Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS AND COMPOSITIONS FOR TREATING CONDITIONS ASSOCIATED WITH STING ACTIVITY
Document Type and Number:
WIPO Patent Application WO/2023/137041
Kind Code:
A1
Abstract:
This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.

Inventors:
VENKATRAMAN SHANKAR (US)
KATZ JASON (US)
ROUSH WILLIAM R (US)
SEIDEL HANS MARTIN (US)
Application Number:
PCT/US2023/010561
Publication Date:
July 20, 2023
Filing Date:
January 11, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
IFM DUE INC (US)
International Classes:
C07D209/40; A61K31/404; A61P35/00; C07D403/12; C07D405/12; C07D409/12; C07D413/12; C07D471/08; C07D493/08
Domestic Patent References:
WO2008061109A22008-05-22
WO2021236405A12021-11-25
WO2015061294A22015-04-30
Foreign References:
US20040106667A12004-06-03
US6043248A2000-03-28
US6124306A2000-09-26
EP0029546A11981-06-03
US7511136B22009-03-31
US20160289196A12016-10-06
US7927613B22011-04-19
US20120202848A12012-08-09
Other References:
"Remington: The Science and Practice of Pharmacy", 2012, LIPPINCOTT WILLIAMS & WILKINS
"Pharmaceutical Preformulation and Formulation", 2009, THE PHARMACEUTICAL PRESS AND THE AMERICAN PHARMACEUTICAL ASSOCIATION
"Handbook of Pharmaceutical Additives", 2007, GOWER PUBLISHING COMPANY
LAMMERS ET AL.: "Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems", NEOPLASIA, vol. 10, 2006, pages 788 - 795
FILIPSKI, K.J. ET AL., CURRENT TOPICS IN MEDICINAL CHEMISTRY, vol. 13, 2013, pages 776 - 802
POSTOW, M., J. CLIN. ONCOL., vol. 33, 2015, pages 1
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
T. W. GREENERGM. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS
L. FIESERM. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
Attorney, Agent or Firm:
KENDALL, John T. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of Formula (I):

Formula I or a pharmaceutically acceptable salt thereof or a tautomer thereof, wherein:

LA is - (L1)ai-(L2)a2-(L3)a3-(L4)a4-(L5)a5-*, wherein * represents the point of attachment to Q1; al, a2, a3, a4, and a5 are each independently 0 or 1, provided that al + a2 + a3 + a4 + a5 > 1, and each of L1, L3, and L5 is independently selected from the group consisting of: -O-, -N(H)-, -N(Rd)-, S(0)o-2, and -C(=O)-; provided that when one or both of a2 and a4 is 0, then the combinations of L1, L3, and L5 cannot form 0-0 , N-O, N-N, O-S, S-S, or N-S(0)o bonds, and each of L2 and L4 is independently selected from the group consisting of:

• straight-chain Ci-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb;

• C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc provided the C3-10 cycloalkylene or C3-10 cycloalkenylene is not directly connected to the 6-membered ring containing Y1, Y2, and Y3; and

• heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(0)o-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc, provided the heterocyclylene or heterocycloalkenylene is not directly connected to the 6-membered ring containing Y1, Y2, and Y3; further provided that LA cannot include a cyclic group directly attached to the 6- membered ring containing Y1, Y2, and Y3;

Q1 is -Rg;

Y1, Y2, and Y3 are each independently selected from the group consisting of CR1, C(=O), N, and NR2;

X1 is selected from the group consisting of O, S, N, NR2, and CR1;

X2 is selected from the group consisting of O, S, N, NR4, and CR5; each — is independently a single bond or a double bond, provided that the fivemembered ring comprising X1 and X2 is heteroaryl, and that the six-membered ring comprising Y1, Y2, and Y3 is aryl or heteroaryl; each occurrence of R1 and R5 is independently selected from the group consisting of: H; Rc; Rg; and -(Lg)bg-Rg; each occurrence of R2 and R4 is independently selected from the group consisting of: H; Rd; Rg; and -(Lg)bg-Rg;

R6 is selected from the group consisting of: H; Rd; and Rg;

W is selected from the group consisting of:

• Ci-io alkyl, C2-10 alkenyl, or C2-10 alkynyl, each of which is optionally substituted with 1-6 Ra2 or Rg;

• C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; and

• heteroaryl, heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc, provided that when W is heteroaryl, heterocyclyl or heterocycloalkenyl, it is attached to the C(=O)NR6 group via a ring carbon atom; each occurrence of Ra and Ra2 is independently selected from the group consisting of: -OH; -halo; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=O)O(Ci-4 alkyl); -C(=O)(Ci-4 alkyl); -C(=O)OH; -CONR’R”; -S(O)I-2NR’R”; -S(O)i-2(Ci-4 alkyl); and cyano; each occurrence of Rb and Rc is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; Ci-4 alkoxy; Ci-4 haloalkoxy; -S(O)i-2(Ci-4 alkyl); - S(O)(=NH)(CI-4 alkyl); -NReRf; -OH; -S(O)I-2NR’R”; -Ci-4 thioalkoxy; -NO2; - C(=0)(Ci-io alkyl); -C(=O)O(Ci-4 alkyl); -C(=O)OH; -C(=O)NR’R”; and -SF5; each occurrence of Rd is independently selected from the group consisting of: Ci-6 alkyl optionally substituted with 1-3 independently selected Ra; -C(O)(Ci-4 alkyl); - C(O)O(Ci-4 alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)i-2(Ci-4 alkyl); -OH; and Ci-4 alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; Ci-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, halo, Ci-4 alkoxy, and Ci-4 haloalkoxy; - C(O)(Ci-4 alkyl); -C(O)O(Ci-4 alkyl); -CONR’R”; -S(O)i-2NR’R”; -S(O)i-2(Ci-4 alkyl); - OH; and Ci-4 alkoxy; each occurrence of Rg is independently selected from the group consisting of:

• Cs-12 cycloalkyl or Cs-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;

141 • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;

• heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh ; and

• Ce-io aryl optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh; each occurrence of Rh is independently selected from the group consisting of:

• C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 R‘;

• heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 R‘;

• heteroaryl of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R‘; and

• Ce-io aryl optionally substituted with 1-4 R‘; each occurrence of R‘ is independently selected from the group consisting of: C1-6 alkyl; Ci-4 haloalkyl; Ci-4 alkoxy; Ci-4 haloalkoxy; and halo; each occurrence of Lg is independently selected from the group consisting of: -O-, -NH-, -NRd, -S(0)o-2, C(O), and C1-3 alkylene optionally substituted with 1-3 Ra; each occurrence of bg is independently 1, 2, or 3; and each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; and Ci-4 alkyl.

2. The compound of claim 1, wherein a2 is 1.

3. The compound of claims 1 or 2, wherein L2 is straight-chain Ci-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb, optionally wherein L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb; optionally wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.

4. The compound of claims 1 or 2, wherein L2 is selected from the group consisting of:

• C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc; and

• heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(0)o-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc.

5. The compound of any one of claims 1-4, wherein al is 1.

6. The compound of any one of claims 1-5, wherein L1 is selected from the group consisting of: -O-, -N(H)-, -N(Rd)-, and -S-, optionally wherein L1 is -O-.

7. The compound of any one of claims 1-3, wherein al is 0.

8. The compound of any one of claims 1-7, wherein a3 is 1.

9. The compound of any one of claims 1-8, wherein L3 is selected from the group consisting of: -O-, -N(H)-, -N(Rd)-, and -S-, optionally wherein L3 is -O-.

10. The compound of any one of claims 1-7, wherein a3 is 0.

11. The compound of any one of claims 1-10, wherein a4 is 1.

12. The compound of claim 1, wherein: al and a2 are each 1; optionally, wherein: al and a2 are each 1;

L1 is -O-, -N(H)-, or -N(Rd)-; and

L2 is selected from the group consisting of:

• straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb;

• C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc; optionally wherein: al and a2 are each 1;

L1 is -O-; and

L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb; optionally wherein: al and a2 are each 1;

L1 is -O-; and L2 is C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; optionally wherein L2 is: which is optionally substituted with 1-2 Rc,

144 wherein nl and n2 are independently 0, 1, or 2; Q2 is CH, CRC, or N; and the asterisk represents the point of attachment to -(L3)a3-; optionally wherein nl and n2 are independently 0 or 1, optionally 0; and Q2 is CH; optionally wherein nl and n2 are 0 and Q2 is CH; optionally wherein L2 is cyclobutanediyl optionally substituted with 1-2 Rc;optionally wherein L2 is cyclobutane-l,3-diyl optionally substituted with 1-2 Rc; optionally wherein L2 is unsubstituted cyclobutane-diyl; optionally wherein L2 is unsubstituted cyclobutane- 1,3 -diyl.

13. The compound of claim 12, wherein a3, a4, and a5 are each 0, optionally wherein (such

), wherein * represents the point of attachment to Q1.

14. The compound of claim 1, wherein al is 0; a2 is 1; optionally wherein L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb, optionally wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.

15. The compound of claim 14, wherein a3 is 1; optionally, wherein L3 is selected from the group consisting of: is -O-, -N(H)-, and -N(Rd)-, optionally wherein L3 is -O-.

16. The compound of claims 14 or 15, wherein a4 is 0; and a5 is 0, optionally wherein LA is -CH2CH2-O-*, wherein * represents to point of attachment to Q1.

17. The compound of any one of claims 1-16, wherein Q1 is selected from the group consisting of:

• heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-3 Rc’; and

• phenyl optionally substituted with 1-3 Rc’.

145

18. The compound of any one of claims 1-16, wherein Q1 is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc’; optionally wherein wherein ml and m2 are each independently 0, 1, or 2; and wherein Q1 is optionally substituted with 1-2 Rc’; and optionally wherein each Rd present in Q1 is independently selected from the group consisting of: -C(O)O(Ci-4 alkyl); and Ci-6 alkyl optionally substituted with 1-3 independently selected Ra.

19. The compound of any one of claims 1-18, wherein Y1 is CR1; Y2 is CR1; and/or Y3 is CR1.

20. The compound of any one of claims 1-19, wherein X1 is NR2; and X2 is CR5; optionally wherein X1 is NH; and X2 is CH.

21. The compound of any one of claims 1-20, wherein R6 is H.

22. The compound of any one of claims 1-21, wherein W is:

(i) Ci-io alkyl, C2-10 alkenyl, or C2-10 alkynyl, each of which is optionally substituted with 1 -6 Ra2 or Rg; optionally C1-10 alkyl, which is optionally substituted with 1-6 Ra2 or Rg; optionally Ci-4 alkyl, which is optionally substituted with 1-6 Ra2 or Rg; optionally wherein C1-10 alkyl, C2-10 alkenyl, or C2-10 alkynyl is substituted with one Rg, optionally wherein Rg is cycloalkyl, heteroary or heterocyclyl.

23. The compound of any one of claims 1-21, wherein W is:

146 (i) C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; optionally C3-12 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc.

(ii) heteroaryl, heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;

24. The compound of claim 1 , wherein the compound is selected from the group consisting of the compounds delineated in Table Cl or a pharmaceutically acceptable salt thereof.

25. A pharmaceutical composition comprising a compound of any one of claims 1-24, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.

26. A method for inhibiting STING activity, the method comprising contacting STING with a compound as claimed in any one of claims 1-24, or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition as claimed in claim 25.

27. A method of inducing an immune response in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound as claimed in any one of claims 1-24, or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition as claimed in claim 25.

28. A method of treatment of disease, disorder, or condition associated with STING, such as a disease, disorder, or condition, in which increased STING signaling, such as excessive STING signaling, contributes to the pathology and/or symptoms and/or progression of the disease, such as cancer, comprising administering to a subject in need of such treatment an effective amount of a compound as claimed in any one of claims 1-

147 24, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as claimed in claim 25.

148

Description:
Compounds and Compositions for Treating Conditions Associated with STING Activity

CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application Serial No. 63/298,893, filed on January 12, 2022, which is incorporated herein by reference in its entirety.

TECHNICAL FIELD

This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.

BACKGROUND

STING, also known as transmembrane protein 173 (TMEM173) and MPYS/MITA/ERIS, is a protein that in humans is encoded by the TMEM173 gene. STING has been shown to play a role in innate immunity. STING induces type I interferon production when cells are infected with intracellular pathogens, such as viruses, mycobacteria and intracellular parasites. Type I interferon, mediated by STING, protects infected cells and nearby cells from local infection in an autocrine and paracrine manner.

The STING pathway is pivotal in mediating the recognition of cytosolic DNA. In this context, STING, a transmembrane protein localized to the endoplasmic reticulum (ER), acts as a second messenger receptor for 2', 3' cyclic GMP-AMP (hereafter cGAMP), which is produced by cGAS after dsDNA binding. In addition, STING can also function as a primary pattern recognition receptor for bacterial cyclic dinucleotides (CDNs) and small molecule agonists. The recognition of endogenous or prokaryotic CDNs proceeds through the carboxy-terminal domain of STING, which faces into the cytosol and creates a V-shaped binding pocket formed by a STING homodimer. Ligand-induced activation of STING triggers its re-localization to the Golgi, a process essential to promote the interaction of STING with TBK1. This protein complex, in turn, signals through the transcription factors IRF-3 to induce type I interferons (IFNs) and other co-regulated antiviral factors. In addition, STING was shown to trigger NF-KB and MAP kinase activation. Following the initiation of signal transduction, STING is rapidly degraded, a step considered important in terminating the inflammatory response.

Excessive activation of STING is associated with a subset of monogenic autoinflammatory conditions, the so-called type I interferonopathies. Examples of these diseases include a clinical syndrome referred to as STING-associated vasculopathy with onset in infancy (SAVI), which is caused by gain-of-function mutations in TMEM173 (the gene name of STING). Moreover, STING is implicated in the pathogenesis of Aicardi- Goutieres Syndrome (AGS) and genetic forms of lupus. As opposed to SAVI, it is the dysregulation of nucleic acid metabolism that underlies continuous innate immune activation in AGS. Apart from these genetic disorders, emerging evidence points to a more general pathogenic role for STING in a range of inflammation-associated disorders such as systemic lupus erythematosus, rheumatoid arthritis and cancer. Thus, small moleculebased pharmacological interventions into the STING signaling pathway hold significant potential for the treatment of a wide spectrum of diseases

SUMMARY

This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same. An "antagonist" of STING includes compounds that, at the protein level, directly bind or modify STING such that an activity of STING is decreased, e.g., by inhibition, blocking or dampening agonist-mediated responses, altered distribution, or otherwise. STING antagonists include chemical entities, which interfere or inhibit STING signaling.

In one aspect, compounds of Formula (I), or a pharmaceutically acceptable salt thereof, are featured: in which Q 1 , L A , Y 1 , Y 2 , Y 3 , X 1 , X 2 , R 6 , and W can be as defined anywhere herein.

In one aspect, pharmaceutical compositions are featured that include a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) and one or more pharmaceutically acceptable excipients.

In one aspect, methods for inhibiting (e.g., antagonizing) STING activity are featured that include contacting STING with a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same). Methods include in vitro methods, e.g., contacting a sample that includes one or more cells comprising STING (e.g., innate immune cells, e.g., mast cells, macrophages, dendritic cells (DCs), and natural killer cells) with the chemical entity. Methods can also include in vivo methods; e.g., administering the chemical entity to a subject (e.g., a human) having a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease.

In one aspect, methods of treating a condition, disease or disorder ameliorated by antagonizing STING are featured, e.g., treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). The methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).

In another aspect, methods of treating cancer are featured that include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).

In a further aspect, methods of treating other STING-associated conditions are featured, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis. The methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).

In another aspect, methods of suppressing STING-dependent type I interferon production in a subject in need thereof are featured that include administering to the subject an effective amount of a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).

In a further aspect, methods of treating a disease in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the disease are featured. The methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).

In another aspect, methods of treatment are featured that include administering an effective amount of a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) to a subject; wherein the subject has (or is predisposed to have) a disease in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the disease.

In a further aspect, methods of treatment that include administering to a subject a chemical entity described herein (e.g., a compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same), wherein the chemical entity is administered in an amount effective to treat a disease in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.

In another aspect, there is provided is a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein, for use in the treatment of a disease, condition or disorder modulated by STING inhibition.

In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of a condition, disease or disorder associated with increased (e.g., excessive) STING activation.

In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, described herein for use in the treatment of cancer.

In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of cancer selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of type I interferonopathies. In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of type I interferonopathies selected from STING-associated vasculopathywith onset in infancy (SA VI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation- associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of a condition, disease or disorder associated with increased (e.g., excessive) STING activation.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of cancer.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of cancer selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of type I interferonopathies.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the manufacture of a medicament for the treatment of type I interferonopathies selected from STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis. In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein, for the treatment of a disease, condition or disorder modulated by STING inhibition.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of a condition, disease or disorder associated with increased (e.g., excessive) STING activation.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of cancer.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of cancer selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of type I interferonopathies.

In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of type I interferonopathies selected from STING-associated vasculopathy with onset in infancy (SA VI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation- associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.

Embodiments can include one or more of the following features.

The chemical entity can be administered in combination with one or more additional therapeutic agents and/or regimens. For examples, methods can further include administering one or more (e.g., two, three, four, five, six, or more) additional agents.

The chemical entity can be administered in combination with one or more additional therapeutic agents and/or regimens that are useful for treating other STING- associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.

The chemical entity can be administered in combination with one or more additional cancer therapies (e.g., surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof; e.g., chemotherapy that includes administering one or more (e.g., two, three, four, five, six, or more) additional chemotherapeutic agents. Non-limiting examples of additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g., azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisomerase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an antihelminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 -PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL- 10, transforming growth factor-P (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM

- LIGHT, HVEM-BTLA-CD160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48

- CD244, CD244, ICOS, ICOS-ICOS ligand, B7 H3, B7 H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD1 or PD-L1).

The subject can have cancer; e.g., the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies.

Non-limiting examples of cancer include melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma. In certain embodiments, the cancer can be a refractory cancer.

The chemical entity can be administered intratum orally. The methods can further include identifying the subject.

Other embodiments include those described in the Detailed Description and/or in the claims.

Additional Definitions

To facilitate understanding of the disclosure set forth herein, a number of additional terms are defined below. Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well-known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Each of the patents, applications, published applications, and other publications that are mentioned throughout the specification and the attached appendices are incorporated herein by reference in their entireties.

As used herein, the term “STING” is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous STING molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.

The term “acceptable” with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.

“API” refers to an active pharmaceutical ingredient.

The terms “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of a chemical entity being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case is determined using any suitable technique, such as a dose escalation study.

The term “excipient” or “pharmaceutically acceptable excipient” means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material. In one embodiment, each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed , Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 6th ed. Rowe el al., Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.,' Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed, Gibson Ed.; CRC Press LLC: Boca Raton, FL, 2009.

The term “pharmaceutically acceptable salt” refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In certain instances, pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. In some instances, pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, A-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined. The pharmacologically acceptable salt s not specifically limited as far as it can be used in medicaments. Examples of a salt that the compounds described hereinform with a base include the following: salts thereof with inorganic bases such as sodium, potassium, magnesium, calcium, and aluminum; salts thereof with organic bases such as methylamine, ethylamine and ethanolamine; salts thereof with basic amino acids such as lysine and ornithine; and ammonium salt. The salts may be acid addition salts, which are specifically exemplified by acid addition salts with the following: mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid:organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; acidic amino acids such as aspartic acid and glutamic acid. The term “pharmaceutical composition” refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.

The term “subject” refers to an animal, including, but not limited to, a primate (e.g., human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human.

The terms “treat,” “treating,” and “treatment,” in the context of treating a disease or disorder, are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof. The “treatment of cancer”, refers to one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, (i) slowing down and (ii) complete growth arrest; (2) reduction in the number of tumor cells; (3) maintaining tumor size; (4) reduction in tumor size; (5) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of tumor cell infiltration into peripheral organs; (6) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of metastasis; (7) enhancement of anti-tumor immune response, which may result in (i) maintaining tumor size, (ii) reducing tumor size, (iii) slowing the growth of a tumor, (iv) reducing, slowing or preventing invasion and/or (8) relief, to some extent, of the severity or number of one or more symptoms associated with the disorder.

The term "halo" refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I).

The term "alkyl" refers to a saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, Ci-io indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, z.w-propyl, tert-butyl, //-hexyl. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.

The term "haloalkyl" refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo.

The term "alkoxy" refers to an -O-alkyl radical (e.g., -OCH3).

The term "alkylene" refers to a divalent alkyl (e.g., -CH2-).

The term "alkenyl" refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds. The alkenyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it. Alkenyl groups can either be unsubstituted or substituted with one or more substituents.

The term "alkynyl" refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds. The alkynyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it. Alkynyl groups can either be unsubstituted or substituted with one or more substituents.

The term "aryl" refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, tetrahydronaphthyl, dihydro- IH-indenyl and the like.

The term "cycloalkyl" as used herein refers to cyclic saturated hydrocarbon groups having, e.g., 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyl may include multiple fused and/or bridged rings. Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butanyl, bicyclo[2.1.0]pentanyl, bicyclo[l. l.l]pentanyl, bicyclo[3.1.0]hexanyl, bicyclo[2.1.1 ]hexanyl, bicyclo[3 ,2.0]heptanyl, bicyclo[4.1.0]heptanyl, bicyclo[2.2. l]heptanyl, bicyclo[3.1. l]heptanyl, bicyclo[4.2.0]octanyl, bicyclo[3.2.1]octanyl, bicyclo[2.2.2]octanyl, and the like. Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic cycloalkyls include spiro[2.2]pentanyl, spiro[2.5]octanyl, spiro[3.5]nonanyl, spiro[3.5]nonanyl, spiro[3.5]nonanyl, spiro[4.4]nonanyl, spiro[2.6]nonanyl, spiro[4.5]decanyl, spiro[3.6]decanyl, spiro[5.5]undecanyl, and the like. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms.

The term "cycloalkenyl" as used herein means partially unsaturated cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkenyl group may be optionally substituted. Examples of cycloalkenyl groups include, without limitation, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. As partially unsaturated cyclic hydrocarbon groups, cycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the cycloalkenyl group is not fully saturated overall. Cycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.

The term “heteroaryl”, as used herein, means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic, and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl). Heteroaryl groups can either be unsubstituted or substituted with one or more substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotri azolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-t ]pyrimidinyl, pyrrolo[2,3- Z>]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-Z>]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridinyl, pyrazolo[4,3-Z>]pyridinyl, tetrazolyl, chromanyl, 2,3-dihydrobenzo[Z>][l,4]dioxinyl, benzo[ ][l,3]dioxolyl, 2,3- dihydrobenzofuranyl, tetrahydroquinolinyl, 2,3-dihydrobenzo[Z>][l,4]oxathiinyl, isoindolinyl, and others. In some embodiments, the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl.

The term "heterocyclyl" refers to a mon-, bi-, tri-, or polycyclic saturated ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like. Heterocyclyl may include multiple fused and bridged rings. Non-limiting examples of fused/bridged heteorocyclyl includes: 2-azabicyclo[1.1.0]butanyl, 2-azabicyclo[2.1.0]pentanyl, 2- azabicyclo[l. l.l]pentanyl, 3-azabicyclo[3.1.0]hexanyl, 5-azabicyclo[2.1.1]hexanyl, 3- azabicyclo[3.2.0]heptanyl, octahydrocyclopenta[c]pyrrolyl, 3-azabicyclo[4.1.0]heptanyl, 7-azabicyclo[2.2.1]heptanyl, 6-azabicyclo[3.1.1]heptanyl, 7-azabicyclo[4.2.0]octanyl, 2- azabicyclo[2.2.2]octanyl, 3-azabicyclo[3.2.1]octanyl, 2-oxabicyclo[1.1.0]butanyl, 2- oxabicyclo[2.1.0]pentanyl, 2-oxabicyclo[l. l.l]pentanyl, 3-oxabicyclo[3.1.0]hexanyl, 5- oxabicyclo[2.1.1]hexanyl, 3-oxabicyclo[3.2.0]heptanyl, 3-oxabicyclo[4.1.0]heptanyl, 7- oxabicyclo[2.2.1]heptanyl, 6-oxabicyclo[3.1.1]heptanyl, 7-oxabicyclo[4.2.0]octanyl, 2- oxabicyclo[2.2.2]octanyl, 3-oxabicyclo[3.2.1]octanyl, and the like. Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic heterocyclyls include 2- azaspiro[2.2]pentanyl, 4-azaspiro[2.5]octanyl, 1-azaspiro[3.5]nonanyl, 2 azaspiro[3.5]nonanyl, 7-azaspiro[3.5]nonanyl, 2-azaspiro[4.4]nonanyl, 6 azaspiro[2.6]nonanyl, 1 , 7-diazaspiro[4.5 ] decanyl, 7-azaspiro[4.5] decanyl 2,5 diazaspiro[3.6]decanyl, 3 -azaspiro[5.5]undecanyl, 2-oxaspiro[2.2]pentanyl, 4 oxaspiro [2.5 ] octanyl , 1-oxaspiro[3.5]nonanyl, 2-oxaspiro[3.5]nonanyl, 7 oxaspiro[3.5]nonanyl, 2-oxaspiro[4.4]nonanyl, 6-oxaspiro[2.6]nonane, 1,7 dioxaspiro[4.5]decanyl, 2,5-dioxaspiro[3.6]decanyl, l-oxaspiro[5.5]undecanyl, 3 oxaspiro[5.5]undecanyl, 3-oxa-9-azaspiro[5.5]undecanyl and the like. The term “saturated” as used in this context means only single bonds present between constituent ring atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.

The term "heterocycloalkenyl" as used herein means partially unsaturated cyclic ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocycloalkenyl groups include, without limitation, tetrahydropyridyl, dihydropyrazinyl, dihydropyridyl, dihydropyrrolyl, dihydrofuranyl, dihydrothiophenyl. As partially unsaturated cyclic groups, heterocycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the heterocycloalkenyl group is not fully saturated overall. Heterocycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.

As used herein, when a ring is described as being “aromatic”, it means said ring has a continuous, delocalized 7t-electron system. Typically, the number of out of plane 7t- electrons corresponds to the Hiickel rule (4n+2). Examples of such rings include: benzene, pyridine, pyrimidine, pyrazine, pyridazine, pyridone, pyrrole, pyrazole, oxazole, thioazole, isoxazole, isothiazole, and the like.

As used herein, when a ring is described as being “partially unsaturated”, it means said ring has one or more additional degrees of unsaturation (in addition to the degree of unsaturation attributed to the ring itself; e.g., one or more double or tirple bonds between constituent ring atoms), provided that the ring is not aromatic. Examples of such rings include: cyclopentene, cyclohexene, cycloheptene, dihydropyridine, tetrahydropyridine, dihydropyrrole, dihydrofuran, dihydrothiophene, and the like.

For the avoidance of doubt, and unless otherwise specified, for rings and cyclic groups (e.g., aryl, heteroaryl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, cycloalkyl, and the like described herein) containing a sufficient number of ring atoms to form bicyclic or higher order ring systems (e.g., tricyclic, polycyclic ring systems), it is understood that such rings and cyclic groups encompass those having fused rings, including those in which the points of fusion are located (i) on adjacent ring atoms (e.g., [x.x.O] ring systems, in which 0 represents a zero atom bridge (e.g., )); (ii) a single ring atom (spiro- fused ring systems) (e.g., (iii) a contiguous array of ring atoms (bridged ring systems having all bridge lengths > 0) (e.g.,

In addition, atoms making up the compounds of the present embodiments are intended to include all isotopic forms of such atoms. Isotopes, as used herein, include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include 13 C and 14 C.

In addition, the compounds generically or specifically disclosed herein are intended to include all tautomeric forms. Thus, by way of example, a compound containing the encompasses the tautomeric form containing the moiety: . y, a pyridinyl or pyrimidinyl moiety that is described to be optionally substituted with hydroxyl encompasses pyridone or pyrimidone tautomeric forms.

As used herein, the phrase “optionally substituted” when used in conjunction with a structural moiety (e.g., alkyl) is intended to encompass both the unsubstituted structural moiety (i.e., none of the substitutable hydrogen atoms are replaced with one or more non- hydrogen substituents) and substituted structural moieties substituted with the indicated range of non-hydrogen substituents. For example, “ C1-C4 alkyl optionally substituted with 1-4 R a ” is intended to encompass both unsubstituted C1-C4 alkyl and C1-C4 alkyl substituted with 1-4 R a .

The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features and advantages of the invention will be apparent from the description and drawings, and from the claims.

DETAILED DESCRIPTION

This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.

Formula I Compounds

In one aspect, the disclosure features a compound of Formula (I):

Formula I or a pharmaceutically acceptable salt thereof or a tautomer thereof, wherein:

L A is - (L 1 )ai-(L 2 )a2-(L 3 )a3-(L 4 )a4-(L 5 ) a 5-*, wherein * represents the point of attachment to Q 1 ; al, a2, a3, a4, and a5 are each independently 0 or 1, provided that each of L 1 , L 3 , and L 5 is independently selected from the group consisting of: -O-, -N(H)-, -N(R d )-, S(0)o-2, and -C(=O)-; provided that when one or both of a2 and a4 is 0, then the combinations of L 1 , L 3 , and L 5 cannot form 0-0 , N-O, N-N, O-S, S-S, or N-S(0)o bonds, and each of L 2 and L 4 is independently selected from the group consisting of:

• straight-chain Ci-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 R b ;

• C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 R c , provided the C3-10 cycloalkylene or C3-10 cycloalkenylene is not directly connected to the 6-membered ring containing Y 1 , Y 2 , and Y 3 ; and

• heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 R c , provided the heterocyclylene or heterocycloalkenylene is not directly connected to the 6-membered ring containing Y 1 , Y 2 , and Y 3 ;

Q 1 is -R g ;

Y 1 , Y 2 , and Y 3 are each independently selected from the group consisting of CR 1 , C(=O), N, and NR 2 ;

X 1 is selected from the group consisting of O, S, N, NR 2 , and CR 1 ;

X 2 is selected from the group consisting of O, S, N, NR 4 , and CR 5 ; each == is independently a single bond or a double bond, provided that the fivemembered ring comprising X 1 and X 2 is heteroaryl, and that the six-membered ring comprising Y 1 , Y 2 , and Y 3 is aryl or heteroaryl; further provided that L A cannot include a cyclic group directly attached to the 6- membered ring containing Y 1 , Y 2 , and Y 3 ; each occurrence of R 1 and R 5 is independently selected from the group consisting of: H; R c ; R g ; and -(L g ) bg -R g ; each occurrence of R 2 and R 4 is independently selected from the group consisting of: H; R d ; R g ; and -(L g ) bg -R g ;

R 6 is selected from the group consisting of: H; R d ; and R g ;

W is selected from the group consisting of:

• Ci-io alkyl, C2-10 alkenyl, or C2-10 alkynyl, each of which is optionally substituted with 1-6 R a2 or R g ;

• C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• heteroaryl, heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c , provided that when W is heteroaryl, heterocyclyl or heterocycloalkenyl, it is attached to the C(=O)NR 6 group via a ring carbon atom; each occurrence of R a and R a2 is independently selected from the group consisting of: -OH; -halo; -NR e R f ; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=O)O(Ci-4 alkyl); -C(=O)(Ci-4 alkyl); -C(=O)OH; -CONR’R”; -S(O)I- 2 NR’R”; -S(O)i. 2 (Ci-4 alkyl); and cyano; each occurrence of R b and R c is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected R a ; C2-6 alkenyl; C2-6 alkynyl; Ci-4 alkoxy; Ci-4 haloalkoxy; -S(O)i-2(Ci-4 alkyl); - S(O)(=NH)(CI- 4 alkyl); -NR e R f ; -OH; -S(O)I- 2 NR’R”; -Ci-4 thioalkoxy; -NO2; - C(=0)(Ci-io alkyl); -C(=O)O(Ci-4 alkyl); -C(=O)OH; -C(=O)NR’R”; and -SF 5 ; each occurrence of R d is independently selected from the group consisting of: C1-6 alkyl optionally substituted with 1-3 independently selected R a ; -C(O)(Ci-4 alkyl); - C(0)0(Ci-4 alkyl); -CONR’R”; -S(O)I- 2 NR’R”; - S(O)i- 2 (Ci- 4 alkyl); -OH; and Ci-4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; Ci-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, halo, Ci-4 alkoxy, and Ci-4 haloalkoxy; - C(O)(Ci- 4 alkyl); -C(O)O(Ci- 4 alkyl); -CONR’R”; -S(O)I- 2 NR’R”; -S(O)i- 2 (Ci-4 alkyl); - OH; and Ci-4 alkoxy; each occurrence of R g is independently selected from the group consisting of:

• C3-12 cycloalkyl or Cs-i 2 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ;

• heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ;

• heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ; and

• Ce-io aryl optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ; each occurrence of R h is independently selected from the group consisting of:

• C3-12 cycloalkyl or C3-i 2 cycloalkenyl, each of which is optionally substituted with 1-4 R‘;

• heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 R‘; • heteroaryl of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R‘; and

• Ce-io aryl optionally substituted with 1-4 R‘; each occurrence of R‘ is independently selected from the group consisting of: Ci-6 alkyl; Ci-4 haloalkyl; Ci-4 alkoxy; Ci-4 haloalkoxy; and halo; each occurrence of L g is independently selected from the group consisting of: -O-, -NH-, -NR d , -S(0)o-2, C(O), and C1-3 alkylene optionally substituted with 1-3 R a ; each occurrence of bg is independently 1, 2, or 3; and each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; and Ci-4 alkyl.

Variable L A (-(L 1 ) a i-(L 2 )a2-(L 3 )a3-(L 4 )a4-(L 5 ) a 5-*, wherein * represents the point of attachment to O 1 )

In some embodiments, L A is a divalent moiety having a 1-6 (e.g., 2-6 (e.g., 2, 3, or 4)) linear array of substituted or unsubstituted carbon and/or heteroatoms. In some embodiments, L A is a divalent moiety having a combination of a cyclic moiety and a 1-6 (e.g., 2-6 (e.g., 2, 3, or 4)) linear array of substituted or unsubstituted carbon and/or heteroatoms. For example, one cyclic moiety (e.g., C3-6, e.g., C4 cycloalkylene), and an acyclic moiety (e.g., O).

In some embodiments, provided that when a3 is 0; and a4 is 1, then L 4 is other than straight-chain Ci-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 R b ;

In some embodiments, a2 is 1. In some embodiments, a2 is 0.

In certain embodiments (when a2 is 1), L 2 is straight-chain C1-6 alkylene, straightchain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 R b .

In certain of the foregoing embodiments, L 2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 R b . In certain of the foregoing embodiments, L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

In certain embodiments, L 2 is selected from the group consisting of: -CH2-, -CHR b - , and -C(R b )2-. For example, L 2 can be -CH2-.

In certain embodiments (when L 2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 R b ), L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

In certain of these embodiments, L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b . In certain of the foregoing embodiments, L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -(L 3 ) a 3-. For example, L 2 can be -CH2CH2-.

In certain embodiments, L 2 is straight-chain C3 alkylene which is optionally substituted with 1-3 R b . For example, L 2 can be selected from the group consisting of: wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

In certain embodiments (when a2 is 1), L 2 is straight-chain C2-6 alkenylene, which is optionally substituted with 1-6 R b . In certain of these embodiments, L 2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 R b . For example, L 2 can be selected from the group consisting of: , wherein the asterisk represents the point of attachment to -(L 3 )^-.

In certain embodiments (when a2 is 1), L 2 is selected from the group consisting of:

• C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 R c ; and heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 R c .

In certain of these embodiments, L 2 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

In certain of the foregoing embodiments, L 2 is: which is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0, 1, or 2; Q 2 is CH, CR C , or

N; and the asterisk represents the point of attachment to -(L 3 ) a 3-.

In certain of these embodiments, Q 2 is CH.

In certain embodiments (when L 2 is: defined supra), nl and n2 are each 0.

As a non-limiting example (when L 2 is: defined supra , L 2 can be , wherein the asterisk represents the point of attachment to -(L 3 ) a 3- or -(L 1 ) a i, e.g., -(L 1 ) a i, in which al is 1. For example, L 2 can be , wherein the asterisk represents the point of attachment to -(L 1 )^. In certain of these embodiments, -(L 1 );!! is O.

In certain of the foregoing embodiments, each of a3, a4, and a5 is 0.

In some embodiments, al is 1. In some embodiments, al is 0. In certain embodiments (when al is 1), L 1 is selected from the group consisting of: -O-, -N(H)-, -N(R d )-, and -S-. In certain of these embodiments, L 1 is -O-.

In some embodiments, a3 is 1. In some embodiments, a3 is 0.

In certain embodiments (when a3 is 1), L 3 is selected from the group consisting of: -O-, -N(H)-, -N(R d )-, and -S- . In certain of these embodiments, L 3 is -O-. In certain other embodiments, L 3 is -N(H)- or -N(R d )- (e.g., -N(H)-).

In some embodiments, a4 is 1. In some embodiments, a4 is 0.

In certain embodiments (when a4 is 1), L 4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b . In certain of these embodiments, L 4 is -CH2-.

In certain embodiments (when a4 is 1), L 4 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

In certain of these embodiments, L 4 is: which is optionally substituted with 1-2 R c , wherein n3 and n4 are independently 0, 1, or 2; Q 3 is CH, CR C , or N; and the asterisk represents the point of attachment to -(L 5 ) a s-.

In certain embodiments (when L 4 is: are each 1. In certain embodiments (when L 4 is:

As a non-limiting example of the foregoing embodiments, L 4 can be , wherein the asterisk represents the point of attachment to -(L 5 ) a s-. In some embodiments, a5 is 0.

Non-Limiting Combinations of —(L 1 ) a i-(L 2 ) a2-(L 3 ) a3-(L 4 ) a -i-( 5 ) a s- *

In some embodiments, -(L 1 ) a i-(L 2 )a2-(L 3 )a3-(L 4 )a4-(L 5 ) a 5-* has a length of from 1 atom to 8 atoms (as used here and for counting purposes only, moieties such as CH2, C(O), CF2 and the like, whether present in acyclic or cyclic moieties, count as 1 atom); e.g., from 1 atom to 6 atoms, or from 1 atom to 5 atoms, or from 1 atom to 4 atoms ; or from from 1 atom to 3 atoms; or from 2 atoms to 6 atoms; or from 2 atoms to 4 atoms.

In certain embodiments, one of al, a3, and a5 is 1, and the other two of al, a3, and a5 are 0. In certain embodiments, al is 1, e.g., when L 2 is a cyclic group (e.g., cycloalkylene).

In certain embodiments, one of a2 and a4 is 1, and the other of a2 and a4 is 0 or 1.

In certain of the foregoing embodiments, one of al, a3, and a5 is 1, and the other two of al, a3, and a5 are 0; and one of a2 and a4 is 1, and the other of a2 and a4 is 0 or 1.

In certain embodiments, 1 < al+a2+a3+a4+a5 < 4. In certain of these embodiments, 1 < al+a2+a3+a4+a5 < 3.

In certain embodiments, al and a2 are each 1.

[AA1] In certain embodiments, al and a2 are each 1;

L 1 is -O-, -N(H)-, or -N(R d )-;

L 2 is selected from the group consisting of:

• straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b ;

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

[AA2] In certain embodiments, al and a2 are each 1;

L 1 is -O-; and

L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

[AA3] In certain embodiments, al and a2 are each 1;

L 1 is -O-; and

L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-.

[AA4] In certain embodiments, al and a2 are each 1;

L 1 is -O-; and

L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

In certain embodiments of [AA4], L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b . As non-limiting examples of the foregoing embodiments, L 2 can be selected from the group consisting of: -CH2CH2-, -CH2CH(R b )- *, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -(L 3 ) a 3-. For example, L 2 can be -CH2CH2-.

[AA5] In certain embodiments, al and a2 are each 1;

L 1 is -O-;

L 2 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

In certain embodiments of [AA5], L 2 is: which is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0, 1, or 2; Q 2 is CH, CR C , or N; and the asterisk represents the point of attachment to -(L 3 ) a 3-.

In certain of these embodiments, nl and n2 are independently 0 or 1, optionally 0; and Q 2 is CH. For example, nl and n2 can both be 0; and Q 2 can be CH, e.g., L 2 can be optionally substituted cyclobutane-diyl, e.g, optionally substituted cyclobutane-l,3-diyl.

In certain embodiments when al and a2 are each 1, a3, a4, and a5 are each 0.

In certain embodiments of [AA1], a3, a4, and a5 are each 0. In certain embodiments of [AA2], a3, a4, and a5 are each 0. In certain embodiments of [AA3], a3, a4, and a5 are each 0. In certain embodiments of [AA4], a3, a4, and a5 are each 0. In certain embodiments of [AA5], a3, a4, and a5 are each 0.

In certain embodiments when al and a2 are each 1, a3 and a5 are 0; and a4 is 1.

In certain embodiments of [AA1], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA2], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA3], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA4], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA5], a3 and a5 are 0; and a4 is 1.

In certain embodiments (when al and a2 are each 1, a3 and a5 are 0; and a4 is 1), L 4 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c . In certain of these embodiments, L 4 is: which is optionally substituted with 1-2 R c , wherein n3 and n4 are independently 0, 1, or 2; Q 3 is CH, CR C , or N; and the asterisk represents the point of attachment to -(L 5 ) a s-. In certain of the foregoing embodiments, n3 and n4 are independently 0 or 1; and Q 3 is N.

In certain embodiments, al is 0; and a2 is 1.

[BB1] In certain embodiments, al is 0; a2 is 1; and L 2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 R b .

In certain embodiments of [BB1], L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b . In certain of the foregoing embodiments, L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-. For example, L 2 can be -CH2-

In certain embodiments of [BB1], L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b . In certain of the foregoing embodiments, L 2 is straightchain C2 alkylene, which is optionally substituted with 1-3 R b . As non-limiting examples, L 2 can be selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and - CH2C(R b )2-*, wherein the asterisk represents point of attachment to -(L 3 ) a 3-. For example, L 2 can be -CH2CH2-.

In certain embodiments of [BB1], L 2 is straight-chain C3 alkylene, which is optionally substituted with 1-3 R b . In certain of these embodiments, L 2 is selected from the group consisting of: wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

In certain embodiments (when al is 0; and a2 is 1) , a3 is 0; and a4 is 0.

In certain embodiments of [BB1], a3 is 0; and a4 is 0.

In certain embodiments (when al is 0; and a2 is 1) , a3 is 1. In certain embodiments of [BB1], a3 is 1. In certain embodiments (when al is 0; and a2 is 1) or in certain embodiments of [BB1], a3 is 1; and L 3 is selected from the group consisting of: is -O-, -N(H)-, and -N(R d )- . In certain of these embodiments, a3 is 1; and L 3 is -O-. In certain other embodiments, a3 is 1; and L 3 is -N(H)- or -N(R d )-, optionally -N(H)-.

In certain embodiments (when al is 0; and a2 is 1) or in certain embodiments of [BB1], a4 is 1; and L 4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b . In certain of these embodiments, a4 is 1; and L 4 is -CH2-.

In certain embodiments (when al is 0; and a2 is 1) or in certain embodiments of [BB1], a4 is 0

In certain embodiments (when al is 0; and a2 is 1) or in certain embodiments of [BB1], a5 is 0.

In certain embodiments (when al is 0; and a2 is 1) or in certain embodiments of

[BB1], L A is -CH2-O-CH2- .

[CC1] In certain embodiments, al is 0; a2 is 1; L 2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 R b .

In certain embodiments of [CC1], L 2 is selected from the group consisting of: , wherein the asterisk represents the point of attachment to - (L 3 )a3-.

In certain embodiments of [CC1], a3 is 0; and a4 is 0.

For the avoidance of doubt when any one or more of al, a2, a3, a4, and a5 are 0, this means that the corresponding variable (L 1 -! ) is absent from L A . For example, when each of a3, a4, and a5 are 0, this means that L A has the formula -L 1 -! -.

In certain embodiments, L A is -L 1 -!?-.

In certain embodiments, L A is - L 2 -L 3 -.

In certain embodiments, L A is -L 2 -L 3 -L 4 -.

In certain embodiments, L A can be -CH2CH2-O-*, wherein * represents the point of attachment to Q 1 . In certain embodiments, L A can be -O-CH2CH2-*, wherein * represents the point of attachment to Q 1 .

In certain embodiments, L A can be -CH2-O-CH2-.

A

In certain embodiments, L A can be (such as wherein * represents the point of attachment to Q 1 .

Variable Q 1

In some embodiments, Q 1 is selected from the group consisting of:

• heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R c ’ ; and

• Ce-io aryl optionally substituted with 1-4 R c ’.

In certain of these embodiments, Q 1 is selected from the group consisting of:

• heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’.

In certain of the foregoing embodiments, Q 1 is selected from the group consisting of:

• heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’. In certain embodiments, Q 1 is phenyl optionally substituted with 1-3 R c ’. In certain of these embodiments, Q 1 is selected from the group consisting of:

In certain embodiments, Q 1 is heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 R c ’. In certain of these embodiments, Q 1 is pyridyl, which is optionally substituted with 1-3 R c ’. In certain of the foregoing embodiments, Q 1 is selected from the group consisting of:

In certain embodiments, Q 1 is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’.

In certain of these embodiments, Q 1 is heterocyclyl of 4-10 ring atoms, wherein 1-

3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with 1-

4 substituents independently selected from the group consisting of oxo and R c ’.

In certain of the foregoing embodiments, Q 1 is heterocyclyl of 4-8 ring atoms, wherein 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, provided that one ring atom is N(R d ), and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’. As non-limiting examples of the foregoing embodiments, Q 1 can or , wherein ml and m2 are each independently 0, 1, or 2; and wherein Q 1 is optionally substituted with 1-2 R c ’. For example, Q 1 can be . As another non-limiting example, Q 1 can be

In certain embodiments, each R d present in Q 1 is independently selected from the group consisting of: -C(O)O(Ci-4 alkyl); and Ci-6 alkyl optionally substituted with 1-3 independently selected R a .

In certain of the foregoing embodiments, each R d present in Q 1 is Ci-6 alkyl optionally substituted with 1-3 independently selected halo.

In certain of the foregoing embodiments, each R d present in Q 1 is Ci-4 alkyl substituted with 1-3 -F. In certain embodiments, each R d present in Q 1 is C2-3 alkyl substituted with 1-3 -F. For example, each R d present in Q 1 can be -CH2CF3.

In certain embodiments, each occurrence of R c ’ is an independently selected R c .

In certain embodiments, each occurrence of R c ’ is independently selected from the group consisting of: (i) C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 independently selected R c ; (ii) heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected R c ; (iii) heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R c ; and (iv) Ce-io aryl optionally substituted with 1-4 R c . In certain embodiments, each occurrence of R c ’ is any combination of an independently selected R c and a cyclic moiety independently selected from the group consisting of: (i) C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 independently selected R c ; (ii) heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected R c ; (iii) heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R c ; and (iv) Ce-io aryl optionally substituted with 1-4 R c .

In certain embodiment, the cyclic moiety is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected R c .

In certain embodiment, the cyclic moiety is Ce-io aryl optionally substituted with 1- 4 R c .

In certain embodiments, each R c present in Q 1 is independently selected from the group consisting of: halo; cyano; Ci-4 alkoxy; Ci-4 haloalkoxy; and C1-10 alkyl which is optionally substituted with 1-6 independently selected R a .

In certain embodiments, each R c present in Q 1 is independently selected from the group consisting of: halo; cyano; Ci-4 alkoxy; Ci-4 haloalkoxy; and C1-6 alkyl which is optionally substituted with 1-6 independently selected halo.

In certain of the foregoing embodiments, each R c present in Q 1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo.

In certain embodiments, each R c present in Q 1 is C1-3 alkyl which is optionally substituted with 1-6 -F. For example, each R c present in Q 1 can be CF3.

In certain embodiments, each R c present in Q 1 is an independently selected halo (e.g., -F or -Cl). Variables Y 1 , Y 2 , Y 3 , X 1 , and X 2

In some embodiments, Y 1 is CR 1 .

In some embodiments, Y 2 is CR 1 .

In some embodiments, Y 3 is CR 1 .

In certain embodiments, each occurrence of R 1 is independently H or R c . In certain of these embodiments, each occurrence of R 1 is H.

In certain other embodiments, 1-2 occurrence of R 1 is R c ; and each remaining occurrence of R 1 is H. For example, one occurrence of R 1 can be halo (e.g., -F or -Cl); and each remaining occurrence of R 1 can be H.

In certain embodiments, Y 1 , Y 2 , and Y 3 are each independently selected CR 1 .

In certain embodiments, Y 1 , Y 2 , and Y 3 are each CH.

In certain embodiments, one of Y 1 , Y 2 , and Y 3 is CR C , optionally C-halo; and each of the remaining two Y 1 , Y 2 , and Y 3 is CH.

In some embodiments, X 1 is NR 2 . In certain of these embodiments, X 1 is NH.

In some embodiments, X 2 is CR 5 . In certain of these embodiments, X 2 is CH.

In certain embodiments, X 1 is NR 2 ; and X 2 is CR 5 . In certain of the foregoing embodiments, X 1 is NH; and X 2 is CH.

In certain embodiments, Y 1 , Y 2 , and Y 3 are each an independently selected CR 1 ; X 1 is NR 2 ; and X 2 is CR 5 . In certain of the foregoing embodiments, Y 1 , Y 2 , and Y 3 are each CH; X 1 is NH; and X 2 is CH.

Variables R 6 and W

In some embodiments, R 6 is H.

In some embodiments, W is Ci-io alkyl, C2-10 alkenyl, or C2-10 alkenyl, each of which is optionally substituted with 1-6 R a2 . In certain of these embodiments, W is Ci-io alkyl, which is optionally substituted with 1-6 R a2 . In certain of the foregoing embodiments, W is Ci-6 alkyl, which is optionally substituted with 1-6 R a2 .

In some embodiments, W is Ci-io alkyl, C2-10 alkenyl, or C2-10 alkenyl, each of which is optionally substituted with 1-6 R a2 or R g .

In certain of these embodiments, W is C1-10 alkyl, which is optionally substituted with 1-6 R a2 or R g . In certain of the foregoing embodiments, W is C1-6 alkyl, which is optionally substituted with 1-6 R a2 or R g .

In certain embodiments, W is Ci-4 alkyl, which is optionally substituted with 1-6 R a2 or R g . In certain embodiments, W is Ci-4 alkyl, which is optionally substituted with 1- 6 R a2 .

In certain embodiments, W is Ci-4 alkyl, which is optionally substituted with 1-6

R g .

In certain embodiments, W is Ci-4 alkyl substituted one R g .

In certain embodiments, W can

In certain of the foregoing embodiments, W is unsubstituted Ci-4 alkyl. As nonlimiting examples of the foregoing embodiments, W can be selected from the group consisting of: methyl, ethyl, n-propyl, isopropyl, and isobutyl. For example, W can be methyl or ethyl.

In certain embodiments, W is Ci-4 alkyl, which is substituted with 1-6 R a2

In certain of these embodiments, each R a2 is independently selected from the group consisting of: -OH; -halo; -NR e R f ; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=O)O(Ci-4 alkyl); - C(=O)(Ci-4 alkyl); and cyano. For example, each R a2 can be independently selected from the group consisting of halo; -OH; Ci-4 alkoxy; and Ci-4 haloalkoxy.

In certain embodiments, W is Ci-4 alkyl which is substituted with 1-3 substituents each independently selected from the group consisting of: halo; -OH; Ci-4 alkoxy; and Ci-

4 haloalkoxy. As non-limiting examples, W can be /^°X

In some embodiments, W is selected from the group consisting of:

• monocyclic C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• monocyclic heterocyclyl or heterocycloalkenyl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain of the foregoing embodiments, W is monocyclic C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain of these embodiments, W is monocyclic C3-8 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments, W is unsubstituted C3-8 cycloalkyl. As non-limiting examples of the foregoing embodiments, W can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. For example, W can be cyclobutyl.

In some embodiments, W is selected from the group consisting of: • bicyclic C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• bicylic heterocyclyl or heterocycloalkenyl of from 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In some embodiments, W is selected from the group consisting of:

• bicyclic C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• bicylic heterocyclyl or heterocycloalkenyl of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In some embodiments, W is selected from the group consisting of:

• bicyclic C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• bicylic heterocyclyl or heterocycloalkenyl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c . In certain of the foregoing embodiments, W is bicyclic C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain of these embodiments, W is bicyclic C3-8 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments, W is unsubstituted bicyclic C3-8 cycloalkyl.

In certain of these embodiments, W is bicyclic C3-8 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments, W is unsubstituted bicyclic C3-8 cycloalkyl.

In certain of the forgoing embodiments, W is bicylic heterocyclyl or heterocycloalkenyl of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .In certain of the forgoing embodiments, W is bicylic heterocyclyl or heterocycloalkenyl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain of the forgoing embodiments, W is bicylic heterocyclyl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain of the forgoing embodiments, W can be In certain embodiments, W is heteroaryl of from 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments, W is heteroaryl of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments, W is heteroaryl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

R c

In certain embodiments, W can be

Non-Limiting Combinations

In certain embodiments, the compound is a compound of Formula (I-a):

Formula (I-a) or a pharmaceutically acceptable salt thereof, wherein:

L 1 is selected from the group consisting of: -O-, -N(H)-, and -N(R d )-;

L 2 is selected from the group consisting of:

• straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b ; • C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

In certain embodiments of Formula (I-a), L 1 is -O-.

In certain embodiments of Formula (I-a), L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

In certain embodiments of Formula (I-a), L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-, optionally wherein L 2 is -CH2-.

In certain embodiments of Formula (I-a), L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b . In certain of these embodiments, L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -Q 1 . For example, L 2 can be -CH2CH2-.

In certain embodiments of Formula (I-a), L 2 is straight-chain C3 alkylene which is optionally substituted with 1-3 R b .

In certain embodiments of Formula (I-a), L 2 is: which is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0, 1, or 2; Q 2 is CH, CR C , or N; and the asterisk represents the point of attachment to Q 1 .

In certain of these embodiments, nl and n2 are independently 0 or 1, optionally 0; and Q 2 is CH. For example, nl and n2 can both be 0; and Q 2 can be CH, e.g., L 2 can be optionally substituted optionally substituted cyclobutane-diyl, e.g, optionally substituted cyclobutane- 1 , 3 -diyl .

In certain embodiments of Formula (I-a), L 1 is -O-; and L 2 is: is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0 or 1, optionally 0; and Q 2 is CH. For example, nl and n2 can both be 0; and Q 2 can be CH, e.g., L 2 can be optionally substituted cyclobutane-diyl, e.g, optionally substituted 1,3- cyclobutane-l,3-diyl, e.g., unsubstituted cyclobutane-diyl, e.g, unsubstituted cyclobutane- 1,3-diyl.

In certain embodiments of Formula (I-a), L 1 is -O-; and L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

In certain of the foregoing embodiments of Formula (I-a), L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b .

In certain of the foregoing embodiments, L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -Q 1 . For example, L 2 can be -CH2CH2-.

In certain embodiments of Formula (I-a), L 1 is -O-; and L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2. For example, L 2 can be -CH2-.

In certain embodiments, the compound is a compound of Formula (I-b):

Formula (I-b) or a pharmaceutically acceptable salt thereof, wherein:

L 2 is straight-chain C1-6 alkylene or straight-chain C2-6 alkenylene, each of which is optionally substituted with 1-6 R b .

In certain embodiments of Formula (I-b), L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

In certain embodiments of Formula (I-b), L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b . In certain of these embodiments, L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -Q 1 . For example, L 2 can be -CH2CH2-. In certain embodiments of Formula (I-b), L 2 is straight-chain C3 alkylene which is optionally substituted with 1-3 R b . In certain of these embodiments, L 2 is selected from the group consisting of: wherein the asterisk represents point of attachment to -Q 1 . For example, L 2 can be

In certain embodiments of Formula (I-b), L 2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 R b .

In certain of these embodiments, L 2 is selected from the group consisting of: , wherein the asterisk represents the point of attachment to -

Q 1

In certain embodiments, the compound is a compound of Formula (I-c):

Formula (I-c) or a pharmaceutically acceptable salt thereof, wherein:

L 2 and L 4 are independently selected straight-chain C1-3 alkylene which is optionally substituted with 1-6 R b ; and

L 3 is selected from the group consisting of: -O-, -N(H)-, and -N(R d )-.

In certain embodiments of Formula (I-c), L 2 and L 4 are independently selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2. In certain of these embodiments, L 2 and L 4 are each -CH2-.

In certain embodiments of Formula (I-c), L 3 is -O-. In certain embodiments of Formula (I-c), L 3 is -N(H)- or -N(R d )-. For example, L 3 can be -N(H)-.

In certain embodiments, the compound is a compound of Formula (I-d):

Formula (I-d) or a pharmaceutically acceptable salt thereof, wherein:

L 2 is straight-chain C1-3 alkylene which is optionally substituted with 1-6 R b ; and

L 3 is selected from the group consisting of: -O-, -N(H)-, and -N(R d )-.

In certain embodiments of Formula (I-d), L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b ) 2 .

In certain embodiments of Formula (I-d), L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b . In certain of these embodiments, L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -L 3 . For example, L 2 can be -CH2CH2-.

In certain embodiments of Formula (I-d), L 3 is -O-.

In certain embodiments of Formula (I-d), L 3 is -N(H)- or -N(R d )-. For example, L 3 can be -N(H)-.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is selected from the group consisting of:

• heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’. In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is selected from the group consisting of:

• heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is phenyl or pyridyl, each optionally substituted with 1-3 R c ’.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is phenyl or pyridyl, each optionally substituted with 1-3 R c ’, wherein each R c present in Q 1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is is independently selected from the group consisting of: -F, -Cl, and -CF3.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’. In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is: , wherein ml and m2 are each independently 0, 1, or 2.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is: the R d present in Q 1 is selected from the group consisting of: -C(O)O(Ci-4 alkyl); and Ci-6 alkyl optionally substituted with 1-3 independently selected R a ; or wherein the R d present in Q 1 is C2-3 alkyl substituted with 1-3 -F.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q 1 is:

; and the R d present in Q 1 is selected from the group consisting of: -C(O)O(Ci-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected R a ; or wherein the R d present in Q 1 is C2-3 alkyl substituted with 1-3 -F.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), R c is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected R c .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), R c is Ce-io aryl optionally substituted with 1-4 R c .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), each R 1 is H. In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), one occurrence of R 1 is R c ; and each remaining R 1 is H.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), R 2 is H; and R 5 is H.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is Ci-6 alkyl, which is optionally substituted with 1-6 R a2 .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is unsubstituted Ci-4 alkyl. For example, W can be methyl or ethyl.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is Ci-4 alkyl, which is substituted with 1-6 R a2 .

In certain of these embodiments, W is: Ci-4 alkyl which is substituted with 1-3 substituents each independently selected from the group consisting of: halo; -OH; Ci-4 alkoxy; and Ci-4 haloalkoxy.

As non-limiting examples of the foregoing embodiments, W can be

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is Ci-6 alkyl, which is optionally substituted with 1-6 R g .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is Ci-4 alkyl, which is substituted with 1-6 R g .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W. can be

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is selected from the group consisting of: • C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• heteroaryl, heterocyclyl or heterocycloalkenyl of from 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is selected from the group consisting of:

• C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• heteroaryl, heterocyclyl or heterocycloalkenyl of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is selected from the group consisting of:

• C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• Heteroaryl, heterocyclyl or heterocycloalkenyl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c . In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is monocyclic C3- 8 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c . In certain of these embodiments, W is unsubstituted C3-8 cycloalkyl. For example, W can be cyclobutyl.

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is heteroaryl of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c . In certain embodiments,

W can

In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is heterocycly of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c . In certain embodiments, W is unsubstituted heterocycly of from 3-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, In certain embodiments, W can be

Non-Limiting Exemplary Compounds

In some embodiments, the compound is selected from the group consisting of the compounds delineated in Table Cl or a pharmaceutically acceptable salt thereof.

Table Cl

Pharmaceutical Compositions and Administration

General In some embodiments, a chemical entity (e.g., a compound that inhibits (e.g., antagonizes) STING, or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination thereof) is administered as a pharmaceutical composition that includes the chemical entity and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein.

In some embodiments, the chemical entities can be administered in combination with one or more conventional pharmaceutical excipients. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, and wool fat. Cyclodextrins such as a-, 0, and y-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl-P-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein. Dosage forms or compositions containing a chemical entity as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared. The contemplated compositions may contain 0.001%-100% of a chemical entity provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22 nd Edition (Pharmaceutical Press, London, UK. 2012). Routes of Administration and Composition Components

In some embodiments, the chemical entities described herein or a pharmaceutical composition thereof can be administered to subject in need thereof by any accepted route of administration. Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intraci sternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, nasal, nasogastric, oral, parenteral, percutaneous, peridural, rectal, respiratory (inhalation), subcutaneous, sublingual, submucosal, topical, transdermal, transmucosal, transtracheal, ureteral, urethral and vaginal. In certain embodiments, a preferred route of administration is parenteral (e.g., intratumoral).

Compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. The preparation of such formulations will be known to those of skill in the art in light of the present disclosure.

The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.

The carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof.

Intratumoral injections are discussed, e.g., in Lammers, et al., “Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems” Neoplasia. 2006, 10, 788-795.

Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository, include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p- oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylocaprate, isopropyl alcohol, propylene glycol, liquid paraffin, xanthan gum, carboxy-metabisulfite, sodium edetate, sodium benzoate, potassium metabisulfite, grapefruit seed extract, methyl sulfonyl methane (MSM) , lactic acid, glycine, vitamins, such as vitamin A and E and potassium acetate.

In certain embodiments, suppositories can be prepared by mixing the chemical entities described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound. In other embodiments, compositions for rectal administration are in the form of an enema.

In other embodiments, the compounds described herein or a pharmaceutical composition thereof are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.).

Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the chemical entity is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

In one embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a chemical entity provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEG’s, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which one or more chemical entities provided herein or additional active agents are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two- compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.

Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid.

In certain embodiments the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient.

In certain embodiments, solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the chemical entity to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel. Exemplary formulation techniques are described in, e.g., Filipski, K.J., et al., Current Topics in Medicinal Chemistry, 2013, 13, 776-802, which is incorporated herein by reference in its entirety.

Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls.

Other examples include lower-GI targeting techniques. For targeting various regions in the intestinal tract, several enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid-methyl methacrylate copolymers), and Marcoat). Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap.

Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, ETDA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxychloro complex; Allergan, Inc.)).

Topical compositions can include ointments and creams. Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives. Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil. Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase, also sometimes called the “internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and nonsensitizing.

In any of the foregoing embodiments, pharmaceutical compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers.

Dosages

The dosages may be varied depending on the requirement of the patient, the severity of the condition being treating and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts. The total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery.

In some embodiments, the compounds described herein are administered at a dosage of from about 0.001 mg/Kg to about 500 mg/Kg (e.g., from about 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0. 1 mg/Kg to about 100 mg/Kg; from about 0. 1 mg/Kg to about 10 mg/Kg).

Regimens

The foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month).

In some embodiments, the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In an embodiment, a therapeutic compound is administered to an individual for a period of time followed by a separate period of time. In another embodiment, a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped. In an aspect of this embodiment, the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time. In a further embodiment, a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.

Methods of Treatment

In some embodiments, methods for treating a subject having condition, disease or disorder in which increased (e.g., excessive)STING activity (e.g., , e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., immune disorders, cancer) are provided.

Indications

In some embodiments, the condition, disease or disorder is cancer. Non-limiting examples of cancer include melanoma, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include breast cancer, colon cancer, rectal cancer, colorectal cancer, kidney or renal cancer, clear cell cancer lung cancer including small-cell lung cancer, non- small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, squamous cell cancer (e.g. epithelial squamous cell cancer), cervical cancer, ovarian cancer, prostate cancer, prostatic neoplasms, liver cancer, bladder cancer, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, gastrointestinal stromal tumor, pancreatic cancer, head and neck cancer, glioblastoma, retinoblastoma, astrocytoma, thecomas, arrhenoblastomas, hepatoma, hematologic malignancies including non-Hodgkins lymphoma (NHL), multiple myeloma, myelodysplasia disorders, myeloproliferative disorders, chronic myelogenous leukemia, and acute hematologic malignancies, endometrial or uterine carcinoma, endometriosis, endometrial stromal sarcoma, fibrosarcomas, choriocarcinoma, salivary gland carcinoma, vulval cancer, thyroid cancer, esophageal carcinomas, hepatic carcinoma, anal carcinoma, penile carcinoma, nasopharyngeal carcinoma, laryngeal carcinomas, Kaposi's sarcoma, mast cell sarcoma, ovarian sarcoma, uterine sarcoma, melanoma, malignant mesothelioma, skin carcinomas, Schwannoma, oligodendroglioma, neuroblastomas, neuroectodermal tumor, rhabdomyosarcoma, osteogenic sarcoma, leiomyosarcomas, Ewing Sarcoma, peripheral primitive neuroectodermal tumor, urinary tract carcinomas, thyroid carcinomas, Wilm's tumor, as well as abnormal vascular proliferation associated with phakomatoses, edema (such as that associated with brain tumors), and Meigs' syndrome. In some cases, the cancer is melanoma.

In some embodiments, the condition, disease or disorder is a neurological disorder, which includes disorders that involve the central nervous system (brain, brainstem and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (parts of which are located in both central and peripheral nervous system). Non-limiting examples of neurological disorders include acquired epileptiform aphasia; acute disseminated encephalomyelitis; adrenoleukodystrophy; age-related macular degeneration; agenesis of the corpus callosum; agnosia; Aicardi syndrome; Alexander disease; Alpers' disease; alternating hemiplegia; Alzheimer's disease; Vascular dementia; amyotrophic lateral sclerosis; anencephaly; Angelman syndrome; angiomatosis; anoxia; aphasia; apraxia; arachnoid cysts; arachnoiditis; Anronl-Chiari malformation; arteriovenous malformation; Asperger syndrome; ataxia telegiectasia; attention deficit hyperactivity disorder; autism; autonomic dysfunction; back pain; Batten disease; Behcet's disease; Bell's palsy; benign essential blepharospasm; benign focal; amyotrophy; benign intracranial hypertension; Binswanger's disease; blepharospasm; Bloch Sulzberger syndrome; brachial plexus injury; brain abscess; brain injury; brain tumors (including glioblastoma multiforme); spinal tumor; Brown-Sequard syndrome; Canavan disease; carpal tunnel syndrome; causalgia; central pain syndrome; central pontine myelinolysis; cephalic disorder; cerebral aneurysm; cerebral arteriosclerosis; cerebral atrophy; cerebral gigantism; cerebral palsy; Charcot-Marie-Tooth disease; chemotherapy-induced neuropathy and neuropathic pain; Chiari malformation; chorea; chronic inflammatory demyelinating polyneuropathy; chronic pain; chronic regional pain syndrome; Coffin Lowry syndrome; coma, including persistent vegetative state; congenital facial diplegia; corticobasal degeneration; cranial arteritis; craniosynostosis; Creutzfeldt-Jakob disease; cumulative trauma disorders; Cushing's syndrome; cytomegalic inclusion body disease; cytomegalovirus infection; dancing eyes-dancing feet syndrome; Dandy-Walker syndrome; Dawson disease; De Morsier's syndrome; Dejerine-Klumke palsy; dementia; dermatomyositis; diabetic neuropathy; diffuse sclerosis; dysautonomia; dysgraphia; dyslexia; dystonias; early infantile epileptic encephalopathy; empty sella syndrome; encephalitis; encephaloceles; encephalotrigeminal angiomatosis; epilepsy; Erb's palsy; essential tremor; Fabry's disease; Fahr's syndrome; fainting; familial spastic paralysis; febrile seizures; Fisher syndrome; Friedreich's ataxia; fronto-temporal dementia and other “tauopathies”; Gaucher's disease; Gerstmann's syndrome; giant cell arteritis; giant cell inclusion disease; globoid cell leukodystrophy; Guillain-Barre syndrome; HTLV-1- associated myelopathy; Hallervorden-Spatz disease; head injury; headache; hemifacial spasm; hereditary spastic paraplegia; heredopathia atactica polyneuritiformis; herpes zoster oticus; herpes zoster; Hirayama syndrome; HIV-associated dementia and neuropathy (also neurological manifestations of AIDS); holoprosencephaly; Huntington's disease and other polyglutamine repeat diseases; hydranencephaly; hydrocephalus; hypercortisolism; hypoxia; immune-mediated encephalomyelitis; inclusion body myositis; incontinentia pigmenti; infantile phytanic acid storage disease; infantile refsum disease; infantile spasms; inflammatory myopathy; intracranial cyst; intracranial hypertension; Joubert syndrome; Kearns-Sayre syndrome; Kennedy disease Kinsboume syndrome; Klippel Feil syndrome; Krabbe disease; Kugelberg- Welander disease; kuru; Lafora disease; Lambert-Eaton myasthenic syndrome; Landau-Kleffner syndrome; lateral medullary (Wallenberg) syndrome; learning disabilities; Leigh's disease; Lennox-Gustaut syndrome; Lesch-Nyhan syndrome; leukodystrophy; Lewy body dementia; Lissencephaly; locked-in syndrome; Lou Gehrig's disease (i.e., motor neuron disease or amyotrophic lateral sclerosis); lumbar disc disease; Lyme disease — neurological sequelae; Machado-Joseph disease; macrencephaly; megalencephaly; Melkersson-Rosenthal syndrome; Menieres disease; meningitis; Menkes disease; metachromatic leukodystrophy; microcephaly; migraine; Miller Fisher syndrome; mini-strokes; mitochondrial myopathies; Mobius syndrome; monomelic amyotrophy; motor neuron disease; Moyamoya disease; mucopolysaccharidoses; milti-infarct dementia; multifocal motor neuropathy; multiple sclerosis and other demyelinating disorders; multiple system atrophy with postural hypotension; p muscular dystrophy; myasthenia gravis; myelinoclastic diffuse sclerosis; myoclonic encephalopathy of infants; myoclonus; myopathy; myotonia congenital; narcolepsy; neurofibromatosis; neuroleptic malignant syndrome; neurological manifestations of AIDS; neurological sequelae of lupus; neuromyotonia; neuronal ceroid lipofuscinosis; neuronal migration disorders; Niemann-Pick disease; O'Sullivan-McLeod syndrome; occipital neuralgia; occult spinal dysraphism sequence; Ohtahara syndrome; olivopontocerebellar atrophy; opsoclonus myoclonus; optic neuritis; orthostatic hypotension; overuse syndrome; paresthesia; Parkinson's disease; paramyotonia congenital; paraneoplastic diseases; paroxysmal attacks; Parry Romberg syndrome; Pelizaeus-Merzbacher disease; periodic paralyses; peripheral neuropathy; painful neuropathy and neuropathic pain; persistent vegetative state; pervasive developmental disorders; photic sneeze reflex; phytanic acid storage disease; Pick's disease; pinched nerve; pituitary tumors; polymyositis; porencephaly; post-polio syndrome; postherpetic neuralgia; postinfectious encephalomyelitis; postural hypotension; Prader-Willi syndrome; primary lateral sclerosis; prion diseases; progressive hemifacial atrophy; progressive multifocal leukoencephalopathy; progressive sclerosing poliodystrophy; progressive supranuclear palsy; pseudotumor cerebri; Ramsay-Hunt syndrome (types I and II); Rasmussen's encephalitis; reflex sympathetic dystrophy syndrome; Refsum disease; repetitive motion disorders; repetitive stress injuries; restless legs syndrome; retrovirus- associated myelopathy; Rett syndrome; Reye's syndrome; Saint Vitus dance; Sandhoff disease; Schilder's disease; schizencephaly; septo-optic dysplasia; shaken baby syndrome; shingles; Shy -Drager syndrome; Sjogren's syndrome; sleep apnea; Soto's syndrome; spasticity; spina bifida; spinal cord injury; spinal cord tumors; spinal muscular atrophy; Stiff-Person syndrome; stroke; Sturge-Weber syndrome; subacute sclerosing panencephalitis; subcortical arteriosclerotic encephalopathy; Sydenham chorea; syncope; syringomyelia; tardive dyskinesia; Tay-Sachs disease; temporal arteritis; tethered spinal cord syndrome; Thomsen disease; thoracic outlet syndrome; Tic Douloureux; Todd's paralysis; Tourette syndrome; transient ischemic attack; transmissible spongiform encephalopathies; transverse myelitis; traumatic brain injury; tremor; trigeminal neuralgia; tropical spastic paraparesis; tuberous sclerosis; vascular dementia (multi-infarct dementia); vasculitis including temporal arteritis; Von Hippel-Lindau disease; Wallenberg's syndrome; Werdnig-Hoffman disease; West syndrome; whiplash; Williams syndrome; Wildon's disease; amyotrophe lateral sclerosis and Zellweger syndrome.

In some embodiments, the condition, disease or disorder is STING-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis. In certain embodiments, the condition, disease or disorder is an autoimmune disease (e.g., a cytosolic DNA-triggered autoinflammatory disease). Non-limiting examples include rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel diseases (IBDs) comprising Crohn disease (CD) and ulcerative colitis (UC), which are chronic inflammatory conditions with polygenic susceptibility. In certain embodiments, the condition is an inflammatory bowel disease. In certain embodiments, the condition is Crohn’s disease, autoimmune colitis, iatrogenic autoimmune colitis, ulcerative colitis, colitis induced by one or more chemotherapeutic agents, colitis induced by treatment with adoptive cell therapy, colitis associated by one or more alloimmune diseases (such as graft-vs-host disease, e.g., acute graft vs. host disease and chronic graft vs. host disease), radiation enteritis, collagenous colitis, lymphocytic colitis, microscopic colitis, and radiation enteritis. In certain of these embodiments, the condition is alloimmune disease (such as graft-vs-host disease, e.g., acute graft vs. host disease and chronic graft vs. host disease), celiac disease, irritable bowel syndrome, rheumatoid arthritis, lupus, scleroderma, psoriasis, cutaneous T-cell lymphoma, uveitis, and mucositis (e.g., oral mucositis, esophageal mucositis or intestinal mucositis).

In some embodiments, modulation of the immune system by STING provides for the treatment of diseases, including diseases caused by foreign agents. Exemplary infections by foreign agents which may be treated and/or prevented by the method of the present invention include an infection by a bacterium (e.g., a Gram-positive or Gramnegative bacterium), an infection by a fungus, an infection by a parasite, and an infection by a virus. In one embodiment of the present invention, the infection is a bacterial infection (e.g., infection by E. coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Salmonella spp., Staphylococcus aureus, Streptococcus spp., or vancomycin-resistant enterococcus), or sepsis. In another embodiment, the infection is a fungal infection (e.g. infection by a mould, a yeast, or a higher fungus). In still another embodiment, the infection is a parasitic infection (e.g., infection by a single-celled or multicellular parasite, including Giardia duodenalis, Cryptosporidium parvum, Cyclospora cayetanensis, and Toxoplasma gondiz). In yet another embodiment, the infection is a viral infection (e.g., infection by a virus associated with AIDS, avian flu, chickenpox, cold sores, common cold, gastroenteritis, glandular fever, influenza, measles, mumps, pharyngitis, pneumonia, rubella, SARS, and lower or upper respiratory tract infection (e.g., respiratory syncytial virus)).

In some embodiments, the condition, disease or disorder is hepatits B (see, e.g., WO 2015/061294).

In some embodiments, the condition, disease or disorder is selected from cardiovascular diseases (including e.g., myocardial infarction).

In some embodiemnts, the condition, disease or disorder is age-related macular degeneration.

In some embodiments, the condition, disease or disorder is mucositis, also known as stomatitits, which can occur as a result of chemotherapy or radiation therapy, either alone or in combination as well as damage caused by exposure to radiation outside of the context of radiation therapy.

In some embodiments, the condition, disease or disorder is uveitis, which is inflammation of the uvea (e.g., anterior uveitis, e.g., iridocyclitis or iritis; intermediate uveitis (also known as pars planitis); posterior uveitis; or chorioretinitis, e.g., pan-uveitis).

In some embodiments, the condition, disease or disorder is selected from the group consisting of a cancer, a neurological disorder, an autoimmune disease, hepatitis B, uvetitis, a cardiovascular disease, age-related macular degeneration, and mucositis.

Still other examples can include those indications discussed herein and below in contemplated combination therapy regimens.

Combination therapy

This disclosure contemplates both monotherapy regimens as well as combination therapy regimens. In some embodiments, the methods described herein can further include administering one or more additional therapies (e.g., one or more additional therapeutic agents and/or one or more therapeutic regimens) in combination with administration of the compounds described herein.

In certain embodiments, the methods described herein can further include administering one or more additional cancer therapies.

The one or more additional cancer therapies can include, without limitation, surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy, cancer vaccines (e.g., HPV vaccine, hepatitis B vaccine, Oncophage, Provenge) and gene therapy, as well as combinations thereof. Immunotherapy, including, without limitation, adoptive cell therapy, the derivation of stem cells and/or dendritic cells, blood transfusions, lavages, and/or other treatments, including, without limitation, freezing a tumor.

In some embodiments, the one or more additional cancer therapies is chemotherapy, which can include administering one or more additional chemotherapeutic agents.

In certain embodiments, the additional chemotherapeutic agent is an immunomodulatory moiety, e.g., an immune checkpoint inhibitor. In certain of these embodiments, the immune checkpoint inhibitor targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD-1 - PD- L2, interleukin -2 (IL -2), indoleamine 2,3 -dioxygenase (IDO), IL - 10, transforming growth factor-P (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM - LIGHT, HVEM-BTLA-CD160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48 - CD244, CD244, ICOS, ICOS-ICOS ligand, B7 H3, B7 H4, VISTA, TMIGD2, HHLA2- TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39-CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155; e.g., CTLA-4 orPDl or PD-L1). See, e.g., Postow, M. J. Clin. Oncol. 2015, 33, 1.

In certain of these embodiments, the immune checkpoint inhibitor is selected from the group consisting of: Urelumab, PF-05082566, MEDI6469, TRX518, Varlilumab, CP-870893, Pembrolizumab (PD1), Nivolumab (PD1), Atezolizumab (formerly MPDL3280A) (PDL1), MEDI4736 (PD-L1), Avelumab (PD-L1), PDR001 (PD1), BMS-986016, MGA271, Lirilumab, IPH2201, Emactuzumab, INCB024360, Galunisertib, Ulocuplumab, BKT140, Bavituximab, CC-90002, Bevacizumab, and MNRP1685A, and MGA271.

In certain embodiments, the additional chemotherapeutic agent is an alkylating agent. Alkylating agents are so named because of their ability to alkylate many nucleophilic functional groups under conditions present in cells, including, but not limited to cancer cells. In a further embodiment, an alkylating agent includes, but is not limited to, Cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin. In an embodiment, alkylating agents can function by impairing cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules or they can work by modifying a cell's DNA. In a further embodiment an alkylating agent is a synthetic, semisynthetic or derivative.

In certain embodiments, the additional chemotherapeutic agent is an antimetabolite. Anti-metabolites masquerade as purines or pyrimidines, the building-blocks of DNA and in general, prevent these substances from becoming incorporated in to DNA during the "S" phase (of the cell cycle), stopping normal development and division. Antimetabolites can also affect RNA synthesis. In an embodiment, an antimetabolite includes, but is not limited to azathioprine and/or mercaptopurine. In a further embodiment an antimetabolite is a synthetic, semisynthetic or derivative.

In certain embodiments, the additional chemotherapeutic agent is a plant alkaloid and/or terpenoid. These alkaloids are derived from plants and block cell division by, in general, preventing microtubule function. In an embodiment, a plant alkaloid and/or terpenoid is a vinca alkaloid, a podophyllotoxin and/or a taxane. Vinca alkaloids, in general, bind to specific sites on tubulin, inhibiting the assembly of tubulin into microtubules, generally during the M phase of the cell cycle. In an embodiment, a vinca alkaloid is derived, without limitation, from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea). In an embodiment, a vinca alkaloid includes, without limitation, Vincristine, Vinblastine, Vinorelbine and/or Vindesine. In an embodiment, a taxane includes, but is not limited, to Taxol, Paclitaxel and/or Docetaxel. In a further embodiment a plant alkaloid or terpernoid is a synthetic, semisynthetic or derivative. In a further embodiment, a podophyllotoxin is, without limitation, an etoposide and/or teniposide. In an embodiment, a taxane is, without limitation, docetaxel and/or ortataxel. [021] In an embodiment, a cancer therapeutic is a topoisomerase. Topoisomerases are essential enzymes that maintain the topology of DNA. Inhibition of type I or type II topoisomerases interferes with both transcription and replication of DNA by upsetting proper DNA supercoiling. In a further embodiment, a topoisomerase is, without limitation, a type I topoisomerase inhibitor or a type II topoisomerase inhibitor. In an embodiment a type I topoisomerase inhibitor is, without limitation, a camptothecin. In another embodiment, a camptothecin is, without limitation, exatecan, irinotecan, lurtotecan, topotecan, BNP 1350, CKD 602, DB 67 (AR67) and/or ST 1481. In an embodiment, a type II topoisomerase inhibitor is, without limitation, epipodophyllotoxin. In a further embodiment an epipodophyllotoxin is, without limitation, an amsacrine, etoposid, etoposide phosphate and/or teniposide. In a further embodiment a topoisomerase is a synthetic, semisynthetic or derivative, including those found in nature such as, without limitation, epipodophyllotoxins, substances naturally occurring in the root of American Mayapple (Podophyllum peltatum).

In certain embodiments, the additional chemotherapeutic agent is a stilbenoid. In a further embodiment, a stilbenoid includes, but is not limited to, Resveratrol, Piceatannol, Pinosylvin, Pterostilbene, Alpha- Viniferin, Ampelopsin A, Ampelopsin E, Diptoindonesin C, Diptoindonesin F, Epsilon- Vinferin, Flexuosol A, Gnetin H, Hemsleyanol D, Hopeaphenol, Trans-Diptoindonesin B, Astringin, Piceid and Diptoindonesin A. In a further embodiment a stilbenoid is a synthetic, semisynthetic or derivative.

In certain embodiments, the additional chemotherapeutic agent is a cytotoxic antibiotic. In an embodiment, a cytotoxic antibiotic is, without limitation, an actinomycin, an anthracenedione, an anthracycline, thalidomide, dichloroacetic acid, nicotinic acid, 2- deoxyglucose and/or chlofazimine. In an embodiment, an actinomycin is, without limitation, actinomycin D, bacitracin, colistin (polymyxin E) and/or polymyxin B. In another embodiment, an antracenedione is, without limitation, mitoxantrone and/or pixantrone. In a further embodiment, an anthracycline is, without limitation, bleomycin, doxorubicin (Adriamycin), daunorubicin (daunomycin), epirubicin, idarubicin, mitomycin, plicamycin and/or valrubicin. In a further embodiment a cytotoxic antibiotic is a synthetic, semi synthetic or derivative.

In certain embodiments, the additional chemotherapeutic agent is selected from endostatin, angiogenin, angiostatin, chemokines, angioarrestin, angiostatin (plasminogen fragment), basement-membrane collagen-derived anti -angiogenic factors (tumstatin, canstatin, or arrestin), anti-angiogenic antithrombin III, signal transduction inhibitors, cartilage-derived inhibitor (CDI), CD59 complement fragment, fibronectin fragment, gro- beta, heparinases, heparin hexasaccharide fragment, human chorionic gonadotropin (hCG), interferon alpha/beta/gamma, interferon inducible protein (IP- 10), interleukin- 12, kringle 5 (plasminogen fragment), metalloproteinase inhibitors (TIMPs), 2-methoxyestradiol, placental ribonuclease inhibitor, plasminogen activator inhibitor, platelet factor-4 (PF4), prolactin 16 kD fragment, proliferin-related protein (PRP), various retinoids, tetrahydrocortisol-S, thrombospondin- 1 (TSP-1), transforming growth factor-beta (TGF- P), vasculostatin, vasostatin (calreticulin fragment) and the like.

In certain embodiments, the additional chemotherapeutic agent is selected from abiraterone acetate, altretamine, anhydrovinblastine, auri statin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly-l-Lpro line-t- butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3',4'-didehydro-4'- deoxy-8'-norvin-caleukoblastine, docetaxol, doxetaxel, cyclophosphamide, carboplatin, carmustine, cisplatin, cryptophycin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, daunorubicin, decitabine dolastatin, doxorubicin (adriamycin), etoposide, 5- fluorouracil, finasteride, flutamide, hydroxyurea and hydroxyureataxanes, ifosfamide, liarozole, lonidamine, lomustine (CCNU), MDV3100, mechlorethamine (nitrogen mustard), melphalan, mivobulin isethionate, rhizoxin, sertenef, streptozocin, mitomycin, methotrexate, taxanes, nilutamide, onapristone, paclitaxel, prednimustine, procarbazine, RPR109881, stramustine phosphate, tamoxifen, tasonermin, taxol, tretinoin, vinblastine, vincristine, vindesine sulfate, and vinflunine.

In certain embodiments, the additional chemotherapeutic agent is platinum, cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, azathioprine, mercaptopurine, vincristine, vinblastine, vinorelbine, vindesine, etoposide and teniposide, paclitaxel, docetaxel, irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide, 5-fluorouracil, leucovorin, methotrexate, gemcitabine, taxane, leucovorin, mitomycin C, tegafur-uracil, idarubicin, fludarabine, mitoxantrone, ifosfamide and doxorubicin. Additional agents include inhibitors of mTOR (mammalian target of rapamycin), including but not limited to rapamycin, everolimus, temsirolimus and deforolimus.

In still other embodiments, the additional chemotherapeutic agent can be selected from those delineated in U.S. Patent 7,927,613, which is incorporated herein by reference in its entirety.

In some embodiments, the additional therapeutic agent and/or regimen are those that can be used for treating other STING-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis and the like.

Non-limiting examples of additional therapeutic agents and/or regimens for treating rheumatoid arthritis include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), corticosteroids (e.g, prednisone), disease-modifying antirheumatic drugs (DMARDs; e.g., methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), leflunomide (Arava®), hydroxychloroquine (Plaquenil), PF-06650833, iguratimod, tofacitinib (Xeljanz®), ABBV-599, evobrutinib, and sulfasalazine (Azulfidine®)), and biologies (e.g., abatacept (Orencia®), adalimumab (Humira®), anakinra (Kineret®), certolizumab (Cimzia®), etanercept (Enbrel®), golimumab (Simponi®), infliximab (Remicade®), rituximab (Rituxan®), tocilizumab (Actemra®), vobarilizumab, sarilumab (Kevzara®), secukinumab, ABP 501, CHS-0214, ABC-3373, and tocilizumab (ACTEMRA®)). Non-limiting examples of additional therapeutic agents and/or regimens for treating lupus include steroids, topical immunomodulators (e.g., tacrolimus ointment (Protopic®) and pimecrolimus cream (Elidel®)), thalidomide (Thalomid®), non-steroidal antiinflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), antimalarial drugs (e.g., Hydroxychloroquine (Plaquenil)), corticosteroids (e.g, prednisone) and immunomodulators (e.g., evobrutinib, iberdomide, voclosporin, cenerimod, azathioprine (Imuran®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral, Sandimmune®, Gengraf®), and mycophenolate mofetil) baricitinb, iguratimod, filogotinib, GS-9876, rapamycin, and PF-06650833), and biologies (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, obinutuzumab, vobarilizumab, lulizumab, atacicept, PF-06823859, and lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-a-kinoid, OMS721, RC18, RSLV- 132, theralizumab, XmAb5871, and ustekinumab (Stelara®)). For example, non-limiting treatments for systemic lupus erythematosus include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), antimalarial drugs (e.g., Hydroxychloroquine (Plaquenil)), corticosteroids (e.g, prednisone) and immunomodulators (e.g., iberdomide, voclosporin, azathioprine (Imuran®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral, Sandimmune®, Gengraf®), and mycophenolate mofetil, baricitinb, filogotinib, and PF-06650833), and biologies (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, vobarilizumab, lulizumab, atacicept, PF-06823859, lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-a-kinoid, RC18, RSLV-132, theralizumab, XmAb5871, and ustekinumab (Stelara®)). As another example, non-limiting examples of treatments for cutaneous lupus include steroids, immunomodulators (e.g., tacrolimus ointment (Protopic®) and pimecrolimus cream (Elidel®)), GS-9876, filogotinib, and thalidomide (Thalomid®). Agents and regimens for treating drug-induced and/or neonatal lupus can also be administered.

Non-limiting examples of additional therapeutic agents and/or regimens for treating STING-associated vasculopathy with onset in infancy (SAVI) include JAK inhibitors (e.g., tofacitinib, ruxolitinib, filgotinib, and baricitinib). Non-limiting examples of additional therapeutic agents and/or regimens for treating Aicardi-Goutieres Syndrome (AGS) include physiotherapy, treatment for respiratory complications, anticonvulsant therapies for seizures, tube-feeding, nucleoside reverse transcriptase inhibitors (e.g., emtricitabine (e.g., Emtriva®), tenofovir (e.g., Viread®), emtricitabine/tenofovir (e.g., Truvada®), zidovudine, lamivudine, and abacavir), and JAK inhibitors (e.g., tofacitinib, ruxolitinib, filgotinib, and baricitinib).

Non-limiting examples of additional therapeutic agents and/or regimens for treating IBDs include 6-mercaptopurine, AbGn-168H, ABX464, ABT-494, adalimumab, AJM300, alicaforsen, AMG139, anrukinzumab, apremilast, ATR-107 (PF0530900), autologous CD34-selected peripheral blood stem cells transplant, azathioprine, bertilimumab, BI 655066, BMS-936557, certolizumab pegol (Cimzia®), cobitolimod, corticosteroids (e.g., prednisone, Methylprednisolone, prednisone), CP-690,550, CT-P13, cyclosporine, DIMS0150, E6007, E6011, etrasimod, etrolizumab, fecal microbial transplantation, figlotinib, fmgolimod, firategrast (SB-683699) (formerly T-0047), GED0301, GLPG0634, GLPG0974, guselkumab, golimumab, GSK 1399686, HMPL-004 (Andrographis paniculata extract), IMU-838, infliximab, Interleukin 2 (IL-2), Janus kinase (JAK) inhibitors, laquinimod, masitinib (AB 1010), matrix metalloproteinase 9 (MMP 9) inhibitors (e.g., GS-5745), MEDI2070, mesalamine, methotrexate, mirikizumab (LY3074828), natalizumab, NNC 0142-0000-0002, NNC0114-0006, ozanimod, peficitinib (JNJ-54781532), PF-00547659, PF-04236921, PF-06687234, QAX576, RHB- 104, rifaximin, risankizumab, RPC1063, SB012, SHP647, sulfasalazine, TD-1473, thalidomide, tildrakizumab (MK 3222), TJ301, TNF-Kinoid®, tofacitinib, tralokinumab, TRK-170, upadacitinib, ustekinumab, UTTR1147A, V565, vatelizumab, VB-201, vedolizumab, and vidofludimus.

Non-limiting examples of additional therapeutic agents and/or regimens for treating irritable bowel syndrome include alosetron, bile acid sequesterants (e.g., cholestyramine, colestipol, colesevelam), chloride channel activators (e.g., lubiprostone), coated peppermint oil capsules, desipramine, dicyclomine, ebastine, eluxadoline, farnesoid X receptor agonist (e.g., obeticholic acid), fecal microbiota transplantation, fluoxetine, gabapentin, guanylate cyclase-C agonists (e.g., linaclotide, plecanatide), ibodutant, imipramine, JCM- 16021, loperamide, lubiprostone, nortriptyline, ondansetron, opioids, paroxetine, pinaverium, polyethylene glycol, pregabalin, probiotics, ramosetron, rifaximin, and tanpanor.

Non-limiting examples of additional therapeutic agents and/or regimens for treating scleroderma include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), corticosteroids (e.g, prednisone), immunomodulators (e.g., azathioprine, methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral®, Sandimmune®, Gengraf®), antithymocyte globulin, mycophenolate mofetil, intravenous immunoglobulin, rituximab, sirolimus, and alefacept), calcium channel blockers (e.g., nifedipine), alpha blockers, serotonin receptor antagonists, angiotensin II receptor inhibitors, statins, local nitrates, iloprost, phosphodiesterase 5 inhibitors (e.g., sildenafil), bosentan, tetracycline antibiotics, endothelin receptor antagonists, prostanoids, and tyrosine kinase inhibitors (e.g., imatinib, nilotinib and dasatinib).

Non-limiting examples of additional therapeutic agents and/or regimens for treating Crohn’s Disease (CD) include adalimumab, autologous CD34-selected peripheral blood stem cells transplant, 6-mercaptopurine, azathioprine, certolizumab pegol (Cimzia®), corticosteroids (e.g., prednisone), etrolizumab, E6011, fecal microbial transplantation, figlotinib, guselkumab, infliximab, IL-2, JAK inhibitors, matrix metalloproteinase 9 (MMP 9) inhibitors (e.g., GS-5745), MEDI2070, mesalamine, methotrexate, natalizumab, ozanimod, RHB-104, rifaximin, risankizumab, SHP647, sulfasalazine, thalidomide, upadacitinib, V565, and vedolizumab.

Non-limiting examples of additional therapeutic agents and/or regimens for treating UC include AbGn-168H, ABT-494, ABX464, apremilast, PF-00547659, PF-06687234, 6- mercaptopurine, adalimumab, azathioprine, bertilimumab, brazikumab (MEDI2070), cobitolimod, certolizumab pegol (Cimzia®), CP-690,550, corticosteroids (e.g., multimax budesonide, Methylprednisolone), cyclosporine, E6007, etrasimod, etrolizumab, fecal microbial transplantation, figlotinib, guselkumab, golimumab, IL-2, IMU-838, infliximab, matrix metalloproteinase 9 (MMP9) inhibitors (e.g., GS-5745), mesalamine, mesalamine, mirikizumab (LY3074828), RPC1063, risankizumab (BI 6555066), SHP647, sulfasalazine, TD-1473, TJ301, tildrakizumab (MK 3222), tofacitinib, tofacitinib, ustekinumab, UTTR1147A, and vedolizumab. Non-limiting examples of additional therapeutic agents and/or regimens for treating autoimmune colitis include corticosteroids (e.g., budesonide, prednisone, prednisolone, Beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, mesalamine, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.

Non-limiting examples of additional therapeutic agents and/or regimens for treating iatrogenic autoimmune colitis include corticosteroids (e.g., budesonide, prednisone, prednisolone, Beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.

Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis induced by one or more chemotherapeutics agents include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, mesalamine, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.

Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis induced by treatment with adoptive cell therapy include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.

Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis associated with one or more alloimmune diseases include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), sulfasalazine, and eicopentaenoic acid.

Non-limiting examples of additional therapeutic agents and/or regimens for treating radaiation enteritis include teduglutide, amifostine, angiotensin-converting enzyme (ACE) inhibitors (e.g., benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, and trandolapril), probiotics, selenium supplementation, statins (e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, and pitavastatin), sucralfate, and vitamin E. Non-limiting examples of additional therapeutic agents and/or regimens for treating collagenous colitis include 6-mercaptopurine, azathaioprine, bismuth subsalicate, Boswellia serrata extract, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), loperamide, mesalamine, methotrexate, probiotics, and sulfasalazine.

Non-limiting examples of additional therapeutic agents and/or regimens for treating lyphocytic colitis include 6-mercaptopurine, azathioprine, bismuth subsalicylate, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), loperamide, mesalamine, methotrexate, and sulfasalazine.

Non-limiting examples of additional therapeutic agents and/or regimens for treating microscopic colitis include 6-mercaptopurine, azathioprine, bismuth subsalicylate, Boswellia serrata extract, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), fecal microbial transplantation, loperamide, mesalamine, methotrexate, probiotics, and sulfasalazine.

Non-limiting examples of additional therapeutic agents and/or regimens for treating alloimmune disease include intrauterine platelet transfusions, intravenous immunoglobin, maternal steroids, abatacept, alemtuzumab, alphal -antitrypsin, AMG592, antithymocyte globulin, barcitinib, basiliximab, bortezomib, brentuximab, cannabidiol, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, glasdegib, ibrutinib, IL-2, infliximab, itacitinib, LBH589, maraviroc, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, pevonedistat, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.

Non-limiting examples of additional therapeutic agents and/or regimens for treating multiple sclerosis (MS) include alemtuzumab (Lemtrada®), ALKS 8700, amiloride, ATX- MS-1467, azathioprine, baclofen (Lioresal®), beta interferons (e.g., IFN-P-la, IFN-P-lb), cladribine, corticosteroids (e.g., methylprednisolone), daclizumab, dimethyl fumarate (Tecfidera®), fmgolimod (Gilenya®), fluoxetine, glatiramer acetate (Copaxone®), hydroxychloroquine, ibudilast, idebenone, laquinimod, lipoic acid, losartan, masitinib, MD1003 (biotin), mitoxantrone, montel ukast, natalizumab (Tysabri®), NeuroVax™, ocrelizumab, ofatumumab, pioglitazone, and RPC 1063. Non-limiting examples of additional therapeutic agents and/or regimens for treating graft-vs-host disease include abatacept, alemtuzumab, alphal -antitrypsin, AMG592, antithymocyte globulin, barcitinib, basiliximab, bortezomib, brentuximab, cannabidiol, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, glasdegib, ibrutinib, IL-2, imatinib, infliximab, itacitinib, LBH589, maraviroc, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, pevonedistat, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.

Non-limiting examples of additional therapeutic agents and/or regimens for treating acute graft-vs-host disease include alemtuzumab, alpha- 1 antitrypsin, antithymocyte globulin, basiliximab, brentuximab, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, ibrutinib, infliximab, itacitinib, LBH589, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, photopheresis, ruxolitinib, sirolimus, tacrolimus, and tocilizumab.

Non-limiting examples of additional therapeutic agents and/or regimens for treating chronic graft vs. host disease include abatacept, alemtuzumab, AMG592, antithymocyte globulin, basiliximab, bortezomib, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, denileukin diftitox, glasdegib, ibrutinib, IL-2, imatinib, infliximab, mycophenolate mofetil, pentostatin, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.

Non-limiting examples of additional therapeutic agents and/or regimens for treating celiac disease include AMG 714, AMY01, Aspergillus niger prolyl endoprotease, BL- 7010, CALY-002, GBR 830, Hu-Mik-Beta-1, IMGX003, KumaMax, Larazotide Acetate, Nexvan2®, pancrelipase, TIMP-GLIA, vedolizumab, and ZED1227.

Non-limiting examples of additional therapeutic agents and/or regimens for treating psoriasis include topical corticosteroids, topical crisaborole/AN2728, topical SNA-120, topical SAN021, topical tapinarof, topical tocafinib, topical IDP-118, topical M518101, topical calcipotriene and betamethasone dipropionate (e.g., MC2-01 cream and Taclonex®), topical P-3073, topical LEO 90100 (Enstilar®), topical betamethasone dipropriate (Sernivo®), halobetasol propionate (Ultravate®), vitamin D analogues (e.g., calcipotriene (Dovonex®) and calcitriol (Vertical®)), anthralin (e.g., Dritho-scalp® and Dritho-creme®), topical retinoids (e.g., tazarotene (e.g., Tazorac® and Avage®)), calcineurin inhibitors (e.g., tacrolimus (Prograf®) and pimecrolimus (Elidel®)), salicylic acid, coal tar, moisturizers, phototherapy (e.g., exposure to sunlight, UVB phototherapy, narrow band UVB phototherapy, Goeckerman therapy, psoralen plus ultraviolet A (PUVA) therapy, and excimer laser), retinoids (e.g., acitretin (Soriatane®)), methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), Apo805Kl, baricitinib, FP187, KD025, prurisol, VTP-43742, XP23829, ZPL-389, CF101 (piclidenoson), LAS41008, VPD-737 (serlopitant), upadacitinib (ABT-494), aprmilast, tofacitibin, cyclosporine (Neoral®, Sandimmune®, Gengraf®), biologies (e.g., etanercept (Enbrel®), entanercept-szzs (Elrezi®), infliximab (Remicade®), adalimumab (Humira®), adalimumab-adbm (Cyltezo®), ustekinumab (Stelara®), golimumab (Simponi®), apremilast (Otezla®), secukinumab (Cosentyx®), certolixumab pegol, secukinumab, tildrakizumab-asmn, infliximab-dyyb, abatacept, ixekizumab (Taltz®), ABP 710, BCD-057, BI695501, bimekizumab (UCB4940), CHS-1420, GP2017, guselkumab (CNTO 1959), HD203, M923, MSB11022, Mirikizumab (LY3074828), PF-06410293, PF-06438179, risankizumab (BI655066), SB2, SB4, SB5, siliq (brodalumab), namilumab (MT203, tildrakizumab (MK-3222), and ixekizumab (Taltz®)), thioguanine, and hydroxyurea (e.g., Droxia® and Hydrea®).

Non-limiting examples of additional therapeutic agents and/or regimens for treating cutaneous T-cell lymphoma include phototherapy (e.g., exposure to sunlight, UVB phototherapy, narrow band UVB phototherapy, Goeckerman therapy, psoralen plus ultraviolet A (PUVA) therapy, and excimer laser), extracorporeal photopheresis, radiation therapy (e.g., spot radiation and total skin body electron beam therapy), stem cell transplant, corticosteroids, imiquimod, bexarotene gel, topical bis-chloroethyl-nitrourea, mechlorethamine gel, vorinostat (Zolinza®), romidepsin (Istodax®), pralatrexate (Folotyn®) biologies (e.g., alemtuzumab (Campath®), brentuximab vedotin (SGN-35), mogamulizumab, and IPH4102).

Non-limiting examples of additional therapeutic agents and/or regimens for treating uveitis include corticosteroids (e.g., intravitreal triamcinolone acetonide injectable suspensions), antibiotics, antivirals (e.g., acyclovir), dexamethasone, immunomodulators (e.g., tacrolimus, leflunomide, cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral®, Sandimmune®, Gengraf®), chlorambucil, azathioprine, methotrexate, and mycophenolate mofetil), biologies (e.g., infliximab (Remicade®), adalimumab (Humira®), etanercept (Enbrel®), golimumab (Simponi®), certolizumab (Cimzia®), rituximab (Rituxan®), abatacept (Orencia®), basiliximab (Simulect®), anakinra (Kineret®), canakinumab (Haris®), gevokixumab (XOMA052), tocilizumab (Actemra®), alemtuzumab (Campath®), efalizumab (Raptiva®), LFG316, sirolimus (Santen®), abatacept, sarilumab (Kevzara®), and daclizumab (Zenapax®)), cytotoxic drugs, surgical implant (e.g., fluocinolone insert), and vitrectomy. on-limiting examples of additional therapeutic agents and/or regimens for treating mucositis include AG013, SGX942 (dusquetide), amifostine (Ethyol®), cryotherapy, cepacol lonzenges, capsaicin lozenges, mucoadhesives (e.g., MuGard®) oral diphenhydramine (e.g., Benadry® elixir), oral bioadherents (e.g., polyvinylpyrrolidone- sodium hyaluronate gel (Gelclair®)), oral lubricants (e.g., Oral Balance®), caphosol, chamomilla recutita mouthwash, edible grape plant exosome, antiseptic mouthwash (e.g., chlorhexidine gluconate (e.g., Peridex® or Periogard®), topical pain relievers (e.g., lidocaine, benzocaine, dyclonine hydrochloride, xylocaine (e.g., viscous xylocaine 2%), and Ulcerease® (0.6% phenol)), corticosteroids (e.g., prednisone), pain killers (e.g., ibuprofen, naproxen, acetaminophen, and opioids), GC4419, palifermin (keratinocyte growth factor; Kepivance®), ATL-104, clonidine lauriad, IZN-6N4, SGX942, rebamipide, nepidermin, soluble p-1,3/1,6 glucan, P276, LP-0004-09, CR-3294, ALD-518, IZN-6N4, quercetin, granules comprising vaccinium myrtillus extract, macleaya cordata alkaloids and echinacea angustifolia extract (e.g., SAMITAL®), and gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)). For example, nonlimiting examples of treatments for oral mucositis include AG013, amifostine (Ethyol®), cryotherapy, cepacol lonzenges, mucoadhesives (e.g., MuGard®) oral diphenhydramine (e.g., Benadry® elixir), oral bioadherents (e.g., polyvinylpyrrolidone-sodium hyaluronate gel (Gelclair®)), oral lubricants (e.g., Oral Balance®), caphosol, chamomilla recutita mouthwash, edible grape plant exosome, antiseptic mouthwash (e.g., chlorhexidine gluconate (e.g., Peridex® or Peri ogard®), topical pain relievers (e.g., lidocaine, benzocaine, dyclonine hydrochloride, xylocaine (e.g., viscous xylocaine 2%), and Ulcerease® (0.6% phenol)), corticosteroids (e.g., prednisone), pain killers (e.g., ibuprofen, naproxen, acetaminophen, and opioids), GC4419, palifermin (keratinocyte growth factor; Kepivance®), ATL-104, clonidine lauriad, IZN-6N4, SGX942, rebamipide, nepidermin, soluble p-1,3/1,6 glucan, P276, LP-0004-09, CR-3294, ALD-518, IZN-6N4, quercetin, and gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)). As another example, non-limiting examples of treatments for esophageal mucositis include xylocaine (e.g., gel viscous Xylocaine 2%). As another example, treatments for intestinal mucositis, treatments to modify intestinal mucositis, and treatments for intestinal mucositis signs and symptoms include gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)).

In certain embodiments, the second therapeutic agent or regimen is administered to the subject prior to contacting with or administering the chemical entity (e.g., about one hour prior, or about 6 hours prior, or about 12 hours prior, or about 24 hours prior, or about 48 hours prior, or about 1 week prior, or about 1 month prior).

In other embodiments, the second therapeutic agent or regimen is administered to the subject at about the same time as contacting with or administering the chemical entity. By way of example, the second therapeutic agent or regimen and the chemical entity are provided to the subject simultaneously in the same dosage form. As another example, the second therapeutic agent or regimen and the chemical entity are provided to the subject concurrently in separate dosage forms.

In still other embodiments, the second therapeutic agent or regimen is administered to the subject after contacting with or administering the chemical entity (e.g., about one hour after, or about 6 hours after, or about 12 hours after, or about 24 hours after, or about 48 hours after, or about 1 week after, or about 1 month after).

Patient Selection In some embodiments, the methods described herein further include the step of identifying a subject (e.g., a patient) in need of such treatment (e.g., by way of biopsy, endoscopy, or other conventional method known in the art). In certain embodiments, the STING protein can serve as a biomarker for certain types of cancer, e.g., colon cancer and prostate cancer. In other embodiments, identifying a subject can include assaying the patient’s tumor microenvironment for the absence of T-cells and/or presence of exhausted T-cells, e.g., patients having one or more cold tumors. Such patients can include those that are resistant to treatment with checkpoint inhibitors. In certain embodiments, such patients can be treated with a chemical entity herein, e.g., to recruit T-cells into the tumor, and in some cases, further treated with one or more checkpoint inhibitors, e.g., once the T-cells become exhausted.

In some embodiments, the chemical entities, methods, and compositions described herein can be administered to certain treatment-resistant patient populations (e.g., patients resistant to checkpoint inhibitors; e.g., patients having one or more cold tumors, e.g., tumors lacking T-cells or exhausted T-cells).

Compound Preparation

As can be appreciated by the skilled artisan, methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and RGM. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. The starting materials used in preparing the compounds of the invention are known, made by known methods, or are commercially available. The skilled artisan will also recognize that conditions and reagents described herein that can be interchanged with alternative art-recognized equivalents. For example, in many reactions, triethylamine can be interchanged with other bases, such as non- nucleophilic bases (e.g. diisopropylamine, l,8-diazabicycloundec-7-ene, 2,6-di-tert- butylpyridine, or tetrabutylphosphazene).

The skilled artisan will recognize a variety of analytical methods that can be used to characterize the compounds described herein, including, for example, T H NMR, heteronuclear NMR, mass spectrometry, liquid chromatography, and infrared spectroscopy. The foregoing list is a subset of characterization methods available to a skilled artisan and is not intended to be limiting.

To further illustrate the foregoing, the following non-limiting, exemplary synthetic schemes are included. Variations of these examples within the scope of the claims are within the purview of one skilled in the art and are considered to fall within the scope of the invention as described, and claimed herein. The reader will recognize that the skilled artisan, provided with the present disclosure, and skill in the art is able to prepare and use the invention without exhaustive examples.

Examples

Abbreviation of chemical terms

ADDP = l,T-(azodicarbonyl)-dipiperidine

ACN = acetonitrile

BOC2O = di -tert-butyl pyrocarb ornate

DCM = dichloromethane

DMF = VMdi methyl form am ide

DMSO = dimethyl sulfoxide

Dppf = bis(diphenylphosphino)ferrocene

HPLC = high performance liquid chromatography

LC-MS = liquid chromatography - mass spectrometry

NMR = nuclear magnetic resonance

RT = retention time TBUP = Tri-n-butylphosphine

TEA = trimethylamine

TFA = trifluoroacetic acid

THF = tetrahydrofuran

FA = Formic acid

Speedvac = Savant SC250EXP SpeedVac ConcentratorDMSO = Dimethyl SulfoxideNa2SO4 = Sodium sulfate

Materials and Methods

The LC-MS was recorded using one of the following methods.

For examples 1-2:

LCMS Method A: Kinetex EVO C18 100A, 30*3mm, 0.5 pL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.30 min, 95% MPB to 10% in 0.10 min.

LCMS Method B: HALOC18, 30*3mm, 0.5 pL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm LTV detection. Mobile Phase A (MPA): Water/0.05% TFA and Mobile Phase B (MPB): Acetonitrile/0.05% TFA. Elution 5% MPB to 100% in 1.20 min, hold at 100% MPB for 0.60 min, 100% MPB to 5% in 0.02 min, then equilibration to 5% MPB for 0.18 min.

NMR was recorded on BRUKER NMR 300.03 Mz, DUL-C-H, ULTRASHIELDTM 300, AVANCE II 300 B-ACSTM 120 or BRUKER NMR 400.13 Mz, BBFO, ULTRASHIELDTM 400, AVANCE III 400, B-ACSTM 120. For examples 3-24

Method A

Instrument: Agilent LCMS system equipped with DAD and ELSD detector

Ion mode: Positive

Column: Waters X-Bridge C18, 50*2.1 mm*5 pm or equivalent

Mobile Phase: A: H2O (0.04% TFA); B: CH3CN (0.02% TFA)

Gradient: 4.5 min gradient method, actual method would depend on clogP of compound.

Flow Rate: 0.6 mL/min or 0.8 mL/min

Column Temp: 40 °C or 50 °C

UV: 220 nm

Method B

Instrument: Agilent LCMS system equipped with DAD and ELSD detector

Ion mode: Positive

Column: Waters X-Bridge ShieldRP18, 50*2.1 mm*5 pm or equivalent

Mobile PhaseA: H2O (0.05% NH3 H2O) or 10 mM ammonia bicarbonate; B: CH3CN

Gradient: 4.5 min gradient method; actual method would depend on the clogP of the compound.

Flow Rate: 0.6 mL/min or 0.8 mL/min

Column Temp: 40 °C

UV: 220 nm

Prep. HPLC condition

Instrument:

1. GILSON 281 and Shimadzu LCMS 2010A

2. GILSON 215 and Shimadzu LC-20AP

3. GILSON 215

Mobile phase:

A: NH4OH/H2O = 0.05% v/v; B: ACN

A: FA/H2O = 0.225% v/v; B: ACN

Column Xtimate C18 150*25mm*5pm

Flow rate: 25 mL/min or 30 mL/min

Monitor wavelength: 220&254 nm

Gradient: actual method would depend on clog P of compound

Detector: MS Trigger or UV

Preparative examples

Scheme for the preparation of Key Intermediates: Schemes below illustrate the preparation of key intermediates.

Scheme 1: Synthesis of intermediate 1 (5-(2-(4-(trifluoromethyl)phenoxy)ethyl)~ lH-indol-3-amine TFA salt)

Step 1: tert-butyl (5-vinyl-lH-indol-3-yl)carbamate

Ze/V-Butyl M(5-bromo- IT/-indol-3-yl)carbamate (4.0 g, 12.8 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (20 mL) and water (4 ml), then CS2CO3 (8.3 g, 25.7 mmol, 2.0 equiv.), Pd(dppf)C12 (0.9 g, 1.2 mmol, 0.1 equiv.) and 2-ethenyl-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane (2.7 g, 19.2 mmol, 1.2 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was heated to 90 °C for 3 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1 : 1) to give tert-butyl (5-vinyl- U/-indol-3-yl)carbamate (2.0 g) as a brown solid. LCMS Method A: [M+H] + = 259.

Step 2: tert-butyl (5-(2-hydroxyethyl)-LH-indol-3-yl)carbamate tert-Butyl (5-vinyl-U/-indol-3-yl)carbamate (2.0 g, 7.7 mmol, 1.0 equiv.) was dissolved in THF (10 mL) and cooled to 0 °C, then BH3-THF (IM, 46.4 mL, 46.4 mmol, 6.0 equiv.) was added dropwise. After stirred for 2 hours at ambient temperature, a solution of aqueous NaOH (1 M, 14 mL, 14.0 mmol, 2.0 equiv.) was added. This was followed by the addition of H2O2 (30% wt. in water, 2.6 mL, 23.2 mmol, 3.0 equiv.), maintaining the reaction mixture at 0 °C. The reaction mixture was stirred for additional 1 hour at 0 °C, then quenched by the addition of saturated aqueous NH4Q. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1 : 1) to give tert-butyl (5-(2-hydroxyethyl)-U/-indol-3-yl)carbamate (605.0 mg) as a brown solid. LCMS Method A: [M+H] + = 277.

Step 3: tert-butyl /V-(5-[2-[4-(trifluoromethyl)phenoxy]ethyl]-LH-indol-3- yl)carbamate tert-Butyl A-[5-(2-hydroxyethyl)-17/-indol-3-yl]carbamate (338.0 mg, 1.2 mmol, 1.0 equiv.) and 4-(trifluoromethyl)phenol (198.2 mg, 1.2 mmol, 1.0 equiv.) were dissolved in THF (10 mL), then ADDP (612.4 mg, 2.4 mmol, 2.0 equiv.) and TBUP (494.9 mg, 2.4 mmol, 2.0 equiv.) were added. The reaction mixture was heated to 70 °C for 5 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1 : 1) to give tert-butyl 7V-(5-[2-[4- (trifluoromethyl)phenoxy]ethyl]-U/-indol-3-yl)carbamate (260.0 mg) as a brown solid.

LCMS Method A: [M+H] + = 421.

Step 4: 5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-LH-indol-3-amine TFA salt tert-Butyl A-(5-{2-[4-(trifluoromethyl)phenoxy]ethyl}-l#-indol-3-yl)car bamate (260.0 mg, 0.6 mmol, 1.0 equiv.) was dissolved in DCM (2 mL) and TFA (2 mL). The reaction mixture was stirred for 30 min at ambient temperature, then concentrated under vacuum to give 5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-lJ/-indol-3-amine TFA salt (350.0 mg) as a yellow solid. LCMS Method A: [M+H] + = 321.

Example 1 : 1 -(let rahydrofiiran-2-yl)-\-(5-(2-(4-(t rill uoromet hyl )phenoxy) ethyl)- lH-indol-3-yl)methanesulfonamide [Compound 125]

5-[2-[4-(Trifluoromethyl)phenoxy]ethyl]-lH-indol-3-amine trifluoroacetic acid salt (260.0 mg, 0.4 mmol, 1.0 equiv.) and TEA (0.2 mL, 1.4 mmol, 3.0 equiv.) were dissolved in DCM (5 mL), then oxolan-2-ylmethanesulfonyl chloride (89.9 mg, 0.4 mmol, 1.0 equiv.) was added. The reaction mixture was stirred for 2 hours at ambient temperature and then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1 : 1) to give initial material that was further purified by Prep-HPLC with the following conditions: Column: Sunfire prep Cl 8 column, 30*150, 5pm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 45 B to 75 B in 7 min; 254/210 nm; RT1 : 6.42 min. This gave l-(tetrahydrofuran-2-yl)-7V-(5-(2-(4-(trifluoromethyl)phenox y)ethyl)-lJ/-indol-3- yl)methanesulfonamide (12.9 mg) as a white solid. LCMS Method B: [M+H] + = 469. T H NMR (400 MHz, DMSO4): 5 11.01 (d, J= 2.4 Hz, 1H), 9.10 (s, 1H), 7.64 (d, J= 8.8 Hz, 2H), 7.57 (s, 1H), 7.31 (d, J = 8.4 Hz, 1H), 7.26 (d, J= 2.4 Hz, 1H), 7.14-7.09 (m, 3H), 4.30-4.21 (m, 3H), 3.75-3.71 (m, 1H), 3.64-3.62 (m, 1H), 3.18-3.11 (m, 4H), 2.05-2.01 (m, 1H), 1.82-1.78 (m, 2H), 1.64-1.59 (m, 1H).

The analogs prepared in Table below were prepared using the same method described for Example 1.

Example 3: Synthesis of N-(5-{2-[4-(trifluoromethyl)phenyl]ethoxy}-lH-indol-3- yl)ethane-l-sulfonamide tert-butyl 3-{[(tert-butoxy)carbonyl]amino}-5-{2-[4-(trifluoromethyl)ph enyl]ethoxy}- IH-indole-l -carboxylate (83.2 mg, 0.16 mmol, 1.0 equiv.) was dissolved in DCM (2 mL), then TFA (500 pl) was added to the solution. The mixture was heated at 30 °C for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and ethanesulfonyl chloride (41.0 mg, 0.32 mmol, 2.0 equiv.) were dissolved in DCM (2 mL), then TEA (116 pl , 0.8 mmol, 5.0 equiv.) was added. The mixture was heated at 30 °C for 16 hours. The reaction mixture was concentrated by Speedvac to give a residue that was purified by prep HPLC to give N-(5-{2-[4-(trifluoromethyl)phenyl]ethoxy}-lH- indol-3-yl)ethane-l -sulfonamide (14.2 mg, 0.034 mmol) as a powder. MS-ESI, 413.2 [M+H^JH NMR (400 MHz, DMSO-d6) 8 ppm 10.89 (br s, 1 H), 9.01 (s, 1 H), 7.68 (br d, J=8.1 Hz, 2 H), 7.58 (br d, J=8.0 Hz, 2 H), 7.28-7.12 (m, 3 H), 6.73 (dd, J=8.7, 2.4 Hz, 1 H), 4.19 (t, J=6.6 Hz, 2 H), 3.22-3.09 (m, 2 H), 2.92 (q, ./=7,4 Hz, 2 H), 1.22 (t, J=7.3 Hz, 3 H).

Example 4: Synthesis of N-(5-{2-[4-(trifluoromethyl)phenoxy]ethyl}-lH-indol-3- yl)ethane-l-sulfonamide

/c/7-butyl (5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-lH-indol-3-yl)carba mate (67.2 mg, 0.16 mmol, 1.0 equiv.) was dissolved in DCM (2 mL), then TFA (500 pl) was added to the solution. The reaction mixture was heated at 30 °C for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and ethanesulfonyl chloride (41.0 mg, 0.32 mmol, 2.0 equiv.) were dissolved in DCM (2 mL), then TEA (116 pl, 0.8 mmol, 5.0 equiv.) was added. The mixture was heated at 30 °C for 16 hours. The reaction mixture was concentrated by Speedvac to give a residue that was purified by prep HPLC to give N-(5-{2-[4-(trifluoromethyl)phenoxy]ethyl}-lH-indol-3-yl)eth ane-l- sulfonamide (33.3 mg, 0.081 mmol) as a powder. MS-ESI, 413.2 fM+H + ].'H NMR (400 MHz, DMSO-tA) 6 ppm 11.03-10.95 (m, 1 H), 9.07 (s, 1 H), 7.63 (d, J=8.8 Hz, 2 H), 7.55 (s, 1 H), 7.29 (d, J=8.3 Hz, 1 H), 7.24 (d, ./=2,5 Hz, 1 H), 7.14-7.06 (m, 3 H), 4.27 (t, J=6.8 Hz, 2 H), 3.17-3.07 (m, 2 H), 2.93 (q, J=7.4 Hz, 2 H), 1.23 (t, J=7.4 Hz, 3 H).

The compounds in Table below were prepared using the above procedures of example 3 and 4 with the approproate starting material.

Biological Assays

STING pathway activation by the compounds described herein was measured using THP 1 -Dual™ cells (KO-IFNAR2) .

THPl-Dual™ KO-IFNAR2 Cells (obtained from invivogen) were maintained in RPMI, 10% FCS, 5 ml P/S, 2mM L-glut, lOmM Hepes, and 1 mM sodium pyruvate. Compounds were spotted in empty 384 well tissue culture plates (Greiner 781182) by Echo for a final concentration of 0.0017 - 100 pM. Cells were plated into the TC plates at 40 pL per well, 2* 10E6 cells/mL. For activation with STING ligand, 2'3'cGAMP (MW 718.38, obtained from Invivogen), was prepared in Optimem media.

The following solutions were prepared for each 1x384 plate: o Solution A: 2 mL Optimem with one of the following stimuli:

■ 150 pM stock o Solution B: 2 mL Optimem with 60 pL Lipofectamine 2000 Incubate 5 min at RT

2 mL of solution A and 2 ml Solution B was mixed and incubated for 20 min at room temperature (RT). 20 pL of transfection solution (A+B) was added on top of the plated cells, with a final 2’3’cGAMP concentration of 15 pM. The plates were then centrifuged immediately at 340 g for 1 minute, after which they were incubated at 37 °C, 5% CC>2, >98% humidity for 24h. Luciferase reporter activity was then measured. ECso values were calculated by using standard methods known in the art.

Luciferase reporter assay: 10 pL of supernatant from the assay was transferred to white 384-plate with flat bottom and squared wells. One pouch of QUANTI-Luc™ Plus was dissolved in 25 mL of water. 100 pL of QLC Stabilizer per 25 mL of QUANTI- Luc™ Plus solution was added. 50 pL of QUANTI-Luc™ Plus/QLC solution per well was then added. Luminescence was measured on a Platereader (e.g., Spectramax I3X (Molecular Devices GF3637001)).

Luciferase reporter activity was then measured. ECso values were calculated by using standard methods known in the art.

Table BA shows the activity of compounds in STING reporter assay: <0.008 pM = “++++++”; >0.008 and <0.04 pM = “+++++”; >0.04 and <0.2 pM = “++++”; >0.2 and Table BA Numbered Clauses

The compounds, compositions, methods, and other subject matter described herein are further described in the following numbered clauses:

1. A compound of Formula (I):

Formula I or a pharmaceutically acceptable salt thereof or a tautomer thereof, wherein:

L A is - (L 1 ) a i-(L 2 )a2-(L 3 )a3-(L 4 )a4-(L 5 )a5-*, wherein * represents the point of attachment to Q 1 ; al, a2, a3, a4, and a5 are each independently 0 or 1, provided that al + a2 + a3 + a4 + a5 > 1, and each of L 1 , L 3 , and L 5 is independently selected from the group consisting of: -O-, -N(H)-, -N(R d )-, S(0)o-2, and -C(=O)-; provided that when one or both of a2 and a4 is 0, then the combinations of L 1 , L 3 , and L 5 cannot form 0-0 , N-O, N-N, O-S, S-S, or N-S(0)o bonds, and each of L 2 and L 4 is independently selected from the group consisting of:

• straight-chain Ci-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 R b ;

• C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 R c ; and

• heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 R c ; Q 1 is -R g ;

Y 1 , Y 2 , and Y 3 are each independently selected from the group consisting of CR 1 , C(=O), N, and NR 2 ;

X 1 is selected from the group consisting of O, S, N, NR 2 , and CR 1 ;

X 2 is selected from the group consisting of O, S, N, NR 4 , and CR 5 ; each == is independently a single bond or a double bond, provided that the fivemembered ring comprising X 1 and X 2 is heteroaryl, and that the six-membered ring comprising Y 1 , Y 2 , and Y 3 is aryl or heteroaryl; further provided that L A cannot include a cyclic group directly attached to the 6- membered ring containing Y 1 , Y 2 , and Y 3 ; each occurrence of R 1 and R 5 is independently selected from the group consisting of: H; R c ; R g ; and -(L g ) bg -R g ; each occurrence of R 2 and R 4 is independently selected from the group consisting of: H; R d ; R g ; and -(L g ) bg -R g ;

R 6 is selected from the group consisting of: H; R d ; and R g ;

W is selected from the group consisting of:

• Ci-io alkyl, C2-10 alkenyl, or C2-10 alkynyl, each of which is optionally substituted with 1-6 R a2 or R g ;

• C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• heteroaryl, heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c , provided that when W is heteroaryl, heterocyclyl or heterocycloalkenyl, it is attached to the C(=O)NR 6 group via a ring carbon atom; each occurrence of R a and R a2 is independently selected from the group consisting of: -OH; -halo; -NR e R f ; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=O)O(Ci-4 alkyl); -C(=O)(Ci-4 alkyl); -C(=O)OH; -CONR’R”; -S(O)I- 2 NR’R”; -S(O)i- 2 (Ci-4 alkyl); and cyano; each occurrence of R b and R c is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with 1-6 independently selected R a ; C 2 -6 alkenyl; C 2 -6 alkynyl; Ci-4 alkoxy; Ci-4 haloalkoxy; -S(O)i- 2 (Ci-4 alkyl); - S(O)(=NH)(CI- 4 alkyl); -NR e R f ; -OH; -S(O)I- 2 NR’R”; -Ci-4 thioalkoxy; -NO 2 ; - C(=0)(Ci-io alkyl); -C(=O)O(Ci-4 alkyl); -C(=O)OH; -C(=O)NR’R”; and -SF 5 ; each occurrence of R d is independently selected from the group consisting of: Ci-6 alkyl optionally substituted with 1-3 independently selected R a ; -C(O)(Ci-4 alkyl); - C(O)O(Ci-4 alkyl); -CONR’R”; -S(O)I- 2 NR’R”; - S(O)i- 2 (Ci-4 alkyl); -OH; and Ci-4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; Ci-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, halo, Ci-4 alkoxy, and Ci-4 haloalkoxy; - C(O)(Ci- 4 alkyl); -C(O)O(Ci-4 alkyl); -CONR’R”; -S(O)i- 2 NR’R”; -S(O)i- 2 (Ci-4 alkyl); - OH; and Ci-4 alkoxy; each occurrence of R g is independently selected from the group consisting of:

• C3-12 cycloalkyl or Cs-i 2 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ;

• heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ; • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ; and

• Ce-io aryl optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, R c , and R h ; each occurrence of R h is independently selected from the group consisting of:

• C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 R‘;

• heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 R‘;

• heteroaryl of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R‘; and

• Ce-io aryl optionally substituted with 1-4 R‘; each occurrence of R‘ is independently selected from the group consisting of: C1-6 alkyl; Ci-4 haloalkyl; Ci-4 alkoxy; Ci-4 haloalkoxy; and halo; each occurrence of L g is independently selected from the group consisting of: -O-, -NH-, -NR d , -S(0)o-2, C(O), and C1-3 alkylene optionally substituted with 1-3 R a ; each occurrence of bg is independently 1, 2, or 3; and each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; and Ci-4 alkyl.

2. The compound of clause 1, wherein a2 is 1. 3. The compound of clauses 1 or 2, wherein L 2 is straight-chain Ci-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 R b .

4. The compound of any one of clauses 1-3, wherein L 2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 R b .

5. The compound of any one of clauses 1-4, wherein L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

6. The compound of any one of clauses 1-5, wherein L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-.

7. The compound of any one of clauses 1-6, wherein L 2 is -CH2-.

8. The compound of any one of clauses 1-4, wherein L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

9. The compound of any one of clauses 1-4 or 8, wherein L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b .

10. The compound of any one of clauses 1-4 or 8-9, wherein L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

11. The compound of any one of clauses 1-4 or 8-10, wherein L 2 is -CH2CH2-

12. The compound of any one of clauses 1-4 or 8, wherein L 2 is straight-chain C3 alkylene which is optionally substituted with 1-3 R b . 13. The compound of any one of clauses 1-4, 8, or 12, wherein L 2 is selected from the group consisting of: , and

R b , wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

14. The compound of any one of clauses 1-3, wherein L 2 is straight-chain C2-6 alkenylene, which is optionally substituted with 1-6 R b .

15. The compound of any one of clauses 1-3 or 14, wherein L 2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 R b .

16. The compound of any one of clauses 1-3 or 14-15, wherein L 2 is selected from the group consisting of: , wherein the asterisk represents the point of attachment to -(L 3 )a3-.

17. The compound of clauses 1 or 2, wherein L 2 is selected from the group consisting of:

• C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 R c ; and

• heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 R c .

18. The compound of any one of clauses 1-2 or 17, wherein L 2 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c . 19. The compound of any one of clauses 1-2 or 17-18, wherein L 2 is: which is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0, 1, or 2; Q 2 is CH, CR C , or N; and the asterisk represents the point of attachment to -(L 3 )a3-.

20. The compound of clause 19, wherein Q 2 is CH.

21. The compound of clauses 19 or 20, wherein nl and n2 are each 0.

22. The compound of any one of clauses 1-2 or 17-21, wherein L 2 is , wherein the asterisk represents the point of attachment to -( L 3 ) a j- .

23. The compound of clause 1, wherein a2 is 0.

24. The compound of any one of clauses 1-23, wherein al is 1.

25. The compound of any one of clauses 1-24, wherein L 1 is selected from the group consisting of: -O-, -N(H)-, -N(R d )-, and -S-.

26. The compound of any one of clauses 1-25, wherein L 1 is -O-.

27. The compound of any one of clauses 1-23, wherein al is 0.

28. The compound of any one of clauses 1-27, wherein a3 is 1.

29. The compound of any one of clauses 1-28, wherein L 3 is selected from the group consisting of: -O-, -N(H)-, -N(R d )-, and -S-

30. The compound of any one of clauses 1-29, wherein L 3 is -O-. 31. The compound of any one of clauses 1-29, wherein L 3 is -N(H)- or -N(R d )- , optionally -N(H)-.

32. The compound of any one of clauses 1-27, wherein a3 is 0.

33. The compound of any one of clauses 1-32, wherein a4 is 1.

34. The compound of any one of clauses 1-33, wherein L 4 is straight-chain Ci-

3 alkylene, which is optionally substituted with 1-3 R b .

35. The compound of any one of clauses 1-34, wherein L 4 is -CH2-.

36. The compound of any one of clauses 1-33, wherein L 4 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

37. The compound of any one of clauses 1-33 or 36, wherein L 4 is: which is optionally substituted with 1-2 R c , wherein n3 and n4 are independently 0, 1, or 2; Q 3 is CH, CR C , or N; and the asterisk represents the point of attachment to

38. The compound of clause 37, wherein n3 and n4 are each 1.

39. The compound of clauses 37 or 38, wherein Q 3 is N.

40. The compound of any one of clauses 1-33 or 36-39, wherein L 4 is , wherein the asterisk represents the point of attachment to -(L 5 )as-. 41. The compound of any one of clauses 1-32, wherein a4 is 0.

42. The compound of any one of clauses 1-41, wherein a5 is 0.

43. The compound of clause 1, wherein one of al, a3, and a5 is 1, and the other two are 0.

44. The compound of clauses 1 or 43, wherein one of a2 and a4 is 1, and the other is 0 or 1.

45. The compound of any one of clauses 1 or 43-44, wherein al and a2 are each 1.

46. The compound of any one of clauses 1 or 43-45, wherein: al and a2 are each 1;

L 1 is -O-, -N(H)-, or -N(R d )-;

L 2 is selected from the group consisting of:

• straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b ;

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

47. The compound of any one of clauses 1 or 43-46, wherein: al and a2 are each 1;

L 1 is -O-; and

L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

48. The compound of any one of clauses 1 or 43-47, wherein: al and a2 are each 1;

L 1 is -O-; and L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-.

49. The compound of any one of clauses 1 or 43-47, wherein: al and a2 are each 1;

L 1 is -O-; and

L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

50. The compound of clause 49, wherein L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b .

51. The compound of clauses 49 or 50, wherein L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

52. The compound of any one of clauses 49-51, wherein L 2 is -CH2CH2-.

53. The compound of any one of clauses 1 or 43-46, wherein: al and a2 are each 1;

L 1 is -O-;

L 2 is selected from the group consisting of:

C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

54. The compound of clause 53, wherein L 2 is: which is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0, 1, or 2; Q 2 is CH, CR C , or N; and the asterisk represents the point of attachment to -(L 3 )a3-. 55. The compound of clause 54, wherein nl and n2 are independently 0 or 1, optionally 0; and Q 2 is CH; optionally wherein nl and n2 are 0 and Q 2 is CH; optionally wherein L 2 is cyclobutane-diyl optionally substituted with 1-2 R c ; ; optionally wherein L 2 is cyclobutane- 1,3 -diyl optionally substituted with 1-2 R c ; ; optionslly wherein L 2 is unsubstituted cyclobutane-diyl; optionally wherein L 2 is unsubstituted cyclobutane- 1,3- diyl.

56. The compound of any one of clauses 43-55, wherein a3, a4, and a5 are each

0, optionally wherein L A is -O-CH2CH2-*, or (such as wherein * represents the point of attachment to Q 1 .

57. The compound of any one of clauses 43-55, wherein a3 and a5 are 0; and a4 is 1.

58. The compound of clause 57, wherein L 4 is selected from the group consisting of:

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

59. The compound of clauses 57 or 58, wherein L 4 is: optionally substituted with 1-2 R c , wherein n3 and n4 are independently 0, 1, or 2; Q 3 is CH, CR C , or N; and the asterisk represents the point of attachment to -(L 5 )as-.

60. The compound of clause 59, wherein n3 and n4 are independently 0 or 1; and Q 3 is N. 61. The compound of any one of clauses 1 or 43-44, wherein: al is 0; and a2 is

1.

62. The compound of any one of clauses 1, 43-44, or 61, wherein al is 0; a2 is 1; and L 2 is straight-chain Ci-6 alkylene, which is optionally substituted with 1-6 R b .

63. The compound of clauses 61 or 62, wherein L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

64. The compound of any one of clauses 61-63, wherein L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-.

65. The compound of any one of clauses 61-64, wherein L 2 is -CH2-.

66. The compound of any one of clauses 61-63, wherein L 2 is straight-chain C2- 3 alkylene which is optionally substituted with 1-3 R b .

67. The compound of any one of clauses 61-63 or 66, wherein L 2 is straightchain C2 alkylene, which is optionally substituted with 1-3 R b .

68. The compound of any one of clauses 61-63 or 66-67, wherein L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

69. The compound of any one of clauses 61-63 or 66-68, wherein L 2 is - CH2CH2-.

70. The compound of any one of clauses 61-63 or 66, wherein L 2 is straightchain C3 alkylene, which is optionally substituted with 1-3 R b . 71. The compound of any one of clauses 61-63, 66, or 70, wherein L 2 is selected from the group consisting of: , and

R b wherein the asterisk represents point of attachment to -(L 3 ) a 3-.

72. The compound of any one of clauses 61-71, wherein a3 is 0; a4 is 0; and a5 is 0.

73. The compound of any one of clauses 61-71, wherein a3 is 1.

74. The compound of clause 73, wherein a3 is 1; and L 3 is selected from the group consisting of: is -O-, -N(H)-, and -N(R d )-.

75. The compound of clauses 73 or 74, wherein a3 is 1; and L 3 is -O-.

76. The compound of any one of clauses 61-71 or 73-74, wherein a3 is 1; and L 3 is -N(H)- or -N(R d )-, optionally -N(H)-.

77. The compounds of any one of clauses 61-71 or 73-76, wherein a4 is 1; and

L 4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

78. The compound of any one of clauses 61-71 or 73-77, wherein a4 is 1; and L 4 is -CH2-.

79. The compound of any one of clauses 61-71 or 73-77, wherein a4 is 0; and a5 is 0, optionally wherein L A is -CH2CH2-O-*, wherein * represents to point of attachment to Q 1 .

80. The compound of clause 1, wherein al is 0; a2 is 1; L 2 is straight-chain C2- 4 alkenylene, which is optionally substituted with 1-3 R b .

I l l 81. The compound of clause 80, wherein L 2 is selected from the group consisting of: wherein the asterisk represents the point of attachment to -(L 3 )a3-.

82. The compound of clauses 80 or 81, wherein a3 is 0; a4 is 0; and a5 is 0.

83. The compound of any one of clauses 1-82, wherein Q 1 is selected from the group consisting of:

• heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-4 R c ’ ; and

• Ce-io aryl optionally substituted with 1-4 R c ’.

84. The compound of any one of clauses 1-82, wherein Q 1 is selected from the group consisting of:

• heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’.

85. The compound of any one of clauses 1-82, wherein Q 1 is selected from the group consisting of:

• heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’.

86. The compound of any one of clauses 1-85, wherein Q 1 is phenyl optionally substituted with 1-3 R c ’. 87. The compound of any one of clauses 1-86, wherein Q 1 is selected from the group consisting of:

88. The compound of any one of clauses 1-85, wherein Q 1 is heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 R c ’.

89. The compound of any one of clauses 1-85 or 88, wherein Q 1 is pyridyl, which is optionally substituted with 1-3 R c ’.

90. The compound of any one of clauses 1-85 or 88-89, wherein Q 1 is selected from the group consisting of:

91. The compound of any one of clauses 1-82, wherein Q 1 is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’.

92. The compound of any one of clauses 1-82 or 91, wherein Q 1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’.

93. The compound of any one of clauses 1-82 or 91-92, wherein Q 1 is heterocyclyl of 4-8 ring atoms, wherein 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, provided that one ring atom is N(R d ), and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’.

94. The compound of any one of clauses 1-82 or 91-93, wherein Q 1 is wherein ml and m2 are each independently 0, 1, or 2; and wherein Q 1 is optionally substituted with 1-2 R c ’.

95. The compound of any one of clauses 1-82 or 91-94, wherein Q 1 is

96. The compound of any one of clauses 1-82 or 91-94, wherein Q 1 is

97. The compound any one of clauses 91-96, wherein each R d present in Q 1 is independently selected from the group consisting of: -C(O)O(Ci-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected R a .

98. The compound of any one of clauses 91-97, wherein each R d present in Q 1 is C1-6 alkyl optionally substituted with 1-3 independently selected halo.

99. The compound of any one of clauses 91-98, wherein each R d present in Q 1 i. Ci-4 alkyl substituted with 1-3 -F; ii. C2-3 alkyl substituted with 1-3 -F; or iii. -CH2CF3. 100. The compound of any one of clauses 83-99, wherein each R c present in Q 1 is independently selected from the group consisting of: halo; cyano; Ci-4 alkoxy; Ci-4 haloalkoxy; and Ci-io alkyl which is optionally substituted with 1-6 independently selected R a

101. The compound of any one of clauses 83-100, wherein each R c present in Q 1 is independently selected from the group consisting of: halo; cyano; Ci-4 alkoxy; Ci-4 haloalkoxy; and Ci-6 alkyl which is optionally substituted with 1-6 independently selected halo.

102. The compound of any one of clauses 83-101, wherein each R c present in Q 1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo.

103. The compound of any one of clauses 83-102, wherein each R c present in Q 1 is: i. C1-3 alkyl which is optionally substituted with 1-6 -F; or ii. CF3.

104. The compound of any one of clauses 83-102, wherein each R c present in Q 1 is an independently selected halo, optionally -F or -Cl.

105. The compound of any one of clauses 1-104, wherein Y 1 is CR 1 .

106. The compound of any one of clauses 1-105, wherein Y 2 is CR 1 .

107. The compound of any one of clauses 1-106, wherein Y 3 is CR 1 .

108. The compound of any one of clauses 1-107, wherein each occurrence of R 1 is independently H or R c . 109. The compound of any one of clauses 1-108, wherein each occurrence of R 1 is H.

110. The compound of any one of clauses 1-108, wherein 1-2 occurrence of R 1 is R c ; and each remaining occurrence of R 1 is H.

111. The compound of any one of clauses 1-108 or 110, wherein one occurrence of R 1 is halo, optionally -F or -Cl; and each remaining occurrence of R 1 is H.

112. The compound of any one of clauses 1-111, wherein Y 1 , Y 2 , and Y 3 are each independently selected CR 1 .

113. The compound of any one of clauses 1-107 or 112, wherein Y 1 , Y 2 , and Y 3 are each CH.

114. The compound of any one of clauses 1-107 or 112, wherein one of Y 1 , Y 2 , and Y 3 is CR C , optionally C-halo; and each of the remaining two Y 1 , Y 2 , and Y 3 is CH.

115. The compound of any one of clauses 1-114, wherein X 1 is NR 2 .

116. The compound of any one of clauses 1-115, wherein X 1 is NH.

117. The compound of any one of clauses 1-116, wherein X 2 is CR 5 .

118. The compound of any one of clauses 1-117, wherein X 2 is CH.

119. The compound of any one of clauses 1-114, wherein X 1 is NR 2 ; and X 2 is CR 5

120. The compound of any one of clauses 1-114 or 119, wherein X 1 is NH; and X 2 is CH.

121. The compound of any one of clauses 1-104, wherein Y 1 , Y 2 , and Y 3 are each an independently selected CR 1 ; X 1 is NR 2 ; and X 2 is CR 5 . 122. The compound of any one of clauses 1-104 or 121, wherein Y 1 , Y 2 , and Y 3 are each CH; X 1 is NH; and X 2 is CH.

123. The compound of any one of clauses 1-122, wherein R 6 is H.

124. The compound of any one of clauses 1-123, wherein W is Ci-io alkyl, C2-10 alkenyl, or C2-10 alkenyl, each of which is optionally substituted with 1-6 R a2 or R g .

125. The compound of any one of clauses 1-124, wherein W is C1-10 alkyl, which is optionally substituted with 1-6 R a2 .

126. The compound of any one of clauses 1-125, wherein W is C1-6 alkyl, which is optionally substituted with 1-6 R a2 .

127. The compound of any one of clauses 1-126, wherein W is Ci-4 alkyl, which is optionally substituted with 1-6 R a2 .

128. The compound of any one of clauses 1-127, wherein W is unsubstituted Ci- 4 alkyl.

129. The compound of any one of clauses 1-128, wherein W is selected from the group consisting of: methyl, ethyl, n-propyl, isopropyl, and isobutyl

130. The compound of any one of clauses 1-129, wherein W is methyl or ethyl.

131. The compound of any one of clauses 1-126, wherein W is Ci-4 alkyl, which is substituted with 1-6 R a2 .

132. The compound of any one of clauses 1-126 or 131, wherein each R a2 is independently selected from the group consisting of: i. -OH; -halo; -NR e R f ; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=O)O(Ci-4 alkyl); - C(=O)(Ci-4 alkyl); and cyano; or ii. halo; -OH; Ci-4 alkoxy; and Ci-4 haloalkoxy. 133. The compound of any one of clauses 1-126 or 131-132, wherein W: i. Ci-4 alkyl which is substituted with 1-3 substituents each independently selected from the group consisting of: halo; -OH; Ci-4 alkoxy; and Ci-4 haloalkoxy; or

.0, ii. selected from the group consisting of: W is

134. The compound of any one of clauses 1-123, wherein W is selected from the group consisting of:

• C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• heteroaryl, heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl, heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c , provided that when W is heteroaryl, heterocyclyl or heterocycloalkenyl, it is attached to the C(=O)NR 6 group via a ring carbon atom;

135. The compound of any one of clauses 1-123 or 134, wherein W is C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

136. The compound of any one of clauses 1-123 or 134-135, wherein W is C3-12 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

137. The compound of any one of clauses 1-123 or 134-136, wherein W is unsubstituted C3-12 cycloalkyl. 138. The compound of any one of clauses 1-123 or 134-137, wherein W is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

139. The compound of any one of clauses 1-123 or 134-138, wherein W is cyclobutyl.

140. The compound of any one of clauses 1-123, wherein W is H.

141. The compound of clause 1, wherein the compound is a compound of

Formula (I-a):

Formula (I-a) or a pharmaceutically acceptable salt thereof, wherein:

L 1 is selected from the group consisting of: -O-, -N(H)-, and -N(R d )-;

L 2 is selected from the group consisting of:

• straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b ;

• C3-8 cycloalkylene, which is optionally substituted with 1-3 R c ; and

• heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclylene is optionally substituted with 1-3 R c .

142. The compound of clause 141, wherein L 1 is -O-.

143. The compound of clauses 141 or 142, wherein L 2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 R b .

144. The compound of any one of clauses 141-143, wherein L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2-, optionally wherein L 2 is -CH2-. 145. The compound of any one of clauses 141-143 wherein L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b .

146. The compound of any one of clauses 141-143 or 145, wherein L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -Q 1 .

147. The compound of clause 146, wherein L 2 is -CH2CH2-.

148. The compound of any one of clauses 141-143, wherein L 2 is straight-chain C3 alkylene which is optionally substituted with 1-3 R b .

149. The compound of clauses 141 or 142, wherein L 2 is: is optionally substituted with 1-2 R c , wherein nl and n2 are independently 0, 1, or 2; Q 2 is CH, CR C , or N; and the asterisk represents the point of attachment to Q 1 ;

150. The compound of clause 149, wherein nl and n2 are independently 0 or 1, optionally 0; and Q 2 is CH; optionally wherein nl and n2 are 0 and Q 2 is CH; optionslly wherein L 2 is cyclobutane-diyl optionally substituted with 1-2 R c ; optionslly wherein L 2 is cyclobutane-l,3-diyl optionally substituted with 1-2 R c ;optionslly wherein L 2 is cyclobutane-diyl optionally substituted with 1-2 R c ; optionslly wherein L 2 is unsubstituted cyclobutane-diyl; optionally wherein L 2 is unsubstituted cyclobutane-l,3-diyl.

151. The compound of clause 141, wherein L 1 is -O-; and L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

152. The compound of clause 151, wherein L 2 is: i. straight-chain C2 alkylene which is optionally substituted with 1-3 R b ; ii. selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and - CH2C(R b )2-*, wherein the asterisk represents point of attachment to -Q 1 ; or iii. -CH2CH2-. 153. The compound of clause 141, wherein L 1 is -O-; and L 2 is: i. selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2; or ii. -CH2-.

154. The compound of clause 1, wherein the compound is a compound of

Formula (I-b):

Formula (I-b) or a pharmaceutically acceptable salt thereof, wherein:

L 2 is straight-chain C1-6 alkylene or straight-chain C2-6 alkenylene, each of which is optionally substituted with 1-6 R b .

155. The compound of clause 154, wherein L 2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 R b .

156. The compound of clauses 154 or 155, wherein L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b .

157. The compound of any one of clauses 154-156, wherein L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -Q 1 , optionally wherein L 2 is -CH2CH2-.

158. The compound of clauses 154-155, wherein L 2 is straight-chain C3 alkylene which is optionally substituted with 1-3 R b .

159. The compound of any one of clauses 154-155 or 158, wherein L 2 is selected from the group consisting of: , and R b asterisk represents point of attachment to -Q 1 , optionally

160. The compound of clause 154, wherein L 2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 R b .

161. The compound of clauses 154 or 160, wherein L 2 is selected from the group consisting of: wherein the asterisk represents the point of attachment to -Q 1 .

162. The compound of clause 1, wherein the compound is a compound of

Formula (I-c):

Formula (I-c) or a pharmaceutically acceptable salt thereof, wherein:

L 2 and L 4 are independently selected straight-chain C1-3 alkylene which is optionally substituted with 1-6 R b ; and

L 3 is selected from the group consisting of: -O-, -N(H)-, and -N(R d )-.

163. The compound of clause 162, wherein L 2 and L 4 are independently selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2.

164. The compound of clauses 162 or 163, wherein L 2 and L 4 are each -CH2-.

165. The compound of any one of clauses 162-164, wherein L 3 is -O-. 166. The compound of any one of clauses 162-164, wherein L 3 is -N(H)- or - N(R d )-, optionally -N(H)-.

167. The compound of clause 1, wherein the compound is a compound of Formula (I-d):

Formula (I-d) or a pharmaceutically acceptable salt thereof, wherein:

L 2 is straight-chain C1-3 alkylene which is optionally substituted with 1-6 R b ; and L 3 is selected from the group consisting of: -O-, -N(H)-, and -N(R d )-.

168. The compound of clause 167, wherein L 2 is selected from the group consisting of: -CH2-, -CHR b -, and -C(R b )2.

169. The compound of clause 167, wherein L 2 is straight-chain C2 alkylene which is optionally substituted with 1-3 R b .

170. The compound of clauses 167 or 169, wherein L 2 is selected from the group consisting of: -CH2CH2-, -CH2CH(R b )-*, and -CH2C(R b )2-*, wherein the asterisk represents point of attachment to -L 3 , optionally wherein L 2 is -CH2CH2-.

171. The compound of any one of clauses 167-170, wherein L 3 is -O-.

172. The compound of any one of clauses 167-170, wherein L 3 is -N(H)- or - N(R d )-, optionally -N(H)-.

173. The compound of any one of clauses 141-172, wherein Q 1 is selected from the group consisting of: • heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’.

174. The compound of any one of clauses 141-173, wherein Q 1 is selected from the group consisting of:

• heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 R c ’ ; and

• phenyl optionally substituted with 1-3 R c ’.

175. The compound of any one of clauses 141-174, wherein Q 1 is: i. phenyl or pyridyl, each optionally substituted with 1-3 R c ’; iii. any groups of i or ii, wherein each R c present in Q 1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo; or iv. any groups of i or ii, wherein each R c present in Q 1 is independently selected from the group consisting of: -F, -Cl, and -CF3.

176. The compound of any one of clauses 141-172, wherein Q 1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ’.

177. The compound of any one of clauses 141-172 or 176, wherein Q 1 is: i. , wherein ml and m2 are each independently 0, 1, or 2; ii. iii any groups of i or ii, wherein the R d present in Q 1 is selected from the group consisting of: -C(O)O(Ci-4 alkyl); and Ci-6 alkyl optionally substituted with 1-3 independently selected R a ; or iv. any groups of i or ii, wherein the R d present in Q 1 is C2-3 alkyl substituted with 1-3 -F.

178. The compound of any one of clauses 141-177, wherein each R 1 is H.

179. The compound of any one of clauses 141-177, wherein one occurrence of R 1 is R c ; and each remaining R 1 is H.

180. The compound of any one of clauses 141-179, wherein R 2 is H; and R 5 is H.

181. The compound of any one of clauses 141-180, wherein W is Ci-6 alkyl, which is optionally substituted with 1-6 R a2 .

182. The compound of any one of clauses 141-181, wherein W is unsubstituted Ci -4 alkyl.

183. The compound of any one of clauses 141-182, wherein W is methyl or ethyl.

184. The compound of any one of clauses 141-181, wherein W is Ci-4 alkyl, which is substituted with 1-6 R a2 .

185. The compound of any one of clauses 141-181 or 184, wherein W: i. Ci-4 alkyl which is substituted with 1-3 substituents each independently selected from the group consisting of: halo; -OH; Ci-4 alkoxy; and Ci-4 haloalkoxy; or

.0. ii. selected from the group consisting of: W is 186. The compound of any one of clauses 141-181, wherein W is selected from the group consisting of:

• monocyclic C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c ; and

• monocyclic heterocyclyl or heterocycloalkenyl of from 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

187. The compound of any one of clauses 141-181 or 186, wherein W is monocyclic C3-8 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R c .

188. The compound of any one of clauses 141-181 or 186-187, wherein W is: i. unsubstituted C3-8 cycloalkyl; or ii. cyclobutyl.

189. The compound of clause 1, wherein the compound is selected from the group consisting of compounds delineated in Table Cl, and a pharmaceutically acceptable salt thereof.

190. A pharmaceutical composition comprising a compound of clauses 1-189 and one or more pharmaceutically acceptable excipients.

191. A method for inhibiting STING activity, the method comprising contacting STING with a compound or a pharmaceutically acceptable salt thereof as defined in any one of clauses 1-126; or a pharmaceutical composition as defined in clause 190. 192. The method of clause 191, wherein the inhibiting comprises antagonizing STING.

193. The method of any one of clauses 191-192, which is carried out in vitro.

194. The method of clauses 193, wherein the method comprises contacting a sample comprising one or more cells comprising STING with the compound.

195. The method of clauses 193 or 194, wherein the one or more cells are one or more cancer cells.

196. The method of clauses 194 or 195, wherein the sample further comprises one or more cancer cells, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

197. The method of clauses 191 or 192, which is carried out in vivo.

198. The method of clause 197, wherein the method comprises administering the compound to a subject having a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease.

199. The method of clause 198, wherein the subject is a human.

200. The method of clause 199, wherein the disease is cancer. 201. The method of clause 200, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

202. The method of clauses 200 or 201, wherein the cancer is a refractory cancer.

203. The method of clause 198, wherein the compound is administered in combination with one or more additional cancer therapies.

204. The method of clause 203, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.

205. The method of clause 204, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.

206. The method of clause 205, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g., azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisomerase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an antihelminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 -PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL- 10, transforming growth factor-P (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM

- LIGHT, HVEM-BTLA-CD160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48

- CD244, CD244, ICOS, ICOS-ICOS ligand, B7 H3, B7 H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD1 or PD-L1).

207. The method of any one of clauses 198-206, wherein the compound is administered intratum orally.

208. A method of treating cancer, comprising administering to a subject in need of such treatment an effective amount of a compound as defined in any one of clauses 1- 189, or a pharmaceutical composition as defined in clause 190. 209. The method of clause 208, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

210. The method of clause 208 or 209, wherein the cancer is a refractory cancer.

211. The method of clause 208, wherein the compound is administered in combination with one or more additional cancer therapies.

212. The method of clause 211, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.

213. The method of clause 212, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.

214. The method of clause 212, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g., azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisomerase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an antihelminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 -PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL- 10, transforming growth factor-P (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM

- LIGHT, HVEM-BTLA-CD160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48

- CD244, CD244, ICOS, ICOS-ICOS ligand, B7 H3, B7 H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD1 or PD-L1).

215. The method of any one of clauses 208-214, wherein the compound is administered intratum orally.

216. A method of inducing an immune response in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.

217. The method of clause 216, wherein the subject has cancer.

218. The method of clause 217, wherein the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies.

219. The method of clause 217, wherein the cancer selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

220. The method of clause any one of clauses 217-219, wherein the cancer is a refractory cancer.

221. The method of clause 219, wherein the immune response is an innate immune response.

222. The method of clause 221 , wherein the at least one or more cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.

223. The method of clause 222, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents. 224. The method of clause 223, wherein the one or more additional chemotherapeutic agents is selected from alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g., azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisomerase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an antihelminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 -PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL- 10, transforming growth factor-P (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM

- LIGHT, HVEM-BTLA-CD160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48

- CD244, CD244, ICOS, ICOS-ICOS ligand, B7 H3, B7 H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD1 or PD-L1).

225. A method of treatment of a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease, comprising administering to a subject in need of such treatment an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.

226. A method of treatment comprising administering to a subject having a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.

227. A method of treatment comprising administering to a subject a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190, wherein the compound or composition is administered in an amount effective to treat a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.

228. The method of any one of clauses 225-227, wherein the disease is cancer.

229. The method of clause 228, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.

230. The method of clause 228 or 229, wherein the cancer is a refractory cancer.

231. The method of any one of clauses 228-230, wherein the compound is administered in combination with one or more additional cancer therapies.

232. The method of clause 231, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.

233. The method of clause 232, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.

234. The method of clause 233, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g., azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisomerase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an antihelminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 -PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL- 10, transforming growth factor-P (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM

- LIGHT, HVEM-BTLA-CD160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48

- CD244, CD244, ICOS, ICOS-ICOS ligand, B7 H3, B7 H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD1 or PD-L1).

235. The method of any one of clauses 225-234, wherein the compound is administered intratum orally.

236. A method of treatment of a disease, disorder, or condition associated with STING, comprising administering to a subject in need of such treatment an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.

237. The method of clause 236, wherein the disease, disorder, or condition is selected from type I interferonopathies, Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, inflammation-associated disorders, and rheumatoid arthritis. 238. The method of clause 237, wherein the disease, disorder, or condition is a type I interferonopathy (e.g., STING-associated vasculopathywith onset in infancy (SAVI)).

239. The method of clause 238, wherein the type I interferonopathy is STING- associated vasculopathy with onset in infancy (SAVI)).

240. The method of clause 237, wherein the disease, disorder, or condition is Aicardi-Goutieres Syndrome (AGS).

241. The method of clause 237, wherein the disease, disorder, or condition is a genetic form of lupus.

242. The method of clause 237, wherein the disease, disorder, or condition is inflammation-associated disorder.

243. The method of clause 242, wherein the inflammation-associated disorder is systemic lupus erythematosus.

244. The method of any one of clauses 191-243, wherein the method further comprises identifying the subject.

245. A combination comprising a compounds defined in any one of clauses 1 to 126 or a pharmaceutically acceptable salt or tautomer thereof, and one or more therapeutically active agents.

246. A compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 190, for use as a medicament. 247. A compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 190, for use in the treatment of a disease, condition or disorder modulated by STING inhibition. 248. A compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or the pharmaceutical composition defined in clause 190, for use in the treatment of a disease mentioned in any one of clauses 191-244.

249. Use of a compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 190, in the manufacture of a medicament for the treatment of a disease mentioned in in any one of clauses 191-244.