 (t, J = 1.1 Hz, 1H), 7.96 (s, 2H), 7.87 (qd, J = 8.5, 1.1 Hz, 2H), 3.86 – 3.79 (m, 4H), 3.44 – 3.37 (m, 4H), 2.82 (s, 6H), 2.70 (s, 3H). [0808] The following Examples were made in an analogous fashion according to Procedure 43 and are shown below in Table 23. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 43 and are noted in the last column of Table 23 – “Changes to Procedure 43: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 43 were replaced with the different reagents/starting materials noted below. Table 23. Examples 189 Procedure 44: Example 190: [0809] 2-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-5-(4-p iperidyl)-1,3,4- oxadiazole (Example 190): To a solution of 2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indole-6- carboxylic acid (I-34) (60.0 mg, 0.21 mmol) in THF (2 mL) was added tert-butyl 4- (hydrazinecarbonyl)piperidine-1-carboxylate (57.3 mg, 0.24 mmol), 2-(7-Aza-1H-benzotriazole- 1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (60.4 mg, 0.26 mmol) and N,N- diisopropylethylamine (0.075 mL, 0.43 mmol) and the reaction mixture was stirred for 1h. Subsequently, Burgess reagent (265 mg, 1.1 mmol) was added to the reaction mixture and the reaction mixture was stirred at room temperature for 3 hr. The reaction mixture was diluted with EtOAc, and filtered through celite, eluting with EtOAc. The filtrate was concentrated under reduced pressure to give the crude product which was used directly in the next step. The crude residue was dissolved in DCM (1 mL) and trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temperature for 1 hr. The crude mixture was concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 190. ES/MS: 399.3 (M+H + ). 1H NMR (400 MHz, MeOD) δ 8.18 (dd, J = 1.5, 0.7 Hz, 1H), 7.97 (s, 2H), 7.93 – 7.89 (m, 1H), 7.83 (dd, J = 8.5, 1.5 Hz, 1H), 3.55 (ddt, J = 14.0, 10.4, 4.1 Hz, 3H), 3.31 – 3.22 (m, 2H), 2.83 (s, 6H), 2.72 (s, 3H), 2.53 – 2.44 (m, 2H), 2.19 (dtd, J = 14.7, 10.9, 4.0 Hz, 2H). [0810] The following Examples were made in an analogous fashion according to Procedure 44 and are shown below in Table 24. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 44 and are noted in the last column of Table 24 – “Changes to Procedure 44: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 44 were replaced with the different reagents/starting materials noted below. Table 24. Examples 191 and 192 Procedure 45: Example 193: [0811] 6-[6-(1H-benzimidazol-2-yl)-3-methyl-1H-indol-2-yl]-8-methyl - [1,2,4]triazolo[1,5-a]pyridine (Example 193): To a solution of 3-methyl-2-(8-methyl- [1,2,4]triazolo[1,5-a]pyridin-6-yl)-1H-indole-6-carboxylic acid (I-35) (50.0 mg, 0.16 mmol) in THF (2 mL) was added benzene-1,2-diamine (17.7 mg, 0.16 mmol), 2-(7-Aza-1H-benzotriazole- 1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (75.5 mg, 0.20 mmol) and N,N- diisopropylethylamine (0.085 mL, 0.49 mmol) and the reaction mixture was stirred for 1h. The reaction was concentrated in vacuo, taken up in EtOAc and washed with water (1x) and brine (1x). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was dissolved in AcOH (1 mL) and the reaction mixture was heated at 100 °C for 16 h. The crude reaction was subsequently concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 193. ES/MS: 379.2 (M+H + ). 1H NMR (400 MHz, MeOD) δ 9.00 (d, J = 1.7 Hz, 1H), 8.52 (s, 1H), 8.27 (d, J = 1.6 Hz, 1H), 7.95 (d, J = 8.4 Hz, 1H), 7.88 (t, J = 1.4 Hz, 1H), 7.86 – 7.81 (m, 3H), 7.64 (dd, J = 6.1, 3.2 Hz, 2H), 2.76 (d, J = 0.9 Hz, 3H), 2.59 (s, 3H). [0812] The following Examples were made in an analogous fashion according to Procedure 45 and are shown below in Table 25. To prepare the below Examples, different reagents/starting materials were used than some of those described in Procedure 45 and are noted in the last column of Table 25 – “Changes to Procedure 45: Different Reagents/Starting Materials”. A person of ordinary skill in the art will readily recognize which reagents/starting materials of Procedure 45 were replaced with the different reagents/starting materials noted below. Table 25. Example 194 Procedure 46: Example 195: E xample 195 [0813] 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]benzene -1,2-diamine: To a solution of 4-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]-2-nitr o-aniline (synthesized identically as in Procedure 28) (490 mg, 1.32 mmol) in ethanol (10 mL) and ethyl acetate (2 mL), nitrogen was bubbled through the solution for 4 minutes before addition of palladium on carbon (10 wt. %, 152 mg, 1.43 mmol). The reaction mixture was stirred for 5 h under a hydrogen atmosphere. The reaction mixture was subsequently degassed with argon, diluted with EtOAc, and filtered through celite, rinsing with EtOAc. The filtrate was concentrated under reduced pressure to give the crude product which was used directly in the next step. ES/MS: 343.2 (M+H + ). [0814] tert-butyl (1S,3R,4R)-3-[6-[2-(2,6-dimethyl-4-pyridyl)-3-methyl-1H-indo l-6- yl]-1H-benzimidazol-2-yl]-2-azabicyclo[2.2.1]heptane-2-carbo xylate: To a solution of 4-[2- (2,6-dimethyl-4-pyridyl)-3-methyl-1H-indol-6-yl]benzene-1,2- diamine (66.7 mg, 0.20 mmol) in THF (2 mL) was added (1S,3R,4R)-2-tert-butoxycarbonyl-2-azabicyclo[2.2.1]heptane- 3- carboxylic acid (56.4 mg, 0.23 mmol), 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate (HATU) (96.2 mg, 0.25 mmol) and N,N- diisopropylethylamine (0.068 mL, 0.39 mmol) and the reaction mixture was stirred for 1h. The reaction was subsequently concentrated in vacuo, taken up in EtOAc and washed with water (1x) and brine (1x). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude residue was dissolved in AcOH (1 mL) and the reaction mixture was heated at 100 °C for 16 h. The crude reaction was subsequently concentrated under reduced pressure, and taken directly forward. ES/MS: 548.4 (M+H+). [0815] 2-[(1S,3R,4R)-2-azabicyclo[2.2.1]heptan-3-yl]-6-[2-(2,6-dime thyl-4-pyridyl)- 3-methyl-1H-indol-6-yl]-1H-benzimidazole: To tert-butyl (1S,3R,4R)-3-[6-[2-(2,6-dimethyl- 4-pyridyl)-3-methyl-1H-indol-6-yl]-1H-benzimidazol-2-yl]-2-a zabicyclo[2.2.1]heptane-2- carboxylate (50 mg, 0.23 mmol) dissolved in DCM (1 mL) was added trifluoroacetic acid (0.2 mL). The reaction mixture was stirred at room temperature for 1 hr. The crude reaction was subsequently concentrated under reduced pressure, dissolved in acetonitrile (0.75 mL) and water (0.25 mL), and purified by RP-HPLC (0.1% TFA-ACN in 0.1% TFA Water, Column: Gemini 5 uM, NX-C18110 Angstrom, 250 x 21.2 mm) to give the title compound Example 195. ES/MS: 448.3 (M+H + ). Multiplet Report 1H NMR (400 MHz, MeOD) δ 7.93 – 7.88 (m, 3H), 7.79 (d, J = 8.4 Hz, 1H), 7.74 – 7.70 (m, 2H), 7.67 (dd, J = 8.5, 1.6 Hz, 1H), 7.50 (dd, J = 8.5, 1.6 Hz, 1H), 4.78 (s, 1H), 4.31 (s, 1H), 3.12 (t, J = 2.4 Hz, 1H), 2.78 (s, 6H), 2.72 (s, 3H), 2.14 – 1.80 (m, 7H). Biological Examples Example A Human Peripheral Blood Mononuclear Cell (PBMC) Cell-Based Assay [0816] Human peripheral blood mononuclear cells (PBMCs) consist of lymphocytes, monocytes and dendritic cells that express TLR7, TLR8 and TLR9. These cells respond to TLR7, TLR8 and TLR9 ligand stimulation and produce cytokines and chemokines in vitro and in vivo. Human PBMCs are therefore suitable to be used in a cell-based assay to assess the in vitro potency of a TLR 7, 8, and/or 9 antagonist. The result is expected to be more translatable to the pharmacodynamics response in vivo than cell line-based assays. [0817] Cryopreserved human PBMCs from healthy donors were thawed and resuspended in RPMI-1640 media with L-glutamine (Corning) supplemented with 10% Fetal Bovine Serum (Hyclone) and 1X Penicillin-Streptomycin (Corning). After counting, the cell density was adjusted to 2 million cells/ml and incubated for 1 hour at 37°C, 5% CO 2 for recovery. Following the recovery, the cells were plated by adding 50µl per well (100,000 cells) to 384-well cell culture plates (Greiner) containing 250nl of test antagonists in 100% DMSO per well, in a 10 points dose response in quadruplicates. PBMCs were incubated in the presence of test antagonists for one hour at 37°C, 5% CO2 before being stimulated with a TLR7 or TLR9 agonist. GS-986 (Gilead Sciences) was used as the TLR7 agonist at a final concentration of 400nM. ODN-2216 (InvivoGen) was used as the TLR9 agonist at a final concentration of 3µM. PBMCs were incubated in the presence of the test antagonist and the TLR7 (or TLR9) agonist for an additional 6 hours at 37 o C, 5% CO2. At the end of the incubation, the cell culture plates were centrifuged at 500g for 5 min, and the cell culture supernatant was collected. The level of cytokines (IL-6 and IFNα) in the supernatant was measured by electrochemiluminescence immunoassays (Mesoscale Discovery) following manufacturer’s recommended protocols. The level of cytokine measured was plotted against the test antagonist concentration and fitted to a sigmoidal function to determine the EC 50 , which are shown below in Table 26. TLR8 Human Peripheral Blood Mononuclear Cell (PBMC) Cell-Based Assay [0818] Cryopreserved human PBMCs from healthy donors were thawed and resuspended in RPMI-1640 media with L-glutamine (Corning) supplemented with 10% Fetal Bovine Serum (Hyclone) and 1X Penicillin-Streptomycin (Corning). After counting, the cell density was adjusted to 2 million cells/ml and incubated for 1 hour at 37 o C, 5% CO2 for recovery. Following the recovery, the cells were plated by adding 50µl per well (100,000 cells) to 384-well cell culture plates (Greiner) containing 250nl of test antagonists in 100% DMSO per well, in a 10 points dose response in quadruplicates. PBMCs were incubated in the presence of antagonists for one hour at 37 o C, 5% CO 2 before being stimulated with TLR8 agonist. Compound A (Gilead Sciences, US Patent No.10,285,990) was used as the TLR8 agonist at a final concentration of 800nM. PBMCs were incubated in the presence of antagonist and the TLR8 agonist for an additional 6 hours at 37 o C, 5% CO2. At the end of the incubation, the cell culture plates were centrifuged at 500g for 5 min, and the cell culture supernatant was collected. The level of cytokines (TNFa and IL12p40) in the supernatant was measured by electrochemiluminescence immunoassays (Mesoscale Discovery) following manufacturer’s recommended protocols. The level of cytokine measured was plotted against the antagonist concentration and fitted to a sigmoidal function to determine the EC50, which are shown below in Table 11. Compound A has the structure: . Table 26.

Example B Human Ether-à-go-go-Related Gene (hERG) Assay [0819] The FASTPatch® Assay (Charles River) was used to examine the in vitro effects of the compounds provided herein on the cloned hERG potassium channel encoded by the KCNH2 gene and stably expressed in HEK293T cells. Vehicle, test and control article formulations were prepared by diluting DMSO stock solutions in HB-PS (HEPES-buffered physiological saline solution) and were delivered to cells via the QPatch robot pipetting system to a final concentration of 0.3% DMSO. Test compound final concentrations of 1, 3, 10 and 30 uM were applied to cells (n ≥ 3, where n = the number of cells/concentration) in ascending order in at least three minute intervals separated by solution exchange. The positive control, 50 nM cisparide, was applied in the same manner. Cell membrane currents were recorded at room temperature with up to 48 parallel patch clamp amplifiers in the QPatch HT® or QPatch HTX® system, and only cells with validated whole-cell recordings (seal resistance ≥ 200 MΩ and leak current ≤ 25% channel current) were used. Onset and block of hERG current was measured using a stimulus voltage pattern consisting of a 500 ms prepulse to -40 mV, a 2-second activating pulse to +40 mV, followed by a 2-second test pulse to -40 mV. The pulse pattern was repeated continuously at 10-second intervals from a holding potential of -80 mV. %Block = {1- 1/[1+([Test]/IC50) N ]}*100% Where [Test] is the concentration of test article, IC50 is the concentration of the test article producing half-maximal inhibition, N is the Hill coefficient and ĥ Block is the percentage of current inhibited at each concentration of the test article. Nonlinear least squares fits will be solved with the Solver add-in for Excel 2000 or later (Microsoft, Redmon, WA). TurboSol analysis for test compound solubility was performed for each concentration tested using a Nephelostar reader measuring light scatter from a 635 nm laser source. Based on validation experiments, light scatter above four times the background level was considered an indication of the presence of particles in suspension. [0820] The hERG IC50 values are provided below in Table 27. Example C [0821] TLR9 EC 50 values and hERG IC 50 values were determined using the same protocols described in Examples A and B. Table 27 shows the comparison of TLR9 and hERG activity between compounds provided herein and Examples A, B, C, and D. The below table shows that the compounds provided herein (e.g., Examples 1, 64, 53, and 23) have TLR-9 inhibitory values (EC50 value) that are superior (more potent) to Examples A, B, C, and D (e.g., compare Example 1 to Example A, compare Example 64 to Example B, compare Example 53 to Example C, and compare Example 23 to Example D). The below table also shows that the compounds provided herein (exemplified by Examples 1, 64, 53, and 23) have hERG inhibitory values (IC50 value) than are superior (less potent) to Examples A, B, C, and D (e.g., compare Example 1 to Example A, compare Example 64 to Example B, compare Example 53 to Example C, and compare Example 23 to Example D). [0822] Example A is published as Example 257 in WO 2019/126253 and has the structure: . [0823] Example B is published as Example 320 in WO 2019 / 126253 and has the structure: . [0824] Example C is published as Example 187 in WO 2019 / 126113 and has the structure: E xample C . [0825] Example D has the structure: . Table 27. [0826] All references, including publications, patents, and patent documents are incorporated by reference herein, as though individually incorporated by reference. The present disclosure provides reference to various embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the present disclosure. The description is made with the understanding that it is to be considered an exemplification of the claimed subject matter and is not intended to limit the appended claims to the specific embodiments illustrated.