Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
OXALAMIDE COMPOUNDS AND COMPOSITIONS FOR TREATING CONDITIONS ASSOCIATED WITH STING ACTIVITY
Document Type and Number:
WIPO Patent Application WO/2021/067791
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:
SEIDEL HANS (US)
ROUSH WILLIAM (US)
KATZ JASON (US)
VENKATRAMAN SHANKAR (US)
Application Number:
PCT/US2020/054054
Publication Date:
April 08, 2021
Filing Date:
October 02, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
IFM DUE INC (US)
International Classes:
C07D471/04; A61K31/437; A61K31/444; A61K31/4453; A61K31/47; A61P25/00; A61P31/04; A61P31/10; A61P31/14; A61P33/00; A61P35/00; A61P37/06
Domestic Patent References:
WO2017175156A12017-10-12
WO2015061294A22015-04-30
Foreign References:
US7927613B22011-04-19
US20120202848A12012-08-09
Other References:
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 15 October 2017 (2017-10-15), AURORA FINE CHEMICALS: "1-Piperidineacetamide, N-(4-ethyl-1,6-dihydro-6-methylpyrazolo[3,4-c]pyrazol-3-yl)-4-methyl-a-oxo-", XP002801603, Database accession no. 2134768-45-7
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 11 October 2017 (2017-10-11), AURORA FINE CHEMICALS: "1-Piperidineacetamide, N-(4-ethyl-1,6-dihydro-6-methylpyrazolo[3,4-c]pyrazol-3-yl)-3-methyl-a-oxo-", XP002801604, Database accession no. 2133761-75-6
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 22 September 2017 (2017-09-22), AURORA FINE CHEMICALS: "1-Piperidineacetamide, N-(1,6-dihydro-4,6-dimethylpyrazolo[3,4-c]pyrazol-3-yl)-4-methyl-a-oxo", XP002801605, Database accession no. 2129872-27-9
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 3 May 2017 (2017-05-03), ENAMINE LLC: "Ethanediamide, N1-[3-cyano-4-(1-methylethoxy)phenyl]-N2-(4-ethyl-1,6-dihydro-6-methylpyrazolo[3,4-c]pyrazol-3-yl)-", XP002801606, Database accession no. 2094846-52-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 2 May 2017 (2017-05-02), ENAMINE LLC: "Ethanediamide, N1-(4-ethyl-1,6-dihydro-6-methylpyrazolo[3,4-c]pyrazol-3-yl)-N2-(4-methylphenyl)-", XP002801607, Database accession no. 2094483-39-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 29 September 2015 (2015-09-29), ENAMINE LLC: "Ethanediamide, N1-(4-chloro-2-methylphenyl)-N2-(1,6-dihydro-4,6-dimethylpyrazolo[3,4-c]pyrazol-3-yl)-", XP002801608, Database accession no. 1808827-30-6
HAAG SIMONE M ET AL: "Targeting STING with covalent small-molecule inhibitors", NATURE, MACMILLAN JOURNALS LTD, LONDON, vol. 559, no. 7713, 4 July 2018 (2018-07-04), pages 269 - 273, XP036553086, ISSN: 0028-0836, [retrieved on 20180704], DOI: 10.1038/S41586-018-0287-8
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
HIGUCHI, T. ET AL.: "Pro-drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
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: Z is selected from the group consisting of a bond, CR1, C(R3)2, N, and NR2; each of Y1, Y2, and Y3 is independently selected from the group consisting of O, S, CR1, C(R3)2, N, and NR2; Y4 is C or N; 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 five- membered ring comprising Y4, X1, and X2 is heteroaryl; Q-A is defined according to (A) or (B) below: (A) Q is selected from the group consisting of: NH and N(C1-6 alkyl) wherein the C1-6 alkyl is optionally substituted with 1-2 independently selected Ra; and A is: (i) -(YA1)n-YA2, wherein: ● n is 0 or 1; ● YA1 is C1-6 alkylene, which is optionally substituted with 1-6 substituents each indepndently selected from the group consisting of: o Ra; o C6-10 aryl optionally substituted with 1-4 independently selected C1- 4 alkyl; and o heteroaryl of 5-10 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(O)0-2, and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C1-4 alkyl; or ● YA1 is –YA3-YA4-YA5 which is connected to Q via YA3 wherein: o YA3 is a C1-3 alkylene optionally substituted with 1-2 independently selected Ra; o YA4 is –O-, -NH-, or -S-; and o YA5 is a bond or C1-3 alkylene which is optionally substituted with 1-2 independently selected Ra; and ● YA2 is: (a) C3-20 cycloalkyl or C3-20 cycloalkenyl, each of which is optionally substituted with 1-4 Rb, (b) C6-20 aryl, which is optionally substituted with 1-4 Rc; (c) heteroaryl of 5-20 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(O)0-2, and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected Rc; or (d) heterocyclyl or heterocycloalkenyl of 3-16 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(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-4 independently selected Rb, or (ii) -Z1-Z2-Z3, wherein: ● Z1 is C1-3 alkylene, which is optionally substituted with 1-4 Ra; ● Z2 is –N(H)-, -N(Rd)-, -O-, or –S-; and ● Z3 is C2-7 alkyl, which is optionally substituted with 1-4 Ra; or (iii) C1-20 alkyl, which is optionally substituted with 1-6 independently selected Ra, or (B) Q and A, taken together, form: E is a ring of 3-16 ring atoms, wherein, 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the ring is optionally substituted with 1-4 independently selected Rb, each occurrence of R1 is independently selected from the group consisting of H; halo; cyano; C1-6 alkyl optionally substituted with 1-2 Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 haloalkyl; C1-4 alkoxy; C1-4 haloalkoxy; –L3-L4-Ri; -S(O)1-2(C1-4 alkyl); - S(O)(=NH)(C1-4 alkyl); SF5; -NReRf; –OH; oxo; -S(O)1-2(NR’R’’); -C1-4 thioalkoxy; -NO2; -C(=O)(C1-4 alkyl); -C(=O)O(C1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’); or a pair of R1 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C1-6 alkyl, halo, C1-6 haloalkyl, -OH, NReRf, C1-6 alkoxy, and C1-6 haloalkoxy, each occurrence of R2 is independently selected from the group consisting of: (i) C1-6 alkyl, which is optionally substituted with 1-2 independently selected Ra; (ii) C3-6 cycloalkyl or C3-6 cycloalkenyl; (iii) heterocyclyl or heterocycloalkenyl of 3-10 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(O)0-2; (iv) C6-10 aryl; (v) heteroaryl of 5-10 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(O)0-2; (vi) -C(O)(C1-4 alkyl); (vii) -C(O)O(C1-4 alkyl); (viii) -CON(R’)(R’’); (ix) -S(O)1-2(NR’R’’); (x) - S(O)1-2(C1-4 alkyl); (xi) -OH; (xii) C1-4 alkoxy; and (xiii) H; or a pair of R1 and R2 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C1-6 alkyl, halo, C1-6 haloalkyl, -OH, NReRf, C1-6 alkoxy, and C1-6 haloalkoxy, each occurrence of R3 is independently selected from the group consisting of H; C1- 6 alkyl optionally substituted with 1-6 independently selected Ra; C1-4 haloalkyl; –OH; -F; -Cl; -Br; –NReRf; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)(C1-4 alkyl); -C(=O)O(C1-4 alkyl); - C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C1-4 alkyl); cyano; and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C1-4 alkyl; or two R3 on the same carbon combine to form an oxo; or a pair of R3 on the same or on adjacent atoms, taken together with the atom(s) connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C1-6 alkyl, halo, C1-6 haloalkyl, -OH, NReRf, C1-6 alkoxy, and C1-6 haloalkoxy; or a pair of R1 and R3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C1-6 alkyl, halo, C1-6 haloalkyl, -OH, NReRf, C1-6 alkoxy, and C1-6 haloalkoxy; or a pair of R2 and R3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C1-6 alkyl, halo, C1-6 haloalkyl, -OH, NReRf, C1-6 alkoxy, and C1-6 haloalkoxy; R4 is selected from the group consisting of H and C1-6 alkyl optionally substituted with 1-3 independently selected Ra; R5 is selected from the group consisting of H; halo; –OH; -C1-4 alkyl; -C1-4 haloalkyl; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)O(C1-4 alkyl); -C(=O)(C1-4 alkyl); - C(=O)OH; -CON(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C1-4 alkyl); cyano, and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C1-4 alkyl; R6 is selected from the group consisting of H; C1-6 alkyl optionally substituted with 1-3 independently selected Ra; -OH; C1-4 alkoxy; C(=O)H; C(=O)(C1-4 alkyl); C6-10 aryl optionally substituted with 1-4 independently selected C1-4 alkyl; and heteroaryl of 5-10 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(O)0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C1-4 alkyl; each occurrence of Ra is independently selected from the group consisting of: – OH; -F; -Cl; -Br; –NReRf; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)O(C1-4 alkyl); -C(=O)(C1- 4 alkyl); -C(=O)OH; -CON(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C1-4 alkyl); cyano, and C3- 6 cycloalkyl or C3-6 cycloalkenyl , each optionally substituted with 1-4 independently selected C1-4 alkyl; each occurrence of Rb is independently selected from the group consisting of: C1- 10 alkyl optionally substituted with 1-6 independently selected Ra; C1-4 haloalkyl; –OH; oxo; -F; -Cl; -Br; –NReRf; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)(C1-10 alkyl); -C(=O)O(C1- 4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C1-4 alkyl); cyano; and –L1-L2-Rh; each occurrence of Rc is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy optionally substituted with 1-2 independently selected Ra; C1-4 haloalkoxy; -S(O)1-2(C1-4 alkyl) or –S(O)1-2(C1-4 haloalkyl); -NReRf; –OH; -S(O)1- 2(NR’R’’); -C1-4 thioalkoxy or –C1-4 thiohaloalkoxy; -NO2; –SF5; -C(=O)(C1-10 alkyl); - C(=O)O(C1-4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); and –L1-L2-Rh; Rd is selected from the group consisting of: C1-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; -C(O)(C1-4 alkyl); - C(O)O(C1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C1-4 alkyl); -OH; and C1-4 alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; C1-6 alkyl; C1-6 haloalkyl; C3-6 cycloalkyl or C3-6 cycloalkenyl; -C(O)(C1-4 alkyl); - C(O)O(C1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C1-4 alkyl); -OH; and C1-4 alkoxy; or Re and Rf together with the nitrogen atom to which each is attached forms a ring of 3-8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to Re and Rf), which are each independently selected from the group consisting of N(Rd), NH, O, and S; -L1 is a bond or C1-3 alkylene; -L2 is –O-, -N(H)-, -S(O)0-2-, or a bond; Rh is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1- 4 haloalkoxy; ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O)0-2, wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1- 4 haloalkoxy; ● heteroaryl of 5-10 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(O)0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1-4 haloalkoxy; -L3 is a bond or C1-3 alkylene; -L4 is –O-, -N(H)-, -S(O)0-2-, or a bond; Ri is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1- 4 haloalkoxy; ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O)0-2, wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1- 4 haloalkoxy; ● heteroaryl of 5-10 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(O)0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C1-4 alkyl optionally substituted with 1-2 independently selected Ra; C1-4 haloalkyl; cyano; C1-4 alkoxy; and C1-4 haloalkoxy; and each occurrence of R’ and R’’ is independently selected from the group consisting of: H, -OH, C1-4 alkyl, C6-10 aryl optionally substituted with 1-2 substituents selected from the group consisting of halo, C1-4 alkyl, and C1-4 haloalkyl, and heteroaryl of 5-10 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(O)0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, -OH, NH2, NH(C1-4 alkyl), N(C1-4 alkyl)2, C1-4 alkyl, and C1-4 haloalkyl; or R’ and R’’ together with the nitrogen atom to which each is attached forms a ring of 3- 8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R’’), which are each independently selected from the group consisting of N(H), N(C1-6 alkyl), O, and S; provided that one or more of a), b), and c) apply: a) one or more of Z, Y1, Y2, Y3, and Y4 in the ring below independently selected heteroatom; b) the ring that includes Z, Y1, Y2, Y3, and Y4 is partially unsaturated; or c) Z is a bond; and further provided that the compound is other than: . 2. The compound of claim 1, wherein is aromatic. 3. The compound of claims 1 or 2, wherein 1-2 of Y1, Y2, and Y3 is independently N or NR2, such as N; and each of the remaining of Y1, Y2, and Y3 is an independently selected CR1, optionally wherein Z is CR1, such as:

wherein the moiety , wherein the asterisk denotes point of attachment to Y4. 4. The compound of claims 1 or 2, wherein Z is N; and optionally each of Y1, Y2, and Y3 is an independently selected CR1, such as: wherein the moiety is such as or , wherein the asterisk denotes point of attachment to Y4. 5. The compound of any one of claims 1-4, wherein X1 is NR2, such as wherein X1 is NH; and optionally wherein Y4 is C. 6. The compound of any one of claims 1-5, wherein X2 is CR5, such as wherein X2 is CH; and optionally wherein Y4 is C. 7. The compound of any one of claims 1-6, wherein 1-2 occurrences of R1 is other than H, such as: wherein one occurrence of R1 is selected from the group consisting of: halo; cyano; C1-6 alkyl optionally substituted with 1-2 Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 haloalkyl; C1- 4 alkoxy; C1-4 haloalkoxy; -S(O)1-2(C1-4 alkyl); -S(O)1-2(NR’R’’); -NO2; -C(=O)(C1-4 alkyl); -C(=O)O(C1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’), such as: wherein one occurrence of R1 is halo, such as –F or -Cl.

8. The compound of any one of claims 1-7, wherein wherein Q-A is defined according to (A), optionally wherein Q is NH. 9. The compound of any one of claims 1-8, wherein A is -(YA1)n-YA2. 10. The compound of any one of claims 1-9, wherein YA1 is C1-6 alkylene, which is optionally substituted with 1-4 Ra, such as: wherein YA1 is -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH(CF3)-, -CH2CH(OH)-, wherein YA1 is –CH2-. 11. The compound of any one of claims 1-10, wherein YA2 is phenyl substituted with 1-3 Rc, wherein one Rc is at the ring carbon para to the point of attachment to YA1; or wherein YA2 is phenyl substituted with 1-3 Rc, wherein 1-2 Rc is at the ring carbons meta to the point of attachment to YA1; or wherein YA2 is phenyl substituted with 1-3 Rc, wherein 1-2 Rc is at the ring carbons ortho to the point of attachment to YA1; or wherein YA2 is C7-10 bicyclic aryl, which is optionally substituted with 1-3 Rc, such as wherein YA2 is selected from the group consisting of: naphthyl, such as ; indanyl, such a tetrahydronapthyl, each of which is optionally substituted with 1- 3 Rc); optionally wherein each occurrence of Rc is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy; C1-4 haloalkoxy; -S(O)1- 2(C1-4 alkyl); -NReRf; -C1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C1-4 alkyl); and–L1-L2-Rh. 12. The compound of any one of claims 1-10, wherein YA2 is heteroaryl of 5- 14 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(O)0-2, and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected Rc; optionally wherein each occurrence of Rc is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy; C1-4 haloalkoxy; -S(O)1- 2(C1-4 alkyl); -NReRf; -C1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C1-4 alkyl); and–L1-L2-Rh. 13. The compound of any one of claims 1-10, wherein YA2 is C3-6 cycloalkyl or C3-6 cycloalkenyl, each of which is substituted with 1-4, such as 1-2, Rb; such as: wherein YA2 is cyclopropyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with 1-2 Rb; or wherein YA2 is bicyclic, tricyclic, or polycyclic C7-20, such as C7-12, cycloalkyl or C7-20, such as C7-12, cycloalkenyl, each optionally substituted with 1-2 Rb, such as: wherein YA2 is selected from the group consisting of: spiro[5.5]undecanyl, such as ; bicyclo[2.2.1]hept-2-enyl, such as bicyclo[2.2.1]heptanyl, such as ; spiro[2.5]octanyl, such as ; and adamantly, such as ; optionally wherein each occurrence of Rb is independently selected from the group consisting of: C1-10 alkyl optionally substituted with 1-6 independently selected Ra; C1-4 haloalkyl; -F; -Cl; -Br; cyano; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)(C1-10 alkyl); - C(=O)O(C1-4 alkyl); -S(O)1-2(C1-4 alkyl); oxo; cyano; and –L1-L2-Rh. 14. The compound of any one of claims 1-7, wherein Q-A is as defined according to (B); and E a ring of 5-8 ring atoms, wherein aside from the nitrogen atom present, 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the ring is optionally substituted with 1-4 independently selected Rb, such as: wherein E is piperidinyl which is optionally substituted with 1-2 independently selected Rb , such as: wherein E is , and Rb is C1-6 alkyl. 15. The compound of claim 1, wherein the compound is a compound of Formula (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), or (I-8), or a pharmaceutically acceptable salt thereof: wherein n1 is 0, 1, or 2; each of RcA and RcB is an independently selected Rc; and R7 is H or C1-4 alkyl; or wherein n1 is 0, 1, or 2; each of RcA and RcB is an independently selected Rc; and R7 is H or C1-4 alkyl; or wherein one of Q1 and Q2 is N; the other one of Q1 and Q2 is CH; n1 is 0, 1, or 2; each of RcA and RcB is an independently selected Rc; and R7 is H or C1-4 alkyl; or wherein one of Q1, Q2, Q3, and Q4 is N; each of the remaining of Q1, Q2, Q3, Q4 is CH; n1 is 0, 1, or 2; and each of RcA and RcB is an independently selected Rc; and R7 is H or C1-4 alkyl; or wherein B1 is selected from the group consisting of: (a) bicyclic or tricyclic heteroaryl of 7-14 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(O)0-2, and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected Rc; and (b) C7-10 bicyclic aryl, which is optionally substituted with 1-3 Rc; and R7 is H or C1-4 alkyl; or wherein n2 is 0, 1, or 2; each of RbA and RbB is an independently selected Rb; and R7 is H or C1-4 alkyl; or wherein n2 is 0, 1, or 2; each of RbA and RbB is an independently selected Rb; and R7 is H or C1-4 alkyl; or wherein B2 is: bicyclic, tricyclic, or polycyclic C7-20 cycloalkyl or C7-20 cycloalkenyl, each optionally substituted with 1-2 Rb; and R7 is H or C1-4 alkyl; optionally wherein X1 is NH, X2 is CH, and Y4 is C in Formula (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), or (I-8); optionally wherein R6 is H in Formula (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), or (I-8); optionally wherein RcA in Formula (I-1), (I-2), (I-3), or (I-4) is selected from the group consisting of: RcA is selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy; C1-4 haloalkoxy; -S(O)1-2(C1-4 alkyl); -NReRf; -C1-4 thioalkoxy; - C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C1-4 alkyl); and –L1-L2-Rh; and optionally wherein RbA in Formula (I-6) or (I-7) is selected from the group consisting of: C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; -F; -Cl; and –L1-L2-Rh. 16. The compound of claim 1, wherein the compound is selected from the group consisting of the compounds delineated in Table C1, or a pharmaceutically acceptable salt thereof. 17. A pharmaceutical composition comprising a compound of claims 1-16 and one or more pharmaceutically acceptable excipients. 18. A method for inhibiting STING activity, the method comprising contacting STING with a compound as claimed in any one of claims 1-16, or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition as claimed in claim 17. 19. 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 1-16, or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition as claimed in claim 17. 20. 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- 16, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as claimed in claim 17.

Description:
QXALAMIDE 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. 62/910,162, filed on Oct 3, 2019; and U.S. Provisional Application Serial No.62/955,921, filed on Dec 31, 2019; each of 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 prodrug, and/or tautomer, 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-κB 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- Goutières 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 molecule- based 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 prodrug, and/or tautomer, 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 X 1 , X 2 , Y 1 , Y 2 , Y 3 , Y 4 , Z, Q, A, and R 6 can be as defined anywhere herein; and each is independently a single bond or a double bond, provided that the five- membered ring comprising Y 4 , X 1 , and X 2 is heteroaryl (i.e., one or more of Y 4 , X 1 , and X 2 is an independently selected heteroatom; and the 5-membered ring comprising Y 4 , X 1 , and X 2 is aromatic (as a non-limiting example, the ring comprising Y 4 , X 1 , and X 2 can be pyrrole)). In one aspect, compounds of Formula (I), or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a tautomer thereof, or any combination of the foregoing, are featured "Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of Formula (I)). Thus, the term "prodrug" refers to a precursor of a biologically active compound that is pharmaceutically acceptable. In some aspects, a prodrug is inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp.7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol.14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference 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-Goutières 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. 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-Goutières 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, Muromonab-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 anti- helminthic 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-β (TGFβ), 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–4‑1BB ligand, OX40–OX40 ligand, GITR, GITR ligand – GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25–TL1A, CD40L, CD40– CD40 ligand, HVEM–LIGHT–LTA, HVEM, HVEM – BTLA, HVEM – CD160, 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 intratumorally. 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 et al., Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed.; Gibson Ed.; CRC Press LLC: Boca Raton, 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, N-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, C1-10 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, iso-propyl, tert-butyl, n-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, C 2-6 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, C 2-6 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, 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]butane, bicyclo[2.1.0]pentane, bicyclo[1.1.1]pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, 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]pentane, spiro[2.5]octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro[4.5]decane, spiro[3.6]decane, spiro[5.5]undecane, 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, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-d]pyrimidinyl, pyrrolo[2,3- b]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-b]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridine, pyrazolo[4,3-b]pyridinyl, tetrazolyl, chromane, 2,3-dihydrobenzo[b][1,4]dioxine, benzo[d][1,3]dioxole, 2,3- dihydrobenzofuran, tetrahydroquinoline, 2,3-dihydrobenzo[b][1,4]oxathiine, isoindoline, 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]butane, 2-azabicyclo[2.1.0]pentane, 2- azabicyclo[1.1.1]pentane, 3-azabicyclo[3.1.0]hexane, 5-azabicyclo[2.1.1]hexane, 3- azabicyclo[3.2.0]heptane, octahydrocyclopenta[c]pyrrole, 3-azabicyclo[4.1.0]heptane, 7- azabicyclo[2.2.1]heptane, 6-azabicyclo[3.1.1]heptane, 7-azabicyclo[4.2.0]octane, 2- azabicyclo[2.2.2]octane, 3-azabicyclo[3.2.1]octane, 2-oxabicyclo[1.1.0]butane, 2- oxabicyclo[2.1.0]pentane, 2-oxabicyclo[1.1.1]pentane, 3-oxabicyclo[3.1.0]hexane, 5- oxabicyclo[2.1.1]hexane, 3-oxabicyclo[3.2.0]heptane, 3-oxabicyclo[4.1.0]heptane, 7- oxabicyclo[2.2.1]heptane, 6-oxabicyclo[3.1.1]heptane, 7-oxabicyclo[4.2.0]octane, 2- oxabicyclo[2.2.2]octane, 3-oxabicyclo[3.2.1]octane, 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]pentane, 4-azaspiro[2.5]octane, 1-azaspiro[3.5]nonane, 2- azaspiro[3.5]nonane, 7-azaspiro[3.5]nonane, 2-azaspiro[4.4]nonane, 6- azaspiro[2.6]nonane, 1,7-diazaspiro[4.5]decane, 7-azaspiro[4.5]decane 2,5- diazaspiro[3.6]decane, 3-azaspiro[5.5]undecane, 2-oxaspiro[2.2]pentane, 4- oxaspiro[2.5]octane, 1-oxaspiro[3.5]nonane, 2-oxaspiro[3.5]nonane, 7- oxaspiro[3.5]nonane, 2-oxaspiro[4.4]nonane, 6-oxaspiro[2.6]nonane, 1,7- dioxaspiro[4.5]decane, 2,5-dioxaspiro[3.6]decane, 1-oxaspiro[5.5]undecane, 3- oxaspiro[5.5]undecane, 3-oxa-9-azaspiro[5.5]undecane 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, “the ring that includes Z, Y 1 , Y 2 , Y 3 , and Y 4 is partially unsaturated” means that said ring may have any degree of unsaturation provided that the ring is not a , . As used herein, when a ring is described as being “aromatic”, it means said ring has a continuous, delocalized π-electron system. Typically, the number of out of plane π- electrons corresponds to the Hückel 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.0] ring systems, in which 0 represents a zero atom bridge (e.g., (ii) a single ring atom (spiro- fused ring systems) ( r (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. Some non-limiting exemplified compounds of the formulae described herein include one or more stereogenic carbon atoms. This disclosure provides examples of stereoisomer mixtures (e.g., racemic and non-racemic mixture of enantiomers; mixture of diastereomers, meso compounds). This disclosure also describes and exemplifies methods for separating individual components of said stereoisomer mixtures (e.g., resolving the enantiomers of a racemic mixture). In some instances, stereoisomers are graphically depicted using hashed and solid wedge three-dimensional representations. Unless otherwise indicated with “(R)” or “(S)” labels, the hashed and solid wedge three- dimensional representation are intended to convey relative stereochemistry only. Likewise, and unless otherwise indicated, reaction schemes showing resolution of a racemic mixture, the above-mentioned representations are intended only to convey that the constituent enantiomers were resolved in enantiopure pure form (about 98% ee or greater) and are not intended to disclose or imply any correlation between absolute configuration and order of elution. The definitions of certain variables herein include –L 1 -L 2 -R h and –L 3 -L 4 -R i . For avoidance of doubt, when a variable is –L 1 -L 2 -R h ; -L 1 is a bond; and –L 2 is a bond, then said variable is –R h , that is connected to the rest of the compound via a single bond. As a non-limiting example, when one occurrence of R b is –L 1 -L 2 -R h ; -L 1 is a bond; and –L 2 is a bond, then said occurrence of R b is –R h , that is connected to the rest of the compound via a single bond. Similarly, when a variable is –L 3 -L 4 -R i ; -L 3 is a bond; and –L 4 is a bond, then said variable is –R i , that is connected to the rest of the compound via a single bond. 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 prodrug, and/or tautomer, 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, compounds of Formula (I), a pharmaceutically acceptable salt thereof, or a tautomer thereof are featured: wherein: Z is selected from the group consisting of a bond, CR 1 , C(R 3 )2, N, and NR 2 ; each of Y 1 , Y 2 , and Y 3 is independently selected from the group consisting of O, S, CR 1 , C(R 3 )2, N, and NR 2 ; Y 4 is C or N; 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 five- membered ring comprising Y 4 , X 1 , and X 2 is heteroaryl; Q-A is defined according to (A) or (B) below: (A) Q is selected from the group consisting of: NH and N(C 1-6 alkyl) wherein the C 1-6 alkyl is optionally substituted with 1-2 independently selected R a ; and A is: (i) -(Y A1 )n-Y A2 , wherein: ● n is 0 or 1; ● Y A1 is C 1-6 alkylene, which is optionally substituted with 1-6 substituents each indepndently selected from the group consisting of: o R a ; o C6-10 aryl optionally substituted with 1-4 independently selected C1- 4 alkyl; and o heteroaryl of 5-10 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C 1-4 alkyl; or ● Y A1 is –Y A3 -Y A4 -Y A5 which is connected to Q via Y A3 wherein: o Y A3 is a C1-3 alkylene optionally substituted with 1-2 independently selected R a ; o Y A4 is –O-, -NH-, or -S-; and o Y A5 is a bond or C1-3 alkylene which is optionally substituted with 1-2 independently selected R a ; and ● Y A2 is: (a) C 3-20 cycloalkyl or C 3-20 cycloalkenyl, each of which is optionally substituted with 1-4 R b , (b) C 6-20 aryl, which is optionally substituted with 1-4 R c ; (c) heteroaryl of 5-20 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c ; or (d) heterocyclyl or heterocycloalkenyl of 3-16 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-4 independently selected R b , or (ii) -Z 1 -Z 2 -Z 3 , wherein: ● Z 1 is C1-3 alkylene, which is optionally substituted with 1-4 R a ; ● Z 2 is –N(H)-, -N(R d )-, -O-, or –S-; and ● Z 3 is C 2-7 alkyl, which is optionally substituted with 1-4 R a ; or (iii) C1-20 alkyl, which is optionally substituted with 1-6 independently selected R a , or (B) Q and A, taken together, form: E is a ring of 3-16 ring atoms, wherein, 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b , each occurrence of R 1 is independently selected from the group consisting of H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; -S(O) 1-2 (C 1-4 alkyl); - S(O)(=NH)(C 1-4 alkyl); SF5; -NR e R f ; –OH; oxo; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy; - NO2; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’); or a pair of R 1 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy, each occurrence of R 2 is independently selected from the group consisting of: (i) C 1-6 alkyl, which is optionally substituted with 1-2 independently selected R a ; (ii) C3-6 cycloalkyl or C3-6 cycloalkenyl; (iii) heterocyclyl or heterocycloalkenyl of 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(O) 0-2 ; (iv) C6-10 aryl; (v) heteroaryl of 5-10 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(O) 0-2 ; (vi) -C(O)(C 1-4 alkyl); (vii) -C(O)O(C 1-4 alkyl); (viii) -CON(R’)(R’’); (ix) -S(O)1-2(NR’R’’); (x) - S(O)1-2(C 1-4 alkyl); (xi) -OH; (xii) C 1-4 alkoxy; and (xiii) H; or a pair of R 1 and R 2 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R 2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy, each occurrence of R 3 is independently selected from the group consisting of H; C1- 6 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; –OH; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); - C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano; and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; or two R 3 on the same carbon combine to form an oxo; or a pair of R 3 on the same or on adjacent atoms, taken together with the atom(s) connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; or a pair of R 1 and R 3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; or a pair of R 2 and R 3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R 2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; R 4 is selected from the group consisting of H and C 1-6 alkyl optionally substituted with 1-3 independently selected R a ; R 5 is selected from the group consisting of H; halo; –OH; -C 1-4 alkyl; -C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)O(C 1-4 alkyl); -C(=O)(C 1-4 alkyl); - C(=O)OH; -CON(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano, and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; R 6 is selected from the group consisting of H; C 1-6 alkyl optionally substituted with 1-3 independently selected R a ; -OH; C 1-4 alkoxy; C(=O)H; C(=O)(C 1-4 alkyl); C6-10 aryl optionally substituted with 1-4 independently selected C 1-4 alkyl; and heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C 1-4 alkyl; each occurrence of R a is independently selected from the group consisting of: – OH; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)O(C 1-4 alkyl); -C(=O)(C1- 4 alkyl); -C(=O)OH; -CON(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano, and C3- 6 cycloalkyl or C3-6 cycloalkenyl , each optionally substituted with 1-4 independently selected C 1-4 alkyl; each occurrence of R b is independently selected from the group consisting of: C1- 10 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; –OH; oxo; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C 1-10 alkyl); -C(=O)O(C 1- 4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano; and –L 1 -L 2 -R h ; each occurrence of 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 ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl) or –S(O)1-2(C 1-4 haloalkyl); -NR e R f ; –OH; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy or –C 1-4 thiohaloalkoxy; -NO2; –SF5; - C(=O)(C1-10 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); and –L 1 -L 2 -R h ; R d is selected from the group consisting of: C 1-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; -C(O)(C 1-4 alkyl); - C(O)O(C 1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C 1-4 alkyl); -OH; and C 1-4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; C 1-6 alkyl; C 1-6 haloalkyl; C3-6 cycloalkyl or C3-6 cycloalkenyl; -C(O)(C 1-4 alkyl); - C(O)O(C 1-4 alkyl); -CON(R’)(R’’); -S(O) 1-2 (NR’R’’); - S(O) 1-2 (C 1-4 alkyl); -OH; and C 1-4 alkoxy; or R e and R f together with the nitrogen atom to which each is attached forms a ring of 3-8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R e and R f ), which are each independently selected from the group consisting of N(R d ), NH, O, and S; -L 1 is a bond or C1-3 alkylene; -L 2 is –O-, -N(H)-, -S(O) 0-2 -, or a bond; R h is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy (in certain embodiments, it is provided that when R h is C3-6 cycloalkyl or C3- 6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected C1- 4 alkyl, -L 1 is a bond, or –L 2 is –O-, -N(H)-, or -S-); ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy; ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; -L 3 is a bond or C1-3 alkylene; -L 4 is –O-, -N(H)-, -S(O) 0-2 -, or a bond; R i is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy (in certain embodiments, it is provided that when R i is C 3-6 cycloalkyl or C 3-6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected C 1-4 alkyl, -L 1 is a bond, or –L 2 is –O-, -N(H)-, or -S-); ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy; ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and each occurrence of R’ and R’’ is independently selected from the group consisting of: H, -OH, C 1-4 alkyl, C6-10 aryl optionally substituted with 1-2 substituents selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, and heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, -OH, NH2, NH(C 1-4 alkyl), N(C 1-4 alkyl)2, C 1-4 alkyl, and C 1-4 haloalkyl; or R’ and R’’ together with the nitrogen atom to which each is attached forms a ring of 3- 8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R’’), which are each independently selected from the group consisting of N(H), N(C 1-6 alkyl), O, and S; provided that one or more of a), b), and c) apply: a) one or more of Z, Y 1 , Y 2 , Y 3 , and Y 4 in the ring below independently selected heteroatom; b) the ring that includes Z, Y 1 , Y 2 , Y 3 , and Y 4 is partially unsaturated; or c) Z is a bond; and further provided that the compound is other than: . In one aspect, compounds of Formula (I), or a pharmaceutically acceptable salt thereof, are featured: Formula I or a pharmaceutically acceptable salt thereof or a tautomer thereof, wherein: Z is selected from the group consisting of a bond, CR 1 , C(R 3 )2, N, and NR 2 ; each of Y 1 , Y 2 , and Y 3 is independently selected from the group consisting of O, S, CR 1 , C(R 3 )2, N, and NR 2 ; Y 4 is C or N; 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 five- membered ring comprising Y 4 , X 1 , and X 2 is heteroaryl; Q-A is defined according to (A) or (B) below: (A) Q is selected from the group consisting of: NH and N(C 1-6 alkyl) wherein the C 1-6 alkyl is optionally substituted with 1-2 independently selected R a ; and A is: (i) -(Y A1 ) n -Y A2 , wherein: ● n is 0 or 1; ● Y A1 is C 1-6 alkylene, which is optionally substituted with 1-6 substituents each indepndently selected from the group consisting of: o R a ; o C6-10 aryl optionally substituted with 1-4 independently selected C1- 4 alkyl; and o heteroaryl of 5-10 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C 1-4 alkyl; and ● Y A2 is: (a) C 3-20 cycloalkyl or C 3-20 cycloalkenyl, each of which is optionally substituted with 1-4 R b , (b) C6-20 aryl, which is optionally substituted with 1-4 R c ; (c) heteroaryl of 5-20 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c ; or (d) heterocyclyl or heterocycloalkenyl of 3-16 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-4 independently selected R b , or (ii) -Z 1 -Z 2 -Z 3 , wherein: ● Z 1 is C1-3 alkylene, which is optionally substituted with 1-4 R a ; ● Z 2 is –N(H)-, -N(R d )-, -O-, or –S-; and ● Z 3 is C2-7 alkyl, which is optionally substituted with 1-4 R a ; or (iii) C1-10 alkyl, which is optionally substituted with 1-6 independently selected R a , or (B) Q and A, taken together, form: E is a ring of 3-16 ring atoms, wherein 0-3 additional ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b , each occurrence of R 1 is independently selected from the group consisting of H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; S(O)1-2(C 1-4 alkyl); - S(O)(=NH)(C 1-4 alkyl); SF5; -NR e R f ; –OH; oxo; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy; - NO2; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’); or a pair of R 1 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy, each occurrence of R 2 is independently selected from the group consisting of: (i) C 1-6 alkyl, which is optionally substituted with 1-2 independently selected R a ; (ii) C3-6 cycloalkyl or C3-6 cycloalkenyl; (iii) heterocyclyl or heterocycloalkenyl of 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(O) 0-2 ; (iv) C6-10 aryl; (v) heteroaryl of 5-10 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(O)0- 2; (vi) -C(O)(C 1-4 alkyl); (vii) -C(O)O(C 1-4 alkyl); (viii) -CON(R’)(R’’); (ix) -S(O)1-2(NR’R’’); (x) - S(O)1-2(C 1-4 alkyl); (xi) -OH; (xii) C 1-4 alkoxy; and (xiii) H; or a pair of R 1 and R 2 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R 2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy, each occurrence of R 3 is independently selected from the group consisting of H; C 1- 6 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; –OH; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); - C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano; and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; or two R 3 on the same carbon combine to form an oxo; or a pair of R 3 on the same or on adjacent atoms, taken together with the atom(s) connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; or a pair of R 1 and R 3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; or a pair of R 2 and R 3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R 2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; R 4 is selected from the group consisting of H and C 1-6 alkyl optionally substituted with 1-3 independently selected R a ; R 5 is selected from the group consisting of H; halo; –OH; -C 1-4 alkyl; -C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)O(C 1-4 alkyl); -C(=O)(C 1-4 alkyl); - C(=O)OH; -CON(R’)(R’’); -S(O) 1-2 (NR’R’’); -S(O) 1-2 (C 1-4 alkyl); cyano, and C 3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; R 6 is selected from the group consisting of H; C 1-6 alkyl optionally substituted with 1-3 independently selected R a ; -OH; C 1-4 alkoxy; C(=O)H; C(=O)(C 1-4 alkyl); C6-10 aryl optionally substituted with 1-4 independently selected C 1-4 alkyl; and heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C 1-4 alkyl; each occurrence of R a is independently selected from the group consisting of: – OH; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)O(C 1-4 alkyl); -C(=O)(C1- 4 alkyl); -C(=O)OH; -CON(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano, and C3- 6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; each occurrence of R b is independently selected from the group consisting of: C1- 10 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; –OH; oxo; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C1-10 alkyl); -C(=O)O(C1- 4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano; and –L 1 -L 2 -R h ; each occurrence of 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 ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl); - NR e R f ; –OH; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy; -NO2; -C(=O)(C1-10 alkyl); -C(=O)O(C1- 4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); and –L 1 -L 2 -R h ; R d is selected from the group consisting of: C 1-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; C 3-6 cycloalkyl or C 3-6 cycloalkenyl, each optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; -C(O)(C 1-4 alkyl); - C(O)O(C 1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C 1-4 alkyl); -OH; and C 1-4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; C 1-6 alkyl; C 1-6 haloalkyl; C3-6 cycloalkyl or C3-6 cycloalkenyl; -C(O)(C 1-4 alkyl); - C(O)O(C 1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C 1-4 alkyl); -OH; and C 1-4 alkoxy; or R e and R f together with the nitrogen atom to which each is attached forms a ring of 3-8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R e and R f ), which are each independently selected from the group consisting of N(R d ), NH, O, and S; -L 1 is a bond or C1-3 alkylene; -L 2 is –O-, -N(H)-, -S(O) 0-2 -, or a bond; R h is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl (in certain embodiments, it is provided that when R h is C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected C 1-4 alkyl, -L 1 is a bond, or –L 2 is – O-, -N(H)-, or -S-); ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1- 4 haloalkyl; ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C1- 4 haloalkyl; -L 3 is a bond or C1-3 alkylene; -L 4 is –O-, -N(H)-, -S(O) 0-2 -, or a bond; R i is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl (in certain embodiments, it is provided that when R i is C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected C 1-4 alkyl, -L 1 is a bond, or –L 2 is – O-, -N(H)-, or -S-); ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C1- 4 haloalkyl; ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C1- 4 haloalkyl; and each occurrence of R’ and R’’ is independently selected from the group consisting of: H, -OH, C 1-4 alkyl, C6-10 aryl optionally substituted with 1-2 substituents selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, and heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, -OH, NH2, NH(C 1-4 alkyl), N(C 1-4 alkyl)2, C 1-4 alkyl, and C 1-4 haloalkyl; or R’ and R’’ together with the nitrogen atom to which each is attached forms a ring of 3- 8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R’’), which are each independently selected from the group consisting of N(H), N(C 1-6 alkyl), O, and S. In some embodiments, it is provided that one or more of a), b), and c) apply: a) one or more of Z, Y 1 , Y 2 , Y 3 , and Y 4 in the ring below independently selected heteroatom; b) the ring that includes Z, Y 1 , Y 2 , Y 3 , and Y 4 is partially unsaturated; or c) Z is a bond; and further provided that the compound is other than: . Embodiments can include any one or more of the features delineated below and/or in the claims. The Variables Y 1 , Y 2 , Y 3 , Y 4 , and Z In some embodiments, aromatic. In some embodiments, Z is selected from the group consisting of CR 1 , N, and NR 2 . In certain embodiments, Z is CR 1 . In some embodiments, 1-2 of Y 1 , Y 2 , and Y 3 is independently N or NR 2 (e.g., N); and each of the remaining Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . In certain embodiments, one of Y 1 , Y 2 , and Y 3 is independently N or NR 2 ; and each of the remaining of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . In certain embodiments, one of Y 1 , Y 2 , and Y 3 is independently N; and each of the remaining of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . In certain embodiments, the moiety i wherein the asterisk denotes point of attachment to Y 4 . For the sole purpose of clarity and unless otherwise specificied, the statement “wherein the asterisk denotes point of attachment to Y 4 ” means “ ” is the direct bond linked to Y 4 . In another word, when the moiety is In certain embodiments, the moiety is , wherein the asterisk denotes point of attachment to Y 4 . In certain embodiments, the y , wherein the asterisk denotes point of attachment to Y 4 . In certain embodiments, the moiety is , wherein the asterisk denotes point of attachment to Y 4 . In certain embodiments, the moiety i wherein the asterisk denotes point of attachment to Y 4 . In certain embodiments, the moiety is , wherein the asterisk denotes point of attachment to Y 4 . In certain embodiments, the y , wherein the asterisk denotes point of attachment to Y 4 . In certain embodiments, the moiety is , wherein the asterisk denotes point of attachment to Y 4 . In some embodiments, Z is N. In some embodiments (e.g., when Z is N), each of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . In certain embodiments, the ), wherein the asterisk denotes point of attachment to Y 4 . In some embodiments, Y 4 is C. The Variables X 1 and X 2 In some embodiments, X 1 is NR 2 . In certain embodiments, X 1 is NH. In some embodiments, X 2 is CR 5 . In certain embodiments, X 2 is CH. In certain embodiments, X 1 is NR 2 ; and X 2 is CR 5 . In certain embodiments, X 1 is NH; and X 2 is CH. Non-Limiting Combinations of Y 1 , Y 2 , Y 3 , Y 4 , Z, X 1 , and X 2 In some embodiments, the compound is selected from a compound of the following formulae: In certain embodiments, the compound has formula (I-a): (Ia). In certain of these embodiments, the compound has formula (I-a1) or (I-a2): In certain embodiments, the compound has formula (Ia-3): the compound has Formula (I In certain embodiments, the compound has formula (Ib). In certain of these embodiments, the compound has formula (Ib-1): In certain embodiments, the compound has formula (Ic). In certain of these embodiments, the compound has formula (Ic-1): In certain embodiments, the compound has formula (I-d): In certain of these embodiments, the compound has formula (Id-1): In certain embodiments when the compound has formula (Id), the compound has formula (Id-2): (Id-2). In certain embodiments when the compound has formula (Id), the compound has formula (Id-3): (Id-3). The Variable R 1 In some embodiments, each occurrence of R 1 is independently selected from the group consisting of: H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; -S(O)1-2(C1- 4 alkyl); -S(O)(=NH)(C 1-4 alkyl); SF5; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy; -NO2; - C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’). In some embodiments, 0-3 (e.g., 0, 1, 2, or 3) occurrences of R 1 is other than H; and each of the remaining occurrences of R 1 is H. In some embodiments, each occurrence of R 1 is H. In some other embodiments, 1-2 occurrences of R 1 is other than H. In certain of these embodiments, one occurrence of R 1 is other than H. In one or more of the foregoing embodiments, one occurrence of R 1 is selected from the group consisting of: halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C2- 6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl); - S(O)1-2(NR’R’’); -NO2; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and - C(=O)N(R’)(R’’). In certain embodiments, one occurrence of R 1 is halo (e.g., F or Cl (e.g., F)). In certain embodiments, one occurrence of R 1 is –L 3 -L 4 -R i . In certain of these embodiments, L 3 is a bond; and/or L 4 is a bond. As a non-limiting example, one occurrence of R 1 is R i (e.g., R i is heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl; or R i is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl). In certain embodiments, one occurrence of R 1 is –L 3 -L 4 -R i ; and each remaining occurrences of R 1 is H. In certain of these embodiments, –L 3 is a bond; and/or –L 4 is a bond. In certain of the foregoing embodiments, –R i is selected from the group consisting of: ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy. In certain of these embodiments, –R i is selected from the group consisting of: ● heteroaryl of 5-6 ring atoms (e.g., pyrazolyl), wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-2 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and ● phenyl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy. In certain embodiments, one of R 1 is selected from the group consisting of: ● heteroaryl of 5-6 ring atoms (such as pyrazolyl), wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-2 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy ( ● phenyl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy (e.g., ). In certain of these embodiments, each remaining R 1 is H. The Variable R 2 In some embodiments, R 2 is H. In some embodiments, R 2 is selected from the group consisting of: heterocyclyl or heterocycloalkenyl of 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(O) 0-2 ; and heteroaryl of 5-10 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(O) 0-2 . In certain embodiments, R 2 is heterocyclyl or heterocycloalkenyl of 3-10 (e.g., 3, 4, 5, 6, 7, 8, 9, or 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(O) 0-2 . In certain of these embodiments, R 2 is heterocyclyl or heterocycloalkenyl of 4-6 (e.g., 4, 5, or 6) ring atoms, wherein 1-3 (e.g., 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 . As a non-limiting example, R 2 can be azetidinyl, pyrolindyl, piperazinyl, morpholinyl, or piperidinyl (e.g., R 2 can be piperidinyl such as piperidin-4-yl). In certain embodiments, R 2 is heteroaryl of 5-10 (e.g., 5, 6, 7, 8, 9, or 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(O) 0-2 . In certain of these embodiments, R 2 is heteroaryl of 5-10 (e.g., 5 or 6) ring atoms, wherein 1-3 (e.g., 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 . As a non-limiting example, R 2 can be pyridyl, pyrimidyl, or pyrazolyl (e.g., R 2 can be pyrazolyl such as pyrazol-4-yl). The Variable R 5 In some embodiments, R 5 is H or halo. In some embodiments, R 5 is H. The Variable R 6 In some embodiments, R 6 is H. In some embodiments, R 6 is C1-3 alkyl. The Variables Q and A In some embodiments, Q-A is defined according to (A). In certain of these embodiments, Q is NH. In certain other embodiments, Q is N(C1-3 alkyl) (e.g., NMe or NEt). In certain embodiments, A is -(Y A1 ) n -Y A2 . In certain of these embodiments, n is 0. In certain other embodiments, n is 1. In certain embodiments when n is 1, Y A1 is C 1-6 alkylene, which is optionally substituted with 1-4 R a . In certain of these embodiments, Y A1 is -CH2-, -CH2CH2-, -CH2CH2CH2-, - As non-limiting examples of the foregoing embodiments, Y A1 is -CH2- or - CH2CH2-. As a non-limiting example, Y A1 can be -CH2-. As another non-limiting example, Y A1 can be -CH2CH2-. As another non-limiting example, Y A1 can be . In certain embodiments, Y A1 is Y A3 -Y A4 -Y A5 . In certain of these embodiments, Y A3 is C2-3 alkylene; and/or Y A4 is –O-; and/or Y A5 is a bond. As a non-limiting example, Y A1 can be . In certain embodiments, Y A2 can be C6-10 aryl, which is optionally substituted with 1-3 R c . In certain embodiments, Y A2 is C6 aryl, which is optionally substituted with 1-3 R c . In certain embodiments, Y A2 is C6 aryl, which is substituted with 1-3 R c . In certain embodiments, Y A2 is phenyl substituted with 1-3 R c , wherein one R c is at the ring carbon para to the point of attachment to Y A1 . In certain embodiments, Y A2 is phenyl substituted with 1-3 R c , wherein 1-2 R c is at the ring carbons meta to the point of attachment to Y A1 . In certain embodiments, Y A2 is phenyl substituted with 1-3 R c , wherein 1-2 R c is at the ring carbons ortho to the point of attachment to Y A1 . In certain other embodiments, Y A2 is unsubstituted phenyl. In certain embodiments, Y A2 is C7-10 bicyclic aryl, which is optionally substituted with 1-3 R c (e.g., Y A2 is naphthyl ( indanyl tetrahydronapthyl, each of which is optionally substituted with 1- 3 R c ). In certain embodiments, Y A2 is heteroaryl of 5-14 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . In certain of the foregoing embodiments, Y A2 is heteroaryl of 6 ring atoms (e.g., pyridyl or pyrimidinyl (e.g., pyridyl)), wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl ring is optionally substituted with 1-3 independently selected R c . In certain of these embodiments, Y A2 is substituted with 1-3 independently selected R c ; and one occurrence of R c is at the ring carbon atom para to the point of attachment to Y A1 (e.g., In certain embodiments (when Y A2 is heteroaryl of 6 ring atoms (e.g., pyridyl or pyrimidinyl (e.g., pyridyl)), wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl ring is optionally substituted with 1-3 independently selected R c ), Y A2 is substituted with 1-3 independently selected R c ; and one occurrence of R c is at the ring carbon atom meta to the point of attachment to Y A1 . In certain embodiments, Y A2 is bicyclic or tricyclic heteroaryl of 7-14 (e.g., 9-12 (e.g., 9, 10, 11, or 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . In certain of these embodiments, Y A2 is bicyclic heteroaryl of 9-10 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . As a non-limiting example of the foregoing embodiments, Y A2 can be bicyclic heteroaryl of 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(O) 0-2 (e.g., , wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . In certain of the foregoing embodiments (wherein Y A2 is aryl or heteroaryl as described supra), each occurrence of 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 ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1- 2(C 1-4 alkyl); -NR e R f ; -C 1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C 1-4 alkyl); and –L 1 -L 2 -R h . In certain embodiments, one occurrence of R c is halo (e.g., F or Cl (e.g., Cl)). In certain embodiments, one occurrece of R c is C 2-6 alkynyl (e.g., ). In certain embodiments, one occurrence of R c is C 1-4 alkoxy or C 1-4 haloalkoxy (e.g., OCF3 or OCH2CHF2). In certain embodiments, one occurrence of R c is SF5. In certain embodiments, one occurrence of R c is S(O)2(C 1-4 haloalkyl) (e.g., S(O) 2 CF 3 ). In certain embodiments, one occurrence of R c is C 1-4 thiohaloalkoxy (e.g., SCF3). In certain embodiments, one occurrence of R c is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a . In certain embodiments, one occurrence of R c is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10). As a non-limiting example, one occurrence of R c can be ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl, iso-butyl, sec-butyl, tert-butyl), or octyl (e.g., n-octyl). In certain embodiments (when one occurrence of R c is is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a ), the occurrece of R c is C 1-10 alkyl which is substituted with 1-6 independently selected R a . In certain of these embodiments, each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy. As a non-limiting example, each occurrence of R a is halo (e.g., F). In certain embodiments (e.g., when one occurrece of R c is C1-10 alkyl which is substituted with 1-6 independently selected R a ), the occurrence of R c is CF3. In certain embodiments, one occurrence of R c is –L 1 -L 2 -R h . In certain of these embodiments, L 1 is a bond and/or L 2 is a bond; and/or L 2 is CH2. In certain embodiments (when one occurrence of R c is –L 1 -L 2 -R h ), R h is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl. In certain embodiments (when one occurrence of R c is –L 1 -L 2 -R h ), R h is C6 aryl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl (e.g., ). In certain embodiments (when one occurrence of R c is –L 1 -L 2 -R h ), R h is heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3- 10 (e.g., 5-6) ring atoms, wherein 1-3 (e.g,, 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl. As a non-limiting example of the foregoing embodiments, R h can another non-limiting example, R h can b . In certain embodiments (when one occurrence of R c is –L 1 -L 2 -R h ), R h is C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl. In certain of these embodiments, R h is C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl. As non-limiting examples, R h can be selected from the group consisting of: , In any one of more of the foregoing embodiments of R c , each of the remaining occurrences of R c is C 1-6 alkyl or halo. In certain embodiments, Y A2 is monocyclic C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with 1-4 R b . In certain embodiments, Y A2 is C 3 - 6 (e.g., C 3 , C 5 , or C 6 ) cycloalkyl or C 3 - 6 (e.g., C 3 , C5, or C6) cycloalkenyl, each of which is substituted with 1-4 (e.g., 1-2) R b (e.g., Y A2 is cyclopropyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with 1-2 R b ). As a non-limiting example of the foregoing embodiments, Y A2 is cyclohexyl which is optionally substituted with 1-2 R b . In certain embodiments (when Y A2 is cyclohexyl which is optionally substituted with 1-2 R b ), one occurrence of R b is at the ring carbon atom para to the point of attachment to Y A1 ; or one occurrence of R b is at the ring carbon atom meta to the point of attachment to Y A1 . In certain embodiments (when Y A2 is cyclohexyl which is optionally substituted with 1-2 R b ), two occurrences of R b are at the ring carbon atom para to the point of attachment to Y A1 ; or two occurrences of R b are at the ring carbon atom meta to the point of attachment to Y A1 . In certain other embodiments, Y A2 is unsubstituted cyclohexyl. In certain embodiments, Y A2 is cyclopropyl which is substituted with 1-2 R b . As a non-limiting example, Y A2 can be cyclopropyl substituted with –L 1 -L 2 -R h ( In certain embodiments, Y A2 is bicyclic, tricyclic, or polycyclic C7-20 cycloalkyl or C7-20 cycloalkenyl, each optionally substituted with 1-2 R b . In certain embodiments, Y A2 is bicyclic, tricyclic, or polycyclic C 7-12 cycloalkyl or or cycloalkenyl, each optionally substituted with 1-2 R b . In certain embodiments, Y A2 is bicyclic C7-8 cycloalkyl, optionally substituted with 1-2 R b . In certain embodiments, Y A2 is bicyclic, tricyclic, or polycyclic C9-12 cycloalkyl, optionally substituted with 1-2 R b . In certain embodiments, Y A2 is spirobicyclic C 7-12 cycloalkyl, optionally substituted with 1-2 R b (e.g., spiro[5.5]undecanyl (e.g., ) or spiro[2.5]octanyl (e.g., )). In certain of these embodiments, Y A2 is selected from the group consisting of: spiro[5.5]undecanyl (e.g., ), bicyclo[2.2.1]hept-2-enyl (e.g., ), bicyclo[2.2.1]heptanyl (e.g., ), spiro[2.5]octanyl (e.g., ), and adamantly (e.g., ). For example, Y A2 can be . As another example, Y A2 can be or . As a further example. Y A2 can be . In certain embodiments, Y A2 is heterocyclyl or heterocycloalkenyl of 3-16 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-3 independently selected R b (sometimes referred to herein as “each occurrence of R b substituent of Y A2 ” or “one occurrence of R b substituent of Y A2 ”). In certain embodiments, Y A2 is heterocyclyl or heterocycloalkenyl of 4-10 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-3 independently selected R b . In certain embodiments, Y A2 is heterocyclyl or heterocycloalkenyl of 5-7 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-3 independently selected R b . As non-limiting examples, Y A2 can be or . In certain embodiments, each occurrence of R b substituent of Y A2 is independently selected from the group consisting of: C1-10 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; -F; -Cl; -Br; cyano; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C1-10 alkyl); -C(=O)O(C 1-4 alkyl); -S(O)1-2(C 1-4 alkyl); oxo; cyano; and –L 1 -L 2 -R h . In certain embodiments, one occurrece of R b substituent of Y A2 is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a . In certain of these embodiments, one occurrence of R b substituent of Y A2 is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10). As a non-limiting example of the foregoing embodiments, one occurrence of R b substituent of Y A2 can be ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl; or sec-butyl; or tert-butyl; or iso-butyl), or octyl (e.g., n-octyl). In certain embodiments, one occurrece of R b substituent of Y A2 is C1-10 alkyl which is substituted with 1-6 independently selected R a . In certain of these embodiments, each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy. For example, one or more occurrences of R a can be an independently selected halo (which can be the same or different halo); e.g., fluro, and R b can be CF3 or -CF2CH3. In certain embodiments, one occurrence of R b substituent of Y A2 is –L 1 -L 2 -R h (e.g., -R h or –CH2-R h such as benzyl). In certain embodiments, one occurrence of R b substituent of Y A2 is C 1-4 alkoxy or C 1-4 haloalkoxy (e.g., ). In certain embodiments, one occurrence of R b is –F or –Cl (e.g., -F). In certain embodiments, Y A2 is ; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . In certain embodiments, Y A2 is ; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . In certain embodiments, Y A2 is ; one of Q 1 and Q 2 is N; the other one of Q 1 and Q 2 is CH; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . In certain embodiments, Y A2 is ; one of Q 1 , Q 2 , Q 3 , and Q 4 is N; each of the remaining of Q 1 , Q 2 , Q 3 , and Q 4 is CH; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . In certain embodiments (when Y A2 is , , , or ), R cA is selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl); - NR e R f ; -C 1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C 1-4 alkyl); and –L 1 - L 2 -R h . In certain embodiments (when Y A2 is , , , or ), R cA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl, iso-butyl, sec- butyl, tert-butyl), or octyl (e.g., n-octyl). In certain embodiments (when Y A2 is , , , or ), R cA is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy). In certain of these embodiments, R cA is C1-10 alkyl which is substituted with 1-6 independently selected halo (e.g., R cA is CF3). In certain embodiments (when Y A2 is , , , or ), R cA is C 2-6 alkynyl (e.g., ). In certain embodiments (when Y A2 is , , , or ), R cA is C 1-4 haloalkoxy (e.g., -OCF3 or ). In certain embodiments (when Y A2 is , , , or ), R cA is –L 1 -L 2 -R h . In certain of these embodiments, –L 1 is a bond. In certain embodiments (when R cA is –L 1 -L 2 -R h ), –L 2 is a bond. In certain embodiments (when R cA is –L 1 -L 2 -R h ), R h is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl, such as C6 aryl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl ( In certain embodiments (when R cA is –L 1 -L 2 -R h ), R h is heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-10 (e.g., 5-6) ring atoms, wherein 1-3 (e.g,, 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, such as . In certain embodiments (when R cA is –L 1 -L 2 -R h ), R h is C3-8 (e.g., C3-6) cycloalkyl or C3-8 (e.g., C3-6) cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl (e.g., R h is cyclohexyl). In certain embodiments (when Y A2 is , , In certain other embodiments, n1 is 1 or 2 (e.g., 1). In certain of these embodiments, each occurrence of R cB is independently halo or C1-3 alkyl (e.g., halo). In certain embodiments, Y A2 is ; n2 is 0, 1, or 2; and each of R bA and R bB is an independently selected R b . In certain embodiments, Y A2 is ; n2 is 0, 1, or 2; and each of R bA and R bB is an independently selected R b . In certain embodiments (when Y A2 is or ), R bA is C1- 10 alkyl which is optionally substituted with 1-6 independently selected R a . In certain embodiments (when Y A2 is or ), R bA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n- propyl), butyl (e.g., n-butyl; or sec-butyl; or tert-butyl; or iso-butyl), or octyl (e.g., n-octyl). In certain embodiments (when Y A2 is or ), R bA is C 1- 10 alkyl which is substituted with 1-6 independently selected R a (e.g., each R a is selected from the group consisting of halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy) (e.g., R bA is CF3 or -CF2CH3). In certain embodiments (when Y A2 is or ), R bA is –F or –Cl. In certain embodiments (when Y A2 is or ), R bA is –L 1 - L 2 -R h (e.g., -R h or –CH 2 -R h such as benzyl). In certain embodiments (when Y A2 is or ), R bA is C 1-4 alkoxy or C 1-4 haloalkoxy (e.g., ). In certain embodiments (when Y A2 is or ), n2 is 0. In certain other embodiments, n2 is 1 or 2. In certain of these embodiments, each occurrence R bB is selected from the group consisting of –F, -Cl, and C1-3 alkyl. Non-limiting examples of A include: Further non-limiting examples of A include:

In some embodiments, Q-A is as defined according to (B). In certain embodiments, E is a saturated or partially unsaturated ring of 3-16 ring atoms, wherein 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the carbon portion of the ring is optionally substituted with 1-4 independently selected R b . In certain embodiments, E is a ring of 5-8 ring atoms, wherein 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b (e.g., E is piperidinyl which is optionally substituted with 1-2 independently selected R b (e.g., E is , wherein R b is C 1-6 alkyl)). As a non-limiting example, E is . Non-Limiting Combinations In certain embodiments, the compound has the following formula: (I-1), wherein n1 is 0, 1, or 2; each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. In certain embodiments, the compound has the following formula: (I-2) , wherein n1 is 0, 1, or 2; each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. In certain embodiments, the compound has the following formula:

wherein one of Q 1 and Q 2 is N; the other one of Q 1 and Q 2 is CH; n1 is 0, 1, or 2; each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. In certain embodiments, the compound has the following formula: wherein one of Q 1 , Q 2 , Q 3 , and Q 4 is N; each of the remaining of Q 1 , Q 2 , Q 3 , Q 4 is CH; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. In certain embodiments, the compound has the following formula: wherein B 1 is selected from the group consisting of: (a) bicyclic or tricyclic heteroaryl of 7-14 (e.g., 9-12 (e.g., 9, 10, 11, or 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c ; and (b) C 7-10 bicyclic aryl, which is optionally substituted with 1-3 R c ; and R 7 is H or C 1-4 alkyl. In certain embodiments of Formula (I-5), B 1 is bicyclic or tricyclic heteroaryl of 9-10 (e.g., 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . As a non-limiting example of the foregoing embodiments, In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4), R cA is selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl); - NR e R f ; -C 1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C 1-4 alkyl); and –L 1 - L 2 -R h . In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4), R cA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl, iso-butyl, sec-butyl, tert-butyl), or octyl (e.g., n-octyl). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4), R cA is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy). In certain of these embodiments, R cA is C1-10 alkyl which is substituted with 1-6 independently selected halo (e.g., R cA is CF3). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4), R cA is C 1-4 alkoxy or C 1-4 haloalkoxy. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4), R cA is –L 1 -L 2 -R h . In certain of these embodiments, –L 1 is a bond. In certain embodiments (when R cA is –L 1 -L 2 -R h ), –L 2 is a bond. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4) (when R cA is –L 1 -L 2 -R h ), R h is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl, such as C6 aryl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl (e.g., In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4) (when R cA is –L 1 -L 2 -R h ), R h is heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-10 (e.g., 5-6) ring atoms, wherein 1-3 (e.g,, 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, such as . In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4) (when R cA is –L 1 -L 2 -R h ), R h is C3-8 (e.g., C3-6) cycloalkyl or C3-8 (e.g., C3-6) cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl (e.g., R h is cyclohexyl). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), and (I- 4), n1 is 0. In certain other embodiments, n1 is 1 or 2 (e.g., 1). In certain of these embodiments, each occurrence of R cB is independently halo or C1-3 alkyl (e.g., halo). In certain embodiments, the compound has the following formula: wherein n2 is 0, 1, or 2; each of R bA and R bB is an independently selected R b ; and R 7 is H or C 1-4 alkyl. In certain embodiments, the compound has the following formula: wherein n2 is 0, 1, or 2; each of R bA and R bB is an independently selected R b ; and R 7 is H or C 1-4 alkyl. In certain embodiments, the compound has the following formula:

wherein B 2 is: bicyclic, tricyclic, or polycyclic C7-20 cycloalkyl or C7-20 cycloalkenyl, each optionally substituted with 1-2 R b ; and R 7 is H or C 1-4 alkyl. In certain of these embodiments, B 2 is: bicyclic, tricyclic, or polycyclic C7-12 cycloalkyl or C7-12 cycloalkenyl, each optionally substituted with 1-2 R b . In certain embodiments, B 2 is bicyclic C 7-8 cycloalkyl optionally substituted with 1-2 R b . In certain embodiments, B 2 is bicyclic C9-12 cycloalkyl optionally substituted with 1-2 R b . In certain embodiments, B 2 is spirobicyclic C7-12 cycloalkyl optionally substituted with 1-2 R b . For example, B 2 can be spiro[5.5]undecanyl (e.g., or spiro[2.5]octanyl (e In certain embodiments of Formula (I-8), B 2 is selected from the group consisting of: spiro[5.5]undecanyl ( bicyclo[2.2.1]hept-2-enyl (e.g., bicyclo[2.2.1]heptanyl (e.g., spiro[2.5]octanyl (e.g., and adamantly In certain embodiments of any one or more of Formulae (I-6) and (I-7), R bA is C1- 10 alkyl which is optionally substituted with 1-6 independently selected R a . In certain of these embodiments, R bA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl; or sec-butyl; or tert-butyl; or iso-butyl), or octyl (e.g., n-octyl). In certain other embodiments, R bA is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., each R a is selected from the group consisting of halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy) (e.g., R bA is CF3). In certain embodiments of any one or more of Formulae (I-6) and (I-7), R bA is –F or –Cl. In certain embodiments of any one or more of Formulae (I-6) and (I-7), R bA is – L 1 -L 2 -R h (e.g., -R h or –CH 2 -R h such as benzyl). In certain embodiments of any one or more of Formulae (I-6) and (I-7), R bA is C1- 4 alkoxy or C 1-4 haloalkoxy ( In certain embodiments of any one or more of Formulae (I-6) and (I-7), n2 is 0. In certain other embodiments, n2 is 1 or 2. In certain of these embodiments, each occurrence R bB is selected from the group consisting of –F, -Cl, and C1-3 alkyl. In some embodiments, the compound has the following formula:

wherein B 2 is: (a) C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with 1-2 R b , (b) phenyl, which is optionally substituted with 1-2 R c ; (c) heteroaryl of 5-6 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-2 independently selected R c ; and R 7 is H or C 1-4 alkyl. In certain embodiments of Formula (I-10), B 2 is C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with 1-2 R b . As a non-limiting example, B 2 can be C5-7 cycloalkyl which is unsubstituted, such as unsubstituted cyclohexyl. In certain embodiments of Formula (I-10), B 2 is phenyl, which is optionally substituted with 1-2 R c . As a non-limiting example, B 2 can be unsubstituted phenyl. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), and (I-8), n is 0. In certain of these embodiments, Y A1 is -CH2-, -CH2CH2-, -CH2CH2CH2-, - CH(CF3)-, -CH2CH(OH)-, , , , , or . In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), and (I-8), n is 1. In certain of these embodiments, Y A1 is C 1-6 alkylene, which is optionally substituted with 1-2 R a . In certain of these embodiments, Y A1 is - CH2-, -CH2CH2-, -CH2CH2CH2-, -CH(CF3)-, -CH2CH(OH)-, (e.g., CH2), , or (e.g., Y A1 is -CH2- or -CH2CH2-). As a non-limiting example, Y A1 can be -CH2-. As another non-limiting example, Y A1 can be -CH2CH2-. As another non-limitign example, Y A1 can be . As another non-limitign example, Y A1 can be . In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), and (I-10), Y A1 is Y A3 -Y A4 -Y A5 . In certain of these embodiments, Y A3 is C2-3 alkylene; and/or Y A4 can be –O-; and/or Y A5 is a bond. As a non-limiting example, Y A1 can be or . In certain embodiments, the compound has the following formula:

E is a ring of 3-16 ring atoms, wherein 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b . In certain embodiments of Formula (I-9), E is a ring of 5-8 ring atoms, wherein 0- 3 additional ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b (e.g., E is piperidinyl which is optionally substituted with 1-2 independently selected R b (e.g., E is , wherein R b is C 1-6 alkyl)). As a non-limiting example, E is In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), ( In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), (I-9), and (I-10), the moiety is (e.g., ). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), (I-9), and (I-10), the moiety is (e.g., ). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), (I-9), and (I-10), the moiety is (e.g., or ). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), (I-9), and (I-10), R 2 is H. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), R 2 is heterocyclyl or heterocycloalkenyl of 3-10 (e.g., 3, 4, 5, 6, 7, 8, 9, or 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(O) 0-2 . In certain of these embodiments, R 2 is heterocyclyl or heterocycloalkenyl of 4-6 (e.g., 4, 5, or 6) ring atoms, wherein 1-3 (e.g., 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 . As a non-limiting example, R 2 can be azetidinyl, pyrolindyl, piperazinyl, morpholinyl, or piperidinyl (e.g., R 2 can be piperidinyl such as piperidin-4-yl). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), R 2 is heteroaryl of 5-10 (e.g., 5, 6, 7, 8, 9, or 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(O) 0-2 . In certain of these embodiments, R 2 is heteroaryl of 5-10 (e.g., 5 or 6) ring atoms, wherein 1-3 (e.g., 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 . As a non-limiting example, R 2 can be pyridyl, pyrimidyl, or pyrazolyl (e.g., R 2 can be pyrazolyl such as pyrazol-4-yl). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), (I-9), and (I-10), R 5 is H. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), and (I-10), R 7 is H. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6), (I-7), (I-8), (I-9), and (I-10), each R 1 is independently selected from the group consisting of: H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; -S(O)1- 2(C 1-4 alkyl); -S(O)(=NH)(C 1-4 alkyl); SF5; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy; -NO2; - C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’). In certain of these embodiments, each occurrence of R 1 is H. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), 1-2 occurrences of R 1 is other than H. In certain of these embodiments, one occurrence of R 1 is halo (e.g., F). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), one occurrence of R 1 is –L 3 -L 4 -R i . In certain of these embodiments, L 3 is a bond; and/or L 4 is a bond. As a non-limiting example, one occurrence of R 1 is R i (e.g., R i is heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl; or R i is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl). In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), one occurrence of R 1 is –L 3 -L 4 -R i ; and each remaining R 1 is H. In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), one occurrence of R 1 is selected from the group consisting of: ● heteroaryl of 5-6 ring atoms (such as pyrazolyl), wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-2 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy ● phenyl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy (e.g., In certain embodiments of any one or more of Formulae (I-1), (I-2), (I-3), (I-4), (I- 5), (I-6), (I-7), (I-8), (I-9), and (I-10), R 6 is H. Non-Limiting Exemplary Compounds In certain embodiments, the compound is selected from the group consisting of the compounds delineated in Table C1, or a pharmaceutically acceptable salt thereof.

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-α-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 α-, ●, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl-β-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, intracisternal, 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 non- sensitizing. 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 0.001 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 150 mg/Kg; from about 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 50 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 5 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from about 0.01 mg/Kg to about 0.5 mg/Kg; from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0.1 mg/Kg to about 200 mg/Kg; from about 0.1 mg/Kg to about 150 mg/Kg; from about 0.1 mg/Kg to about 100 mg/Kg; from about 0.1 mg/Kg to about 50 mg/Kg; from about 0.1 mg/Kg to about 10 mg/Kg; from about 0.1 mg/Kg to about 5 mg/Kg; from about 0.1 mg/Kg to about 1 mg/Kg; from about 0.1 mg/Kg to about 0.5 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, 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. 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 cancer 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 Kinsbourne 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; Sjögren'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-Goutières 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 Gram- negative 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-β (TGFβ), 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–4‑1BB ligand, OX40–OX40 ligand, GITR, GITR ligand – GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25–TL1A, CD40L, CD40–CD40 ligand, HVEM–LIGHT–LTA, HVEM, HVEM – BTLA, HVEM – CD160, 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). 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 anti- metabolite. 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. Anti- metabolites 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 anti- metabolite 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, semisynthetic 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- β), vasculostatin, vasostatin (calreticulin fragment) and the like. In certain embodiments, the additional chemotherapeutic agent is selected from abiraterone acetate, altretamine, anhydrovinblastine, auristatin, 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-1-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-Goutières 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 biologics (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 anti- inflammatory 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 biologics (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, obinutuzumab, vobarilizumab, lulizumab, atacicept, PF-06823859, and lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-α-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 biologics (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, vobarilizumab, lulizumab, atacicept, PF-06823859, lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-α-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-Goutières 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, fingolimod, firategrast (SB-683699) (formerly T-0047), GED0301, GLPG0634, GLPG0974, guselkumab, golimumab, GSK1399686, HMPL-004 (Andrographis paniculata extract), IMU-838, infliximab, Interleukin 2 (IL-2), Janus kinase (JAK) inhibitors, laquinimod, masitinib (AB1010), 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, alpha1-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-β-1a, IFN-β-1b), cladribine, corticosteroids (e.g., methylprednisolone), daclizumab, dimethyl fumarate (Tecfidera®), fingolimod (Gilenya®), fluoxetine, glatiramer acetate (Copaxone®), hydroxychloroquine, ibudilast, idebenone, laquinimod, lipoic acid, losartan, masitinib, MD1003 (biotin), mitoxantrone, montelukast, natalizumab (Tysabri®), NeuroVax TM , ocrelizumab, ofatumumab, pioglitazone, and RPC1063. Non-limiting examples of additional therapeutic agents and/or regimens for treating graft-vs-host disease include abatacept, alemtuzumab, alpha1-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 (Vectical®)), anthralin (e.g., Dritho-scalp® and Dritho-crème®), 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®), Apo805K1, baricitinib, FP187, KD025, prurisol, VTP-43742, XP23829, ZPL-389, CF101 (piclidenoson), LAS41008, VPD-737 (serlopitant), upadacitinib (ABT-494), aprmilast, tofacitibin, cyclosporine (Neoral®, Sandimmune®, Gengraf®), biologics (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®) biologics (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), biologics (e.g., infliximab (Remicade®), adalimumab (Humira®), etanercept (Enbrel®), golimumab (Simponi®), certolizumab (Cimzia®), rituximab (Rituxan®), abatacept (Orencia®), basiliximab (Simulect®), anakinra (Kineret®), canakinumab (Ilaris®), 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. Non-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 β-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, non- limiting 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 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 β-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, 1,8-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, 1 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. The following abbreviations have the indicated meanings:

LCMS analysis condition Method AA Instrument: Agilent LCMS system equipped with DAD and ELSD detector Ion mode: Positive Column: Waters X-Bridge C18, 50*2.1 mm*5 μm or equivalent Mobile Phase: A: H 2 O (0.04% TFA); B: CH 3 CN (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 AB Instrument: Agilent LCMS system equipped with DAD and ELSD detector Ion mode: Positive Column: Waters X-Bridge ShieldRP18, 50*2.1 mm*5 μm or equivalent Mobile Phase:A: 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 LCMS Method BA: Shim-pack XR-ODS, 50 *3mm, 3.0 µL injection, 1.2 mL/min flowrate, 90- 900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.05% TFA and Mobile Phase B (MPB): Acetonitrile/0.05% TFA. Elution 5% MPB to 100% in 2.00 min, hold at 100% MPB for 0.7 min, 100% MPB to 5% in 0.05 min, then equilibration to 5% MPB for 0.25 min. LCMS Method BB: EVO C18, 50 *3mm, 2.0 µL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.6 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min. LCMS Method BC: XBridge Shield RP18, 50 *4.6mm, 3.0 µL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.04% NH3.H2O and Mobile Phase B (MPB): Acetonitrile. Elution 40% MPB to 70% in 2.80 min, upto 95% in 0.20min, hold at 95% MPB for 0.5 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min. LCMS Method BD: Titank C18, 50 *3mm, 2.0 µL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 40% MPB to 70% in 2.80 min, upto 95% in 0.20min, hold at 95% MPB for 0.5 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min. LCMS Method BE: XBridge BEH C18 , 50 *3mm, 0.7 µL injection, 1.2 mL/min flowrate, 30- 2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5mMNH4CO3 and Mobile Phase B (MPB): Acetonitrile. Elution 5% MPB to 95% in 1.29 min, hold at 95% MPB for 0.90 min, 95% MPB to 5% in 0.05 min, then equilibration to 5% MPB for 0.25 min. LCMS Method CA: Kinetex EVO C18100A, 30*3mm, 0.5 µL 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 2.0 min, hold at 95% MPB for 0.3 min, 95% MPB to 10% in 0.1 min. LCMS Method CB: Xselect CSH C18, 50*3mm, 1.0 µL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.1% FA and Mobile Phase B (MPB): Acetonitrile/0.1% FA. Elution 5% MPB to 100% in 2.00 min, hold at 100% MPB for 0.7 min, 100% MPB to 5% in 0.05 min, then equilibration to 5% MPB for 0.15 min. LCMS Method CC: XBridge Shield RP18, 50*4.6mm, 0.5 µL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.04% NH4OH and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.79 min, 95% MPB to 10% in 0.06 min, then equilibration to 10% MPB for 0.15 min. LCMS Method CD: Kinetex 2.6um EVO C18 100A, 50*3mm, 0.6 µL injection, 1.2 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 1.20 min, hold at 95% MPB for 0.50 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.10 min. LCMS Method CE: EVO C18, 50*3mm, 0.1 µL injection, 1.2 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.60 min, 95% MPB to 10% in 0.15 min, then equilibration to 10% MPB for 0.25 min. LCMS Method CF: kinetex 2.6um EVO, 50*3mm, 0.5 µL injection, 1.2 mL/min flowrate, 30- 2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.70 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min. LCMS Method CG: Titank C18, 50*3mm, 0.5 µL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 1.80 min, hold at 95% MPB for 0.80 min, 95% MPB to 10% in 0.15 min, then equilibration to 10% MPB for 0.25 min. LCMS Method CH: Shim-pack Scepter C18, 30*3mm, 0.5 µL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.04% NH4OH and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.60 min, 95% MPB to 10% in 0.20 min, then equilibration to 10% MPB for 0.20 min. NMR was recorded on BRUKER NMR 300.03 Mz, DUL-C-H, ULTRASHIELD TM 300, AVANCE II 300 B-ACS TM 120 or BRUKER NMR 400.13 Mz, BBFO, ULTRASHIELD TM 400, AVANCE III 400, B-ACS TM 120. Prep. HPLC condition Instrument: 1. GILSON 281 and Shimadzu LCMS 2010A 2. GILSON 215 and Shimadzu LC-20AP 3. GILSON 215 Mobile phase: A: NH 4 OH/H 2 O = 0.05% v/v; B: ACN A: FA/H 2 O = 0.225% v/v; B: ACN Column Xtimate C18150*25mm*5µm 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 Examples Synthesis of Intermediates Intermediate 1 5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-amine Scheme 1 Step 1: 5-fluoro-3-nitro-1H-pyrrolo[2,3-b]pyridine 5-Fluoro-1H-pyrrolo[2,3-b]pyridine (5.0 g, 36.8 mmol, 1.0 equiv.) was dissolved in conc. H2SO4 (25 mL) and the reaction mixture was cooled to 0 °C. KNO3 (4.1 g, 40.5 mmol, 1.1 equiv.) was added portionwise over 3 minutes, maintaining the reaction mixture at 0 °C. The reaction mixture was stirred at 0 °C for 2 hours, then the solution was adjusted to pH 8.0 by the dropwise addition of aqueous 1 M NaOH. The resulting solid was collected by filtration and washed with water. 5-Fluoro-3-nitro-1H-pyrrolo[2,3-b]pyridine (3.6 g, 17.9 mmol) was isolated as a gray solid and used without additional purification. MS-ESI, 182.1 [M+H + ]. Step 2: 5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-amine 5-Fluoro-3-nitro-1H-pyrrolo[2,3-b]pyridine (3.6 g, 19.9 mmol, 1.0 equiv.) was dissolved in 40% HBr / H2O (40 mL). SnCl2 (18.9 g, 99.4 mmol, 5.0 equiv.) was added in one portion,a nd then the reaction mixture was heated to 70 °C for 30 minutes. The resulting mixture was cooled to ambient temperature. The solution was adjusted to pH 8.0 by the dropwise addition of aqueous 1M NaOH solution. The mixture was extracted with DCM, and the organic layers were combined and concentrated in vacuo to give 5-fluoro-1H- pyrrolo[2,3-b]pyridin-3-amine (2.0 g, 11.7 mmol) as a yellow solid that was used without additional purification. MS-ESI, 152.2 [M+H + ]. Intermediate 2 Step 1: 3-nitro-1H-pyrrolo[3,2-c]pyridine 1H-pyrrolo[3,2-c]pyridine (10.0 g, 84.7 mmol, 1.0 equiv.) was dissolved in conc. H2SO4 (50 mL) and cooled to 0 °C. Then KNO3 (9.4 g, 93.1 mmol, 1.1 equiv.) was added portionwise over 3 minutes, maintaining the reaction mixture at 0 °C. The reaction was then stirred for another 16 hours at 0 °C, after which the reaction mixture was adjusted to pH 8 by the dropwise addition of saturated aqueous Na2CO3. The resulting solid was collected by filtration and washed with water to give 3-nitro-1H-pyrrolo[3,2-c]pyridine (5.1 g, 31.3 mmol) as a yellow solid that was used without additional purification. MS-ESI, 164.1 [M+H + ]. Step 2: 1H-pyrrolo[3,2-c]pyridin-3-amine 3-nitro-1H-pyrrolo[3,2-c]pyridine (5.1 g, 31.3 mmol 1.0 equiv.) was dissolved in HCl (6 M, 166.7 mL, 32.0 equiv.). SnCl2•2H2O (35.3 g, 156.3 mmol, 5.0 equiv.) was added in one portion and the reaction mixture was stirred at 25 °C for 16 hours. The resulting mixture was adjusted to pH 8 by the dropwise addition of saturated aqueous Na2CO3. The mixture was extracted with EtOAc, and the organic layers were combined and concentrated in vacuo to give 1H-pyrrolo[3,2-c]pyridin-3-amine (0.9 g, 6.8 mmol) as a black solid that was used without additional purification. MS-ESI, 134.1 [M+H + ]. Intermediate 3 6-cyclohexylpyridin-3-amine Scheme 3 Step 1: 6-(cyclohex-1-en-1-yl)pyridin-3-amine To a mixture of 6-bromopyridin-3-amine (10.0 g, 57.8 mmol, 1.0 equiv.) and cyclohexen-1-ylboronic acid (8.7 g, 69.4 mmol, 1.2 equiv.) in dioxane (200 mL) and H2O (58 mL) was added Cs2CO3 (37.7 g, 115.6 mmol, 2.0 equiv.) and Pd(dppf)Cl2•DCM (2.4 g, 2.9 mmol, 0.05 equiv.) under an atmosphere of N2. The mixture was stirred at 100 °C for 16 hours. The reaction mixture was concentrated in vacuo, and the resulting residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0~50% EtOAc/Petroleum ether gradient) to give 6-(cyclohex-1-en-1-yl)pyridin-3-amine (9.0 g, 51.7 mmol) as a brown oil. MS-ESI, 175.2 [M+H + ]. Step 2: 6-cyclohexylpyridin-3-amine To a mixture of 6-(cyclohexen-1-yl)pyridin-3-amine (3.0 g, 17.2 mmol, 1.0 equiv.) in MeOH (40 mL) under an atmosphere of N2 was added Pd/C (3.0 g, 10% wt/wt%, 0.28 mmol, 0.02 equiv.) in one portion. The suspension evacuated under vacuum and backfilled with H2(g) 3 times. The mixture was stirred at 25 °C for 16 hours under an atmosphere of hydrogen (balloon). The reaction mixture was filtered through a pad of Celite and concentrated in vacuo to give 6-cyclohexylpyridin-3-amine (2.1 g, 11.9 mmol) as a white solid. MS-ESI, 176.8 [M+H + ]. Intermediate 4: 2-oxo-2-((3-(trifluoromethyl)phenyl)amino)acetic acid Scheme 4 Step 1: ethyl 2-oxo-2-((3-(trifluoromethyl)phenyl)amino)acetate To a mixture of 3-(trifluoromethyl)aniline (3.6 g, 23.6 mmol, 1.0 equiv.) in THF (100 mL) at 0 °C was added a solution of ethyl 2-chloro-2-oxoacetate (3.6 g, 26.0 mmol, 1.1 equiv.) in THF (30 mL) over 5 minutes, maintaining the reaction mixture at 0 °C. Then TEA (8.2 mL, 59.0 mmol, 2.5 equiv.) was added to the reaction mixture. The mixture was stirred at 30 °C for 3 hours. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 2-oxo-2-((3-(trifluoromethyl)phenyl)amino)acetate (5.2 g, 19.8 mmol) as a yellow oil that was used without additional purification. Step 2: 2-oxo-2-((3-(trifluoromethyl)phenyl)amino)acetic acid To a mixture of ethyl 2-oxo-2-((3-(trifluoromethyl)phenyl)amino)acetate (5.2 g, 19.8 mmol, 1.0 equiv.) in MeOH (80 mL) was added aqueous NaOH (2 M, 15 mL, 30 mmol, 1.5 equiv.). The mixture was stirred at 30 °C for 2 hours. The reaction mixture concentrated under reduced pressure to give a residue. Then H2O (30 mL) was added and the mixture was adjusted to pH 4 by the dropwise addition of 2 M HCl. The resulting solid was collected by filtration and washed with water to give 2-oxo-2-((3- (trifluoromethyl)phenyl)amino)acetic acid (4.8 g, 20.6 mmol) as a light yellow solid. MS- ESI, 234.1 [M+H + ]. Table I-1. The intermediates in Table I-1 were prepared using the procedure described in Scheme 4, starting from the appropriate amine:

Intermediate 16: 2-((2-methyl-3-(trifluoromethyl)phenyl)amino)-2-oxoacetic acid Scheme 2 Step 1: ethyl 2-((2-methyl-3-(trifluoromethyl)phenyl)amino)-2-oxoacetate 2-methyl-3-(trifluoromethyl)aniline (2.0 g, 11.4 mmol, 1.0 equiv.) was dissolved in THF (30 mL) at 0 °C. To this solution was added a solution of ethyl 2-chloro-2-oxoacetate (1.7 g, 12.5 mmol, 1.1 equiv.) in THF (20 mL) over 5 minutes, maintaining the reaction mixture at 0 °C. Then TEA (4.1 mL, 28.5 mmol, 2.5 equiv.) was added, and the reaction mixture was stirred at 30 °C for 3 hours. The reaction mixture was filtered and concentrated in vacuo to give ethyl 2-((2-methyl-3-(trifluoromethyl)phenyl)amino)-2- oxoacetate (2.8 g, 10.3 mmol) as a yellow oil that was used without additional purification. Step 2: 2-((2-methyl-3-(trifluoromethyl)phenyl)amino)-2-oxoacetic acid Ethyl 2-((2-methyl-3-(trifluoromethyl)phenyl)amino)-2-oxoacetate (2.8 g, 10.3 mmol, 1.0 equiv.) was dissolved in MeOH (50 mL) was added aqueous NaOH (2 M, 7.9 mL, 15.8 mmol, 1.5 equiv.). The mixture was stirred at 30 °C for 2 hours. The reaction mixture concentrated in vacuo to give a residue. Then H2O (30 mL) was added and the mixture was adjusted to pH 4 by the dropwise addition of 2 M HCl. The resulting solid was collected by filtration and washed with water to give 2-((2-methyl-3- (trifluoromethyl)phenyl)amino)-2-oxoacetic acid (2.2 g, 8.9 mmol) as a light yellow solid. MS-ESI, 248.1 [M+H + ]. Table I-2. The compounds in Table I-2 were prepared using the above procedure. Synthesis of intermediate B1 (5-methyl-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride) Step 1: 5-methyl-1H-pyrrolo[3,2-b]pyridine-3-carbonyl azide 5-Methyl-1H-pyrrolo[3,2-b]pyridine-3-carboxylic acid (500.0 mg, 3.0 mmol, 1.0 equiv.) was dissolved in THF (10 mL), then TEA (0.8 mL, 6.0 mmol, 2.0 equiv.) and DPPA (937.0 mg, 3.0 mmol, 1.2 equiv.) were added. The reaction mixture was stirred for 16 hours at ambient temperature and then quenched by the addition of water. The precipitated solids were collected by filtration and washed with water to give 5-methyl-1H-pyrrolo[3,2- b]pyridine-3-carbonyl azide (310.2 mg) as a pale yellow solid. LCMS Method CA: [M+H] + = 202. Step 2: tert-butyl N-[5-methyl-1H-pyrrolo[3,2-b]pyridin-3-yl]carbamate 5-Methyl-1H-pyrrolo[3,2-b]pyridine-3-carbonyl azide (100.0 mg, 0.5 mmol, 1.0 equiv.) was dissolved in t-BuOH (10 mL). The resulting solution was heated to 90 °C for 16 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl N-[5-methyl-1H-pyrrolo[3,2-b]pyridin-3- yl]carbamate (57.3 mg) as a yellow solid. LCMS Method CA: [M+H] + = 248. Step 3: 5-methyl-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride tert-Butyl N-[5-methyl-1H-pyrrolo[3,2-b]pyridin-3-yl]carbamate (238.0 mg, 1.0 mmol, 1.0 equiv.) was dissolved in HCl/1,4-dioxane (4 N, 10 mL). The reaction mixture was stirred for 2 hours at ambient temperature and concentrated under vacuum to give 5- methyl-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride (152.5 mg) as a pale brown solid that was used without additional purification. LCMS Method CA: [M+H] + = 148. Following intermediates were prepared using the method described for Intermediate B1. Synthesis of intermediate B5 (5-(1-isopropyl-1H-pyrazol-4-yl)-1H-pyrrolo[3,2- b]pyridin-3-amine hydrochloride) Compound 4 was prepared using the same methods described for Intermediate B1, Steps 1-2. LCMS: Method CC, [M+H] + = 312. Step 1: isopropyl N-[5-(1-isopropylpyrazol-4-yl)-1H-pyrrolo[3,2-b]pyridin-3- yl]carbamate tert-Butyl N-[5-bromo-1H-pyrrolo[3,2-b]pyridin-3-yl]carbamate (600.0 mg, 1.9 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (15 mL) and water (1.5 mL). Then 1- isopropyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyr azole (453.8 mg, 1.9 mmol, 1.0 equiv.), Cs2CO3 (1.2 g, 3.8 mmol, 2.0 equiv.) and Pd(dppf)Cl2 (140.6 mg, 0.2 mmol, 0.1 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was heated to 100 °C for 4 hours under nitrogen, then cooled to ambient temperature 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 isopropyl N-[5-(1- isopropylpyrazol-4-yl)-1H-pyrrolo[3,2-b]pyridin-3-yl]carbama te (385.2 mg) as a white solid. LCMS Method CA:[M+1] + = 342. Step 2: 5-(1-ethylpyrazol-4-yl)-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride tert-Butyl N-[5-(1-isopropylpyrazol-4-yl)-1H-pyrrolo[3,2-b]pyridin-3-yl ]carbamate (350.0 mg, 1.0 mmol, 1.0 equiv.) was dissolved in HCl/1,4-dioxane (4 N, 5 mL). The reaction mixture was stirred for 1 hour at ambient temperature then concentrated under vacuum to give 5-(1-ethylpyrazol-4-yl)-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride (311.5 mg) as a white solid that was used without further purification. LCMS Method CD: [M+1] + = 242. Synthesis of intermediate B6 (5-methoxy-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride) Step 1: 5-methoxy-3-nitro-1H-pyrrolo[3,2-b]pyridine 5-Methoxy-1H-pyrrolo[3,2-b]pyridine (1.0 g, 6.7 mmol, 1.0 equiv.) was dissolved in concentrated H2SO4 (15 mL) and cooled to 0 °C, then HNO3 (0.3 mL, 6.7 mmol, 1.0 equiv.) was added dropwise, maintaining the solution at 0 °C. The resulting solution was stirred for 2 hours at 0 °C and then the solution was poured into ice-water. The resulting mixture was adjusted to pH 8 with saturated aqueous Na2CO3. The precipitated solids were collected by filtration, washed with water and dried to give 5-methoxy-3-nitro-1H- pyrrolo[3,2-b]pyridine (810.0 mg) as a brown solid. LCMS Method CB: [M-H]- = 192. Step 2: tert-butyl N-[5-methoxy-1H-pyrrolo[3,2-b]pyridin-3-yl]carbamate 5-Methoxy-3-nitro-1H-pyrrolo[3,2-b]pyridine (200.0 mg, 1.0 mmol, 1.0 equiv.) was dissolved in MeOH (50 mL), then Boc2O (225.9 mg, 1.0 mmol, 1.0 equiv.) and Pd/C (10% wt, 20 mg) were added under an atmosphere of nitrogen. The mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred overnight at ambient temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl N-[5-methoxy- 1H-pyrrolo[3,2-b]pyridin-3-yl]carbamate (205.5 mg) as a yellow solid. LCMS Method CC: [M+H] + = 264. Step 3: 5-methoxy-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride The title compound was prepared using the methods described for Intermediate B1, Step 3. LCMS: Method CC, [M+H] + = 164. The following intermediates were prepared using the method described for Intermediate B6. Synthesis of intermediate B12 (8-(2-methylphenyl)-8-azabicyclo[3.2.1]octan-3-amine hydrochloride) Step 1: tert-butyl N-[8-(2-methylphenyl)-8-azabicyclo[3.2.1]octan-3-yl]carbamat e 2-methylphenylboronic acid (500.0 mg, 3.7 mmol, 1.0 equiv.) was dissolved in DCM (15 mL), then tert-butyl N-[8-azabicyclo[3.2.1]octan-3-yl]carbamate (832.3 mg, 3.7 mmol, 1.0 equiv.), Cu(OAc)2 (133.6 mg, 0.7 mmol, 0.2 equiv.) and TEA (1.0 mL, 7.4 mmol, 2.0 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was stirred for 16 hours at ambient temperature then quenched by the addition of water. The resulting mixture 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 (1:5) to give tert-butyl N-[8-(2- methylphenyl)-8-azabicyclo[3.2.1]octan-3-yl]carbamate (82.4 mg) as a colorless oil. LCMS Method CA: [M+H] + = 317. Step 2: 8-(2-methylphenyl)-8-azabicyclo[3.2.1]octan-3-amine hydrochloride The title compound was prepared using the methods described for Intermediate B1, Step 3. LCMS: Method CC: [M+H] + = 217. Synthesis of intermediate B13 ((3-methyl-4-(trifluoromethyl)phenyl)methanamine hydrochloride) Step 1: 4-methyl-3-(trifluoromethyl)benzamide 4-Methyl-3-(trifluoromethyl)benzoic acid (5.0 g, 24.5 mmol, 1.0 equiv.) was dissolved in DCM (100 mL) and cooled to 0 °C, then oxalyl dichloride (4.7 mL, 49.0 mmol, 2.0 equiv.) and DMF (0.1 mL, 1.3 mmol, 0.05 equiv.) were added. The resulting solution was stirred for 1 hour at ambient temperature and concentrated under vacuum. The residue was dissolved in THF (10 mL), then a solution of NH3(g) in THF (0.5 M, 50 mL) was added. The resulting mixture was stirred for 1 hour at ambient temperature and concentrated under vacuum. The residue was diluted with water, then extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to give 4-methyl-3-(trifluoromethyl)benzamide (5.1 g) as a white solid. LCMS Method CA: [M-H]- = 202. Step 2: (3-methyl-4-(trifluoromethyl)phenyl)methanamine hydrochloride 4-Methyl-3-(trifluoromethyl)benzamide (5.1 g, 24.6 mmol, 1.0 equiv) was dissolved in THF (50 mL) and cooled to 0 °C, then BH3-THF (1 M, 492.2 mL, 492.2 mmol, 20.0 equiv.) was added. The reaction mixture was heated to 55 °C for 1 hour, then cooled to 0 °C and quenched by the addition of MeOH. The resulting mixture was concentrated under vacuum and the residue was diluted with water. The solution was adjusted to pH 3 with aqueous HCl (2 M). The resulting solution was washed with ethyl acetate and the aqueous layer was concentrated under vacuum to give 1-[4-methyl-3- (trifluoromethyl)phenyl]methanamine hydrochloride (2.5 g) as a white solid that was used without any additional purification. LCMS: Method CC, [M+H] + = 190. Synthesis of intermediate B14 (5-chloro-6-(4,4-difluoropiperidin-1-yl)pyridin-3- amine)

Step 1: 3-chloro-2-(4,4-difluoropiperidin-1-yl)-5-nitropyridine 2,3-Dichloro-5-nitropyridine (5.0 g, 25.9 mmol, 1.0 equiv.) was dissolved in ACN (10 mL), then 4,4-difluoropiperidine (3.1 g, 25.9 mmol, 1.0 equiv.) and K2CO3 (7.2 g, 51.8 mmol, 2.0 equiv.) were added. The reaction mixture was heated to 80 °C for 16 hours, then cooled to ambient temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum to give 3-chloro-2-(4,4-difluoropiperidin-1-yl)-5- nitropyridine (7.1 g) as a pale yellow solid. LCMS Method CA: [M+H] + = 278. Step 2: 5-chloro-6-(4,4-difluoropiperidin-1-yl)pyridin-3-amine 3-Chloro-2-(4,4-difluoropiperidin-1-yl)-5-nitropyridine (7.0 g, 25.2 mmol, 1.0 equiv.) was dissolved in AcOH (70 mL), then Fe (2.8 g, 50.4 mmol, 2.0 equiv.) was added. The reaction mixture was heated to 60 °C for 16 hours, then cooled to ambient temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum. The resulting mixture was diluted with water, then 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 5-chloro-6-(4,4-difluoropiperidin-1-yl)pyridin-3- amine (5.6 g) as a yellow solid. LCMS Method CA: [M+H] + = 248. Synthesis of intermediate B15 (5-cyclohexylpyridin-2-amine) Step 1: 5-(cyclohex-1-en-1-yl)pyridin-2-amine 5-Bromopyridin-2-amine (10.0 g, 57.8 mmol, 1.0 equiv.) was dissolved in 1,4- dioxane (100 mL) and water (10 mL), then cyclohex-1-en-1-ylboronic acid (10.9 g, 86.7 mmol, 1.5 equiv.), K2CO3 (16.0 g, 115.6 mmol, 2.0 equiv.) and Pd(PPh3)2Cl2 (4.1 g, 5.8 mmol, 0.1 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was heated to 90 °C for 16 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:10) to give 5-(cyclohex-1-en-1-yl)pyridin-2-amine (9.8 g) as yellow oil. LCMS Method CA: [M+H] + = 175. Step 2: 5-cyclohexylpyridin-2-amine 5-(Cyclohex-1-en-1-yl)pyridin-2-amine (9.8 g, 56.2 mmol, 1.0 equiv.) was dissolved in MeOH (100 mL), then Pd/C (10% wt., 3.0 g) was added under nitrogen. The mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred 5 days at ambient temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum to give 5-cyclohexylpyridin-2-amine (7.1 g) as a brown oil. LCMS: Method CC, [M+H] + = 177. Synthesis of intermediate B16 (2-((1H-pyrrolo[2,3-c]pyridin-3-yl)amino)-2-oxoacetic acid)

Step 1: ethyl ([1H-pyrrolo[2,3-c]pyridin-3-yl]carbamoyl)formate 1H-pyrrolo[2,3-c]pyridin-3-amine hydrobromide (320.0 mg, 2.4 mmol, 1.0 equiv.) and TEA (0.7 mL, 4.8 mmol, 2.0 equiv.) were dissolved in DCM (15 mL) and cooled to 0 °C. Then ethyl oxalyl chloride (0.3 mL, 2.4 mmol, 1.0 equiv.) was added dropwise, maintaining the solution at 0 °C. The reaction mixture was stirred for 30 min at ambient temperature and then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, dried over anhydrous Na2SO4 and concentrated under vacuum to give ethyl ([1H-pyrrolo[2,3-c]pyridin-3-yl]carbamoyl)formate (780.0 mg) as a white solid that was used in the next step directly without further purification. LCMS Method CA: [M+H] + = 234. Step 2: ([1H-pyrrolo[2,3-c]pyridin-3-yl]carbamoyl)formic acid Ethyl ([1H-pyrrolo[2,3-c]pyridin-3-yl]carbamoyl)formate (780.0 mg, 3.3 mmol, 1.0 equiv.) was dissolved in MeOH (15 mL) and water (3.0 mL), then KOH (375.2 mg, 6.6 mmol, 2.0 equiv.) was added. The reaction mixture was stirred for 2 hours at ambient temperature and concentrated under vacuum. The residue was diluted with water, then adjusted to pH 3 with aqueous HCl (2M). The resulting solids were collected by filtration and dried to give ([1H-pyrrolo[2,3-c]pyridin-3-yl]carbamoyl)formic acid (352.5 mg) as a white solid. LCMS Method CA: [M+H] + =206. The following intermediates were prepared using the method described for Intermediate B16. Synthesis of intermediate B24 (ethyl 2-((5-fluoro-1H-pyrrolo[2,3-b]pyridin-3- yl)amino)-2-oxoacetate) The title compound was prepared using the same methods described for Intermediate B16, Step 1. LCMS: Method CC, [M+H] + = 252. The following intermediates were prepared using the same method described for Intermediate B24. Example 1: Synthesis of N 1 -(5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl)-N 2 -(3-methyl- 5-(trifluoromethyl)phenyl)oxalamide (Compound 101) To a mixture of 2-((3-methyl-5-(trifluoromethyl)phenyl)amino)-2-oxoacetic acid (61.8 mg, 0.25 mmol, 1.0 equiv.) and 5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-amine (37.8 mg, 0.25 mmol, 1.0 equiv.) in DMF (3 mL) was added HATU (95.0 mg, 0.25 mmol, 1.0 equiv.) and TEA (70 µl, 0.5 mmol, 2.0 equiv.). The mixture was stirred at 30 °C for 2 hours. The solvent was removed in vacuo and the residue was purified by prep HPLC to give N1-(5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl)-N2-(3-methyl-5- (trifluoromethyl)phenyl)oxalamide (40.2 mg, 0.11 mmol) as a white powder. Analysis Condition: Method AA MS-ESI, 381.1 [M+H + ]. 1 H NMR (400 MHz, DMSO-d6) δ = 11.82 (br s, 1H), 11.11 (d, 2H), 8.33–8.23 (m, 2H), 8.17 (s, 1H), 7.98 (d, 2H), 7.35 (s, 1H), 2.40 (s, 3H) Example 2: N 1 -(1H-pyrrolo[3,2-c]pyridin-3-yl)-N 2 -(3- (trifluoromethyl)phenyl)oxalamide (Compound 114) To a mixture of 2-oxo-2-((3-(trifluoromethyl)phenyl)amino)acetic acid (60.6 mg, 0.26 mmol, 1.0 equiv.) and 1H-pyrrolo[3,2-c]pyridin-3-amine (34.6 mg, 0.26 umol, 1.0 equiv.) in ACN (4 mL) was added T3P (50 wt.% in EtOAc, 300 ^l, 0.52 mmol, 2.0 equiv.) and TEA (73 ^l, 0.52 mmol, 2.0 equiv.). The mixture was stirred at 80 °C for 16 hours. After cooling to ambient temperature, the solvent was removed in vacuo. The residue was purified by prep. HPLC to give N 1 -(1H-pyrrolo[3,2-c]pyridin-3-yl)-N 2 -(3- (trifluoromethyl)phenyl)oxalamide (4.6 mg, 0.013 mmol) as an off-white powder. Analysis Condition: Method AA MS-ESI, 349.2 [M+H + ]. 1 H NMR (400MHz, DMSO-d6) δ = 11.52 (br s, 1H), 11.20 (d, 2H), 9.21 (s, 1H), 8.38 (s, 1H), 8.23–8.12 (m, 2H), 7.87 (s, 1H), 7.64 (t, 1H), 7.52 (d, 1H), 7.39 (d, 1H) Table E1. The Examples in Table E1 were prepared in a manner similar to that as described for either Example 1 or Example 2, as indicated in Table E1 below. l C I i Fi l R i I PAC LC MS, - +] y 105 Intermedia Example N 1 -(adamantan- 371.1 1 110 Intermedia Example N 1 -(1H- 439.0 y)oxaam e 108 Intermedia Example N 1 -(1H- 336.2 1 120 Intermedia Example N 1 -(1H- 439.1 y 123 Intermedia Example N 1 - 311.0 0 y Table E2. The Examples in Table E2 were prepared in a manner similar to that as described for Example 1 from the appropriate starting materials.

Table E3. The Examples in Table E3 were prepared in a manner similar to that as described for Example 2 from the appropriate starting materials.

Example 87: N-(5-chloro-6-(4,4-difluoropiperidin-1-yl)pyridin-3-yl)-N'-( 5-methyl- 1H-pyrrolo[3,2-b]pyridin-3-yl)oxalamide (Compound 195)

5-Methyl-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride (150.0 mg, 1.0 mmol, 1.0 equiv.) was dissolved in THF (20 mL), then [[5-chloro-6-(4,4-difluoropiperidin-1- yl)pyridin-3-yl]carbamoyl]formic acid (325.8 mg, 1.0 mmol, 1.0 equiv.), T3P (50% wt. in ethyl acetate, 1.8 mL, 1.5 mmol, 1.5 equiv.) and TEA (0.3 mL, 2.0 mmol, 2.0 equiv.) were added. The reaction mixture was stirred for 16 hours at ambient temperature then concentrated under vacuum. The residue was diluted with water, extracted with ethyl acetate, washed with brine, dried over anhydrous Na 2 SO 4 and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30*150mm 5μm; Mobile Phase A: Water (10 mM NH4HCO3+0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 40 B to 65 B in 8 min; 220 nm. This gave N-[5-chloro-6-(4,4- difluoropiperidin-1-yl)pyridin-3-yl]-N'-[5-methyl-1H-pyrrolo [3,2-b]pyridin-3- yl]ethanediamide (42.5 mg) as a off-white solid. LCMS Method CF: [M+H] + = 449. 1 H NMR (400 MHz, DMSO-d6) δ 11.24 (s, 1H), 11.19–11.18 (m, 1H), 9.90 (s, 1H), 8.74 (d, 1H), 8.35 (d, 1H), 8.01 (d, 1H), 7.73 (d, 1H), 7.09 (d, 1H), 3.39–3.37 (m, 4H), 2.59 (s, 3H), 2.14–2.08 (m, 4H). The following compounds were prepared using the method described for Example 87.

Example 90: N-[5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]-N'-[8-(2-methylph enyl)- 8-azabicyclo[3.2.1]octan-3-yl]ethanediamide (Compound 209) ([5-Fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]carbamoyl)formic acid (70.0 mg, 0.3 mmol, 1.0 equiv.) was dissolved in DCM (15 mL), then 8-(2-methylphenyl)-8- azabicyclo[3.2.1]octan-3-amine hydrochloride (67.8 mg, 0.3 mmol, 1.0 equiv.), HATU (178.9 mg, 0.4 mmol, 1.5 equiv.) and DIEA (0.2 mL, 0.9 mmol, 3.0 equiv.) were added. The reaction mixture was stirred for 16 hours at ambient temperature and then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30*150 mm 5μm; Mobile Phase A: Water (10 mM NH 4 HCO 3 ), Mobile Phase B: ACN; Flow rate: 50 mL/min; Gradient: 55 B to 65 B in 16 min; 220 nm. This gave N-[5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]-N'-[8-(2-methylph enyl)-8- azabicyclo[3.2.1]octan-3-yl]ethanediamide (20.7 mg) as a white solid. LCMS Method CD: [M+H] + = 422. 1 H NMR (300 MHz, DMSO-d6) δ 11.75 (s, 1H), 10.82 (s, 1H), 8.85– 8.82 (m, 1H), 8.23–8.21 (m, 2H), 7.91 (s, 1H), 7.14–7.12 (m, 1H), 7.08–7.02 (m, 1H), 6.88–6.80 (m, 2H), 4.22–4.18 (m, 1H), 3.79 (s, 2H), 2.31 (s, 3H), 1.97–1.94 (m, 4H), 1.88–1.74 (m, 4H). The following compounds were prepared using the method described for Example 90. Example 93: N-(5-cyclohexylpyridin-2-yl)-N'-(5-(1-isopropyl-1H-pyrazol-4 -yl)- 1H-pyrrolo[3,2-b]pyridin-3-yl)oxalamide (Compound 206) 5-(1-Ethylpyrazol-4-yl)-1H-pyrrolo[3,2-b]pyridin-3-amine hydrochloride (300.0 mg, 1.1 mmol, 1.0 equiv.) was dissolved in DMF (15 mL), then NMM (656.3 mg, 6.6 mmol, 6.0 equiv.), PyBop (573.5 mg, 1.1 mmol, 1.0 equiv.) and [(5-cyclohexylpyridin- 2-yl)carbamoyl]formic acid (273.1 mg, 1.1 mmol, 1.0 equiv.) were added. The reaction mixture was stirred for 1 hour at ambient temperature and then 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 Prep-HPLC with the following conditions: Column: XBridge Prep C18 OBD Column, 19*150mm 5μm; Mobile Phase A: Water (0.05% NH4OH), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 40 B to 95 B in 9 min; 210 nm. This gave N-(5- cyclohexylpyridin-2-yl)-N'-[5-(1-isopropylpyrazol-4-yl)-1H-p yrrolo[3,2-b]pyridin-3- yl]ethanediamide (60.4 mg) as a white solid. LCMS Method CE: [M+1] + = 472. 1 H NMR (400 MHz, DMSO-d6) δ 11.28 (d, 1H), 10.26 (s, 1H), 9.99 (s, 1H), 8.36 (s, 1H), 8.31 (d, 1H), 8.07–8.03 (m, 3H), 7.83–7.79 (m, 2H), 7.53 (d, 1H), 4.59–4.54 (m, 1H), 2.55–2.51 (m, 1H), 1.82–1.70 (m, 5H), 1.49–1.34 (m, 7H), 1.26–1.23 (m, 4H). The following compounds were prepared using the method described for Example 93.

Example 98: N-(5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl)-N'-(spiro[4.5]dec an-2- yl)oxalamide (Compound 204) Ethyl ([5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]carbamoyl)formate (50.0 mg, 0.2 mmol, 1.0 equiv.) was dissolved in EtOH (5 mL), then TEA (0.1 mL, 0.4 mmol, 2.0 equiv.) and spiro[4.5]decan-2-amine hydrochloride (75.3 mg, 0.4 mmol, 2.0 equiv.) were added. The resulting solution was heated to 80 °C for 12 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by Prep- HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30*150 mm 5μm; mobile phase A: water (10 mM NH4HCO3); mobile phase B: ACN (35% PhaseB up to 75% in 8 min); Detector, UV 220/254nm. This gave N-[5-fluoro-1H- pyrrolo[2,3-b]pyridin-3-yl]-N'-[spiro[4.5]decan-2-yl]ethaned iamide (6.1 mg) as a white solid. LCMS Method CK: [M+H] + = 359. 1 H NMR (300 MHz, DMSO-d6) δ 11.75 (d, 1H), 10.83 (s, 1H), 8.82 (d, 1H), 8.25–8.22 (m, 2H), 7.91 (s, 1H), 4.20–4.17 (m, 1H), 1.89–1.79 (m, 2H), 1.73–1.55 (m, 2H), 1.44–1.30 (m, 12H). The following compounds were prepared using the method described for Example 98.

xample 103: N-(5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl)-N'-(spiro[4.5]dec an-8- yl)oxalamide (Compound 198) Ethyl ([5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]carbamoyl)formate (150.0 mg, 0.6 mmol, 1.0 equiv.) and spiro[4.5]decan-8-amine (96.9 mg, 0.6 mmol, 1.0 equiv.) were dissolved in MeOH (5 mL) and cooled to 0 °C. Then AlMe3 (2M in toluene, 1.0 mL, 1.8 mmol, 3.0 equiv.) was added dropwise, maintaining the reaction mixture at 0 °C. The reaction mixture was heated to 60 °C overnight, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:1). The resulting material was further purified by Pre-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30*150 mm 5 μm; Mobile Phase A: Water (10 mM NH4HCO3), Mobile Phase B: ACN; Flow rate: 50 mL/min; Gradient: 40 B to 80 B in 7 min; 254 nm. This gave N-[5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]-N'-[spiro[4.5]dec an-8- yl]ethanediamide (28.7 mg) as an off-white solid. LCMS Method CE: [M+H] + = 359. 1 H NMR (400 MHz, DMSO-d6) δ 11.73 (s, 1H), 10.82 (s, 1H), 8.67 (d, 1H), 8.24–8.21 (m, 2H), 7.90 (d, 1H), 3.66–3.62 (m, 1H), 1.65–1.41 (m, 12H), 1.38–1.29 (m, 4H). Examples 104-105: N-[5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]-N'-[(2R or 2S)-spiro[5.5]undecan-2-yl]ethanediamide (Compound 197) and N-[5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]-N'-[(2S or 2R)-spiro[5.5]undecan- 2-yl]ethanediamide (Compound 196) Compound 18 in the scheme above was prepared using the same method described for Example 103 with intermediate B24 (ethyl ([5-fluoro-1H-pyrrolo[2,3-b]pyridin-3- yl]carbamoyl)formate) and spiro[5.5]undecan-2-amine. The racemic N-[5-fluoro-1H-pyrrolo[2,3-b]pyridin-3-yl]-N'-[spiro[5.5]und ecan-2- yl]ethanediamide (42.0 mg) was separated by Prep-Chiral-HPLC with the following conditions: Column: CHIRALPAK IA, 2*25cm, 5μm; Mobile Phase A: EtOH:DCM=1:1--HPLC, Mobile Phase B: EtOH--HPLC; Flow rate: 20 mL/min; Gradient: 50 B to 50 B in 25 min; 220/254 nm; RT1: 13.2 min, RT2: 20.3 min. This gave Compound 197 (peak one, 19.7 mg) as a white solid and Compound 196 (peak two, 18.7 mg) as a white solid. Compound 197: LCMS Method CG: [M-H]- = 371. 1 H NMR (400 MHz, DMSO- d6): δ 11.74 (s, 1H), 10.81 (s, 1H), 8.63 (d, 1H), 8.25–8.21 (m, 2H), 7.91 (d, 1H), 3.85– 3.81 (m, 1H), 1.78–1.76 (m, 2H), 1.60–1.31 (m, 12H), 1.22–1.99 (m, 2H), 1.12–1.09 (m, 1H), 0.97–0.94 (m, 1H). Compound 196: LCMS Method CG: [M-H]- = 371. 1 H NMR (400 MHz, DMSO- d6): δ 11.74 (s, 1H), 10.81 (s, 1H), 8.63 (d, 1H), 8.24–8.21 (m, 2H), 7.91 (d, 1H), 3.85– 3.81 (m, 1H), 1.78–1.76 (m, 2H), 1.60–1.31 (m, 12H), 1.22–1.99 (m, 2H), 1.12–1.09 (m, 1H), 0.97–0.94 (m, 1H). The following compounds were prepared using the same method described for Examples 104-105.

The following compounds were synthesized using methods similar to those described herein from the appropriate starting materials.

Biological Assays STING pathway activation by the compounds described herein was measured using THP1-Dual™ cells (KO-IFNAR2). THP1-Dual™ KO-IFNAR2 Cells (obtained from invivogen) were maintained in RPMI, 10% FCS, 5 ml P/S, 2mM L-glut, 10mM Hepes, and 1 mM sodium pyruvate. Compounds awere spotted in empty 384 well tissue culture plates (Greiner 781182) by Echo for a final concentration of 0.0017 - 100 µM. Cells were plated into the TC plates at 40 μL 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 1×384 plate: ^ Solution A: 2 mL Optimem with one of the following stimuli: o 60 μL of 10 mM 2'3'cGAMP -> 150 μM stock ^ Solution B: 2 mL Optimem with 60 μL 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 uL of transfection solution (A+B) was added on top of the plated cells, with a final 2’3’cGAMP concentration of 15 μM. The plates were then centrifuged immediately at 340 g for 1 minute, after which they were incubated at 37 o C, 5% CO2, >98% humidity for 24h. Luciferase reporter activity was then measured. EC50 values were calculated by using standard methods known in the art. Luciferase reporter assay: 10 µL 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 µL of QLC Stabilizer per 25 mL of QUANTI- Luc™ Plus solution was added.50 µL 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. EC50 values were calculated by using standard methods known in the art. Table BA shows the activity of compounds in STING reporter assay: <0.008 µM = “++++++”; ≥0.008 and <0.04 µM = “+++++”; ≥0.04 and <0.2 µM = “++++”; ≥0.2 and <1 µM = “+++”; ≥1 and <5 µM = “++”; ≥5 and <100 µM = “+”. Table BA Numbered Clauses The compounds, compositions, methods, and other subject matter described herein are futther 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: Z is selected from the group consisting of a bond, CR 1 , C(R 3 )2, N, and NR 2 ; each of Y 1 , Y 2 , and Y 3 is independently selected from the group consisting of O, S, CR 1 , C(R 3 )2, N, and NR 2 ; Y 4 is C or N; 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 five- membered ring comprising Y 4 , X 1 , and X 2 is heteroaryl; Q-A is defined according to (A) or (B) below: (A) Q is selected from the group consisting of: NH and N(C 1-6 alkyl) wherein the C 1-6 alkyl is optionally substituted with 1-2 independently selected R a ; and A is: (i) -(Y A1 ) n -Y A2 , wherein: ● n is 0 or 1; ● Y A1 is C 1-6 alkylene, which is optionally substituted with 1-6 substituents e o C6-10 aryl optionally substituted with 1-4 independently selected C1- 4 alkyl; and o heteroaryl of 5-10 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C 1-4 alkyl; or ● Y A1 is –Y A3 -Y A4 -Y A5 which is connected to Q via Y A3 wherein: o Y A3 is a C1-3 alkylene optionally substituted with 1-2 independently selected R a ; o Y A4 is –O-, -NH-, or -S-; and o Y A5 is a bond or C1-3 alkylene which is optionally substituted with 1-2 independently selected R a ; and ● Y A2 is: (a) C 3-20 cycloalkyl or C 3-20 cycloalkenyl, each of which is optionally substituted with 1-4 R b , (b) C6-20 aryl, which is optionally substituted with 1-4 R c ; (c) heteroaryl of 5-20 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c ; or (d) heterocyclyl or heterocycloalkenyl of 3-16 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-4 independently selected R b , or (ii) -Z 1 -Z 2 -Z 3 , wherein: ● Z 1 is C1-3 alkylene, which is optionally substituted with 1-4 R a ; ● Z 2 is –N(H)-, -N(R d )-, -O-, or –S-; and ● Z 3 is C2-7 alkyl, which is optionally substituted with 1-4 R a ; or (iii) C1-20 alkyl, which is optionally substituted with 1-6 independently selected R a , or (B) Q and A, taken together, form: E is a ring of 3-16 ring atoms, wherein, 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b , each occurrence of R 1 is independently selected from the group consisting of H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; -S(O)1-2(C 1-4 alkyl); - S(O)(=NH)(C 1-4 alkyl); SF5; -NR e R f ; –OH; oxo; -S(O)1-2(NR’R’’); -C 1-4 thioalkoxy; -NO2; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’); or a pair of R 1 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy, each occurrence of R 2 is independently selected from the group consisting of: (i) C 1-6 alkyl, which is optionally substituted with 1-2 independently selected R a ; (ii) C3-6 cycloalkyl or C3-6 cycloalkenyl; (iii) heterocyclyl or heterocycloalkenyl of 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(O) 0-2 ; (iv) C6-10 aryl; (v) heteroaryl of 5-10 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(O) 0-2 ; (vi) -C(O)(C 1-4 alkyl); (vii) -C(O)O(C 1-4 alkyl); (viii) -CON(R’)(R’’); (ix) -S(O)1-2(NR’R’’); (x) - S(O)1-2(C 1-4 alkyl); (xi) -OH; (xii) C 1-4 alkoxy; and (xiii) H; or a pair of R 1 and R 2 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R 2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy, each occurrence of R 3 is independently selected from the group consisting of H; C1- 6 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; –OH; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); - C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano; and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; or two R 3 on the same carbon combine to form an oxo; or a pair of R 3 on the same or on adjacent atoms, taken together with the atom(s) connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; or a pair of R 1 and R 3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; or a pair of R 2 and R 3 on adjacent atoms, taken together with the atoms connecting them, form a ring of 3-10 ring atoms, wherein 0-2 ring atoms (in addition to the nitrogen atom to which the R 2 is attached) are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 ; and wherein the ring is optionally substituted with 1-4 substituents each independently selected from the group consisting of C 1-6 alkyl, halo, C 1-6 haloalkyl, -OH, NR e R f , C 1-6 alkoxy, and C 1-6 haloalkoxy; R 4 is selected from the group consisting of H and C 1-6 alkyl optionally substituted with 1-3 independently selected R a ; R 5 is selected from the group consisting of H; halo; –OH; -C 1-4 alkyl; -C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)O(C 1-4 alkyl); -C(=O)(C 1-4 alkyl); - C(=O)OH; -CON(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano, and C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 independently selected C 1-4 alkyl; R 6 is selected from the group consisting of H; C 1-6 alkyl optionally substituted with 1-3 independently selected R a ; -OH; C 1-4 alkoxy; C(=O)H; C(=O)(C 1-4 alkyl); C6-10 aryl optionally substituted with 1-4 independently selected C 1-4 alkyl; and heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected C 1-4 alkyl; each occurrence of R a is independently selected from the group consisting of: – OH; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)O(C 1-4 alkyl); -C(=O)(C1- 4 alkyl); -C(=O)OH; -CON(R’)(R’’); -S(O) 1-2 (NR’R’’); -S(O) 1-2 (C 1-4 alkyl); cyano, and C 3- 6 cycloalkyl or C3-6 cycloalkenyl , each optionally substituted with 1-4 independently selected C 1-4 alkyl; each occurrence of R b is independently selected from the group consisting of: C1- 10 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; –OH; oxo; -F; -Cl; -Br; –NR e R f ; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C1-10 alkyl); -C(=O)O(C1- 4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); -S(O)1-2(NR’R’’); -S(O)1-2(C 1-4 alkyl); cyano; and –L 1 -L 2 -R h ; each occurrence of 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 ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl) or –S(O)1-2(C 1-4 haloalkyl); -NR e R f ; –OH; -S(O)1- 2(NR’R’’); -C 1-4 thioalkoxy or –C 1-4 thiohaloalkoxy; -NO2; –SF5; -C(=O)(C1-10 alkyl); - C(=O)O(C 1-4 alkyl); -C(=O)OH; -C(=O)N(R’)(R’’); and –L 1 -L 2 -R h ; R d is selected from the group consisting of: C 1-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-3 substituents each independently selected from the group consisting of halo and OH; -C(O)(C 1-4 alkyl); - C(O)O(C 1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C 1-4 alkyl); -OH; and C 1-4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; C 1-6 alkyl; C 1-6 haloalkyl; C3-6 cycloalkyl or C3-6 cycloalkenyl; -C(O)(C 1-4 alkyl); - C(O)O(C 1-4 alkyl); -CON(R’)(R’’); -S(O)1-2(NR’R’’); - S(O)1-2(C 1-4 alkyl); -OH; and C 1-4 alkoxy; or R e and R f together with the nitrogen atom to which each is attached forms a ring of 3-8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R e and R f ), which are each independently selected from the group consisting of N(R d ), NH, O, and S; -L 1 is a bond or C1-3 alkylene; -L 2 is –O-, -N(H)-, -S(O) 0-2 -, or a bond; R h is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy (in certain embodiments, it is provided that when R h is C3-6 cycloalkyl or C3- 6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected C1- 4 alkyl, -L 1 is a bond, or –L 2 is –O-, -N(H)-, or -S-); ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy; ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; -L 3 is a bond or C1-3 alkylene; -L 4 is –O-, -N(H)-, -S(O) 0-2 -, or a bond; R i is selected from the group consisting of: ● C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy (in certain embodiments, it is provided that when R i is C3-6 cycloalkyl or or cycloalkenyl, each optionally substituted with 1-4 substituents independently selected C 1-4 alkyl, -L 1 is a bond, or –L 2 is –O-, -N(H)-, or -S-); ● heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-16 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(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C1- 4 haloalkoxy; ● heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and ● C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo; C 1-4 alkyl optionally substituted with 1-2 independently selected R a ; C 1-4 haloalkyl; cyano; C 1-4 alkoxy; and C 1-4 haloalkoxy; and each occurrence of R’ and R’’ is independently selected from the group consisting of: H, -OH, C 1-4 alkyl, C6-10 aryl optionally substituted with 1-2 substituents selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, and heteroaryl of 5-10 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(O) 0-2 and wherein the heteroaryl ring is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, -OH, NH2, NH(C 1-4 alkyl), N(C 1-4 alkyl)2, C 1-4 alkyl, and C 1-4 haloalkyl; or R’ and R’’ together with the nitrogen atom to which each is attached forms a ring of 3- 8 ring atoms, wherein the ring has: (a) 1-7 ring carbon atoms, each of which is substituted with 1-2 substituents independently selected from the group consisting of H and C1-3 alkyl; and (b) 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R’’), which are each independently selected from the group consisting of N(H), N(C 1-6 alkyl), O, and S; provided that one or more of a), b), and c) apply: a) one or more of Z, Y 1 , Y 2 , Y 3 , and Y 4 in the ring below is an independently selected heteroatom; b) the ring that includes Z, Y 1 , Y 2 , Y 3 , and Y 4 is partially unsaturated; or c) Z is a bond; and further provided that the compound is other than: . 2. The compound of clause 1, wherein is aromatic. 3. The compound of any one of clauses 1-2, wherein Z is selected from the group consisting of CR 1 , N, and NR 2 . 4. The compound of any one of clauses 1-3, wherein Z is CR 1 . 5. The compound of any one of clauses 1-4, wherein 1-2 of Y 1 , Y 2 , and Y 3 is independently N or NR 2 (e.g., N); and each of the remaining of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . 6. The compound of any one of clauses 1-5, wherein one of Y 1 , Y 2 , and Y 3 is independently N or NR 2 ; and each of the remaining of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . 7. The compound of any one of clauses 1-6, wherein one of Y 1 , Y 2 , and Y 3 is independently N; and each of the remaining of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . 8. The compound of any one of clauses 1-7, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 9. The compound of clause 8, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 10. The compound of clause 8, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 11. The compound of clause 8, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 12. The compound of any one of clauses 1-7, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 13. The compound of clause 12, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 14. The compound of any one of clauses 1-7, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 15. The compound of clause 14, wherein the moiety is , wherein the asterisk denotes point of attachment to Y 4 . 16. The compound of any one of clauses 1-3, wherein Z is N. 17. The compound of any one of clauses 1-3 and 16, wherein each of Y 1 , Y 2 , and Y 3 is an independently selected CR 1 . 18. The compound of any one of clauses 1-3 and 16-17, wherein the moiety wherein the asterisk denotes point of attachment to Y 4 . 19. The compound of any one of clauses 1-18, wherein Y 4 is C. 20. The compound of any one of clauses 1-19, wherein X 1 is NR 2 . 21. The compound of any one of clauses 1-20, wherein X 1 is NH. 22. The compound of any one of clauses 1-21, wherein X 2 is CR 5 . 23. The compound of any one of clauses 1-22, wherein X 2 is CH. 24. The compound of any one of clauses 1-23, wherein X 1 is NH; and X 2 is CH. 25. The compound of any one of clauses 1-3, wherein the compound is selected from a compound of the following formulae: (Ia), (Ib), (Ic), and (Id). 26. The compound of any one of clauses 1-3 and 25, wherein the compound has formula (I-a): (Ia). 27. The compound of clause 26, wherein the compound has formula (I-a1) or (I-a2): (Ia-1) or (Ia-2); or wherein the compound has Formula (Ia-3): 28. The compound of any one of clauses 1-3 and 25, wherein the compound has formula (I-b): (Ib). 29. The compound of clause 28, wherein the compound has formula (Ib-1): (Ib-1). 30. The compound of any one of clauses 1-3 and 25, wherein the compound has formula (I-c): (Ic). 31. The compound of clause 30, wherein the compound has formula (Ic-1): (Ic-1). 32. The compound of any one of clauses 1-3 and 25, wherein the compound has formula (I-d): (Id). 33. The compound of clause 32, wherein the compound has formula (Id-1): (Id-1). 34. The compound of clause 32, wherein the compound has formula (Id-2): (Id-2). 35. The compound of clause 32, wherein the compound has formula (Id-3): (Id-3). 36. The compound of any one of clauses 1-35, wherein each occurrence of R 1 is independently selected from the group consisting of: H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; -S(O)1-2(C 1-4 alkyl); -S(O)(=NH)(C 1-4 alkyl); SF5; -S(O)1- 2(NR’R’’); -C 1-4 thioalkoxy; -NO2; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’). 37. The compound of any one of clauses 1-36, wherein 0-3 (e.g., 0, 1, 2, or 3) occurrences of R 1 is other than H; and each of the remaining occurrences of R 1 is H. 38. The compound of any one of clauses 1-37, wherein each occurrence of R 1 is H. 39. The compound of any one of clauses 1-37, wherein 1-2 occurrences of R 1 is other than H. 40. The compound of clause 39, wherein one occurrence of R 1 is other than H. 41. The compound of any one of clauses 1-36 and 39-40, wherein one occurrence of R 1 is selected from the group consisting of: halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl); -S(O)1-2(NR’R’’); -NO2; -C(=O)(C 1-4 alkyl); - C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’). 42. The compound of any one of clauses 1-36 and 39-41, wherein one occurrence of R 1 is halo (e.g., F or Cl (e.g., F)). 43. The compound of any one of clauses 1-42, wherein R 2 is H. 44. The compound of any one of clauses 1-43, wherein R 5 is H. 45. The compound of any one of cliams 1-44, wherein Q-A is defined according to (A). 46. The compound of any one of clauses 1-45, wherein Q is NH. 47. The compound of any one of clauses 1-45, wherein Q is N(C 1-3 alkyl) (e.g., NMe or NEt). 48. The compound of any one of clauses 1-47, wherein A is -(Y A1 )n-Y A2 . 49. The compound of any one of clauses 1-48, wherein n is 0. 50. The compound of any one of clauses 1-48, wherein n is 1. 51. The compound of clause 50, wherein Y A1 is C 1-6 alkylene, which is optionally substituted with 1-4 R a . 52. The compound of any one of clauses 50-51, wherein Y A1 is -CH2-, - CH2CH2-, -CH2CH2CH2-, -CH(CF3)-, -CH2CH(OH)-, , , , , or (e.g., Y A1 is CH2). 53. The compound of clause 52, wherein Y A1 is -CH2- or -CH2CH2-. 54. The compound of any one of clauses 48-53, wherein Y A2 is C6-10 aryl, which is optionally substituted with 1-3 R c . 55. The compound of any one of clauses 48-54, wherein Y A2 is C6 aryl, which is optionally substituted with 1-3 R c . 56. The compound of any one of clauses 48-55, wherein Y A2 is C6 aryl, which is substituted with 1-3 R c . 57. The compound of any one of clauses 48-56, wherein Y A2 is phenyl substituted with 1-3 R c , wherein one R c is at the ring carbon para to the point of attachment to Y A1 . 58. The compound of any one of clauses 48-56, wherein Y A2 is phenyl substituted with 1-3 R c , wherein 1-2 R c is at the ring carbons meta to the point of attachment to Y A1 ; or wherein Y A2 is phenyl substituted with 1-3 R c , wherein 1-2 R c is at the ring carbons ortho to the point of attachment to Y A1 . 59. The compound of any one of clauses 48-54, wherein Y A2 is C7-10 bicyclic aryl, which is optionally substituted with 1-3 R c (e.g., Y A2 is naphthyl indanyl (e r tetrahydronapthyl, each of which is optionally substituted with 1- 3 R c ). 60. The compound of any one of clauses 48-53, wherein Y A2 is heteroaryl of 5- 14 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . 61. The compound of any one of clauses 48-53 and 60, wherein Y A2 is heteroaryl of 6 ring atoms (e.g., pyridyl or pyrimidinyl (e.g., pyridyl)), wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl ring is optionally substituted with 1-3 independently selected R c . 62. The compound of clause 61, wherein Y A2 is substituted with 1-3 independently selected R c ; and one occurrence of R c is at the ring carbon atom para to the point of attachment to Y A1 (e.g., or ). 63. The compound of clause 61, wherein Y A2 is substituted with 1-3 independently selected R c ; and one occurrence of R c is at the ring carbon atom meta to the point of attachment to Y A1 . 64. The compound of any one of clauses 48-53, wherein Y A2 is bicyclic or tricyclic heteroaryl of 7-14 (e.g., 9-12 (e.g., 9, 10, 11, or 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . 65. The compound of clause 64, wherein Y A2 is bicyclic heteroaryl of 9-10 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . 66. The compound of clause 65, wherein Y A2 is bicyclic heteroaryl of 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(O) 0-2 (e.g., , or and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . 67. The compound of any one of clauses 54-66, wherein each occurrence of 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 ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1-2(C 1-4 alkyl); -NR e R f ; -C 1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C 1-4 alkyl); and–L 1 -L 2 -R h . 68. The compound of any one of clauses 54-67, wherein one occurrence of R c is halo (e.g., F or Cl (e.g., Cl)) ; or wherein one occurrece of R c is C 2-6 alkynyl (e.g., ); or wherein one occurrence of R c is C 1-4 alkoxy or C 1-4 haloalkoxy. 69. The compound of any one of clauses 54-67, wherein one occurrence of R c is is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a . 70. The compound of any one of clauses 54-67 and 69, wherein one occurrence of R c is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10). 71. The compound of clause 70, wherein one occurrence of R c is ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl, iso-butyl, sec-butyl, tert-butyl), or octyl (e.g., n-octyl). 72. The compound of clause 69, wherein one occurrece of R c is C1-10 alkyl which is substituted with 1-6 independently selected R a . 73. The compound of clause 72, wherein each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy. 74. The compound of clause 73, wherein each occurrence of R a is halo (e.g., F). 75. The compound of any one of clauses 69 and 72, wherein one occurrence of R c is CF3. 76. The compound of any one of clauses 54-67, wherein one occurrence of R c is –L 1 -L 2 -R h . 77. The compound of clause 76, wherein L 1 is a bond. 78. The compound of any one of clauses 76-77, wherein L 2 is a bond; or wherein L 2 is –CH 2 -. 79. The compound of any one of clauses 76-78, wherein R h is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl. 80. The compound of any one of clauses 76-79, wherein R h is C6 aryl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl (e.g., ). 81. The compound of any one of clauses 76-78, wherein R h is heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-10 (e.g., 5-6) ring atoms, wherein 1-3 (e.g,, 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl. 82. The compound of clause 81, wherein r wherein R h is .. 83. The compound of any one of clauses 76-78, wherein R h is C3-8 cycloalkyl or C3-8 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl. 84. The compound of clause 83, wherein R h is C3-6 cycloalkyl or C3-6 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl. 85. The compound of clause 84, wherein R h is selected from the group consisting of: 86. The compound of any one of clauses 68-85, wherein each of the remaining occurrences of R c is C 1-6 alkyl or halo; or wherein each of the remaining occurrences of R c is C 1-4 alkoxy or C 1-4 haloalkoxy. 87. The compound of any one of clauses 1-53, wherein Y A2 is monocyclic C3- 10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with 1-4 R b . 88. The compound of any one of clauses 1-53 and 87, wherein Y A2 is C3-6 (e.g., C3, C5, or C6) cycloalkyl or C3-6 (e.g., C3, C5, or C6) cycloalkenyl, each of which is substituted with 1-4 (e.g., 1-2) R b (e.g., Y A2 is cyclopropyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with 1-2 R b ). 89. The compound of clause 88, wherein Y A2 is cyclohexyl which is optionally substituted with 1-2 R b . 90. The compound of clause 89, wherein one occurrence of R b is at the ring carbon atom para to the point of attachment to Y A1 ; or one occurrence of R b is at the ring carbon atom meta to the point of attachment to Y A1 . 91. The compound of any one of clauses 1-53, wherein Y A2 is bicyclic, tricyclic, or polycyclic C7-20 (e.g., C7-12) cycloalkyl or C7-20 (e.g., C7-12) cycloalkenyl, each optionally substituted with 1-2 R b . 92. The compound of clause 91, wherein Y A2 is selected from the group consisting of: spiro[5.5]undecanyl (e.g., ), bicyclo[2.2.1]hept-2-enyl (e.g., ), bicyclo[2.2.1]heptanyl (e.g., ), spiro[2.5]octanyl (e.g., ), and adamantly (e.g., ). 93. The compound of any one of clauses 1-53, wherein Y A2 is heterocyclyl or heterocycloalkenyl of 3-16 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(O) 0-2 , and wherein the heterocyclyl or heterocycloalkenyl ring is optionally substituted with 1-3 independently selected R b (e.g., Y A2 is or ). 94. The compound of any one of clauses 87-93, wherein each occurrence of R b substituent of Y A2 is independently selected from the group consisting of: C1-10 alkyl optionally substituted with 1-6 independently selected R a ; C 1-4 haloalkyl; -F; -Cl; -Br; cyano; C 1-4 alkoxy; C 1-4 haloalkoxy; -C(=O)(C1-10 alkyl); -C(=O)O(C 1-4 alkyl); -S(O)1-2(C1- 4 alkyl); oxo; cyano; and –L 1 -L 2 -R h . 95. The compound of any one of clauses 87-94, wherein one occurrece of R b substituent of Y A2 is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a . 96. The compound of clause 95, wherein one occurrence of R b substituent of Y A2 is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10). 97. The compound of clause 96, wherein one occurrence of R b substituent of Y A2 is ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl; or sec-butyl; or tert-butyl; or iso- butyl), or octyl (e.g., n-octyl). 98. The compound of clause 95, wherein one occurrece of R b substituent of Y A2 is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., R b is CF3 or - CF2CH3). 99. The compound of clause 98, wherein each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy. 100. The compound of any one of clauses 87-94, wherein one occurrence of R b substituent of Y A2 is –L 1 -L 2 -R h (e.g., -R h or –CH2-R h such as benzyl); or wherein one occurrence of R b substituent of Y A2 is C 1-4 alkoxy or C 1-4 haloalkoxy (e.g., ). 101. The compound of any one of clauses 87-94, wherein one occurrence of R b is –F or –Cl (e.g., -F). 102. The compound of any one of clauses 1-53, wherein Y A2 is ; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . 103. The compound of any one of clauses 1-53, wherein n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . 104. The compound of any one of clauses 1-53, wherein Y A2 is ; one of Q 1 and Q 2 is N; the other one of Q 1 and Q 2 is CH; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . 105. The compound of any one of clauses 1-53, wherein ; one of Q 1 , Q 2 , Q 3 , and Q 4 is N; each of the remaining of Q 1 , Q 2 , Q 3 , and Q 4 is CH; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c . 106. The compound of any one of clauses 102-105, wherein R cA is selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1- 2(C 1-4 alkyl); -NR e R f ; -C 1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C 1-4 alkyl); and –L 1 -L 2 -R h . 107. The compound of any one of clauses 102-106, wherein R cA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl, iso-butyl, sec-butyl, tert-butyl), or octyl (e.g., n-octyl). 108. The compound of any one of clauses 102-106, wherein R cA is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy), such as C 1-10 alkyl which is substituted with 1-6 independently selected halo (e.g., R cA is CF3). 109. The compound of clause 106, wherein R cA is C 2-6 alkynyl (e.g., ); or wherein R cA is C 1-4 alkoxy or C 1-4 haloalkoxy. 110. The compound of any one of clauses 102-106, wherein R cA is –L 1 -L 2 -R h . 111. The compound of clause 110, wherein –L 1 is a bond. 112. The compound of any one of clauses 110-111, wherein –L 2 is a bond. 113. The compound of any one of clauses 110-112, wherein R h is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl, such as C 6 aryl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl (e.g., ). 114. The compound of any one of clauses 110-112, wherein R h is heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-10 (e.g., 5-6) ring atoms, wherein 1-3 (e.g,, 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, such as . 115. The compound of any one of clauses 110-112, wherein R h is C3-8 (e.g., C3- 6 ) cycloalkyl or C 3-8 (e.g., C 3-6 ) cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl (e.g., R h is cyclohexyl). 116. The compound of any one of clauses 102-115, wherein n1 is 0. 117. The compound of any one of clauses 102-115, wherein n1 is 1 or 2 (e.g., 1). 118. The compound of clause 117, wherein each occurrence of R cB is independently halo or C1-3 alkyl (e.g., halo). 119. The compound of any one of clauses 1-53, wherein Y A2 is ; n2 is 0, 1, or 2; and each of R bA and R bB is an independently selected R b . 120. The compound of any one of clauses 1-53, wherein Y A2 is ; n2 is 0, 1, or 2; and each of R bA and R bB is an independently selected R b . 121. The compound of any one of clauses 119-120, wherein R bA is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a . 122. The compound of clause 121, wherein R bA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl; or sec-butyl; or tert-butyl; or iso-butyl), or octyl (e.g., n-octyl). 123. The compound of clause 121, wherein R bA is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., each R a is selected from the group consisting of halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy) (e.g., R bA is CF3 or -CF2CH3). 124. The compound of any one of clauses 119-120, wherein R bA is –F or –Cl. 125. The compound of any one of clauses 119-120, wherein R bA is –L 1 -L 2 -R h (e.g., -R h or –CH2-R h such as benzyl); or wherein R bA is C 1-4 alkoxy or C 1-4 haloalkoxy ( 126. The compound of any one of clauses 119-125, wherein n2 is 0. 127. The compound of any one of clauses 119-125, wherein n2 is 1 or 2; and/or wherein each occurrence R bB is selected from the group consisting of –F, -Cl, and C1-3 alkyl. 128. The compound of any one of clauses 1-44, wherein Q-A is as defined according to (B); and E a ring of 5-8 ring atoms, wherein aside from the nitrogen atom present, 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b (e.g., E is piperidinyl which is optionally substituted with 1-2 independently selected wherein R b is C 1-6 alkyl)). 129. The compound of clause 1, wherein the compound has the following formula: wherein n1 is 0, 1, or 2; each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. 130. The compound of clause 1, wherein the compound has the following formula: wherein n1 is 0, 1, or 2; each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. 131. The compound of clause 1, wherein the compound has the following formula: wherein one of Q 1 and Q 2 is N; the other one of Q 1 and Q 2 is CH; n1 is 0, 1, or 2; each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. 132. The compound of clause 1, wherein the compound has the following formula:

wherein one of Q 1 , Q 2 , Q 3 , and Q 4 is N; each of the remaining of Q 1 , Q 2 , Q 3 , Q 4 is CH; n1 is 0, 1, or 2; and each of R cA and R cB is an independently selected R c ; and R 7 is H or C 1-4 alkyl. 133. The compound of clause 1, wherein the compound has the following formula: wherein B 1 is selected from the group consisting of: (a) bicyclic or tricyclic heteroaryl of 7-14 (e.g., 9-12 (e.g., 9, 10, 11, or 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c ; and (b) C7-10 bicyclic aryl, which is optionally substituted with 1-3 R c ; and R 7 is H or C 1-4 alkyl. 134. The compound of clause 133, wherein B 1 is bicyclic or tricyclic heteroaryl of 9-10 (e.g., 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(O) 0-2 , and wherein the heteroaryl ring is optionally substituted with 1-4 independently selected R c . 135. The compound of clause 134, wherein 136. The compound of any one of clauses 129-132, wherein R cA is selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 alkoxy; C 1-4 haloalkoxy; -S(O)1- 2(C 1-4 alkyl); -NR e R f ; -C 1-4 thioalkoxy; -C(=O)(C1-10 alkyl); -C(=O)(OH); –C(=O)O(C 1-4 alkyl); and –L 1 -L 2 -R h . 137. The compound of any one of clauses 129-132 and 136, wherein R cA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n- propyl), butyl (e.g., n-butyl, iso-butyl, sec-butyl, tert-butyl), or octyl (e.g., n-octyl). 138. The compound of any one of clauses 129-132 and 136, wherein R cA is C1- 10 alkyl which is substituted with 1-6 independently selected R a (e.g., each occurrence of R a is independently selected from halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy), such as C1- 10 alkyl which is substituted with 1-6 independently selected halo (e.g., R cA is CF3). 139. The compound of clause 138, wherein R cA is R cA is C 2-6 alkynyl (e.g., or wherein R cA is C 1-4 alkoxy or C 1-4 haloalkoxy. 140. The compound of any one of clauses 129-132 and 136, wherein R cA is –L 1 - L 2 -R h . 141. The compound of clause 140, wherein –L 1 is a bond. 142. The compound of any one of clauses 140-141, wherein –L 2 is a bond. 143. The compound of any one of clauses 140-142, wherein R h is C6-10 aryl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl, such as C6 aryl, which is optionally substituted with 1-2 substituents independently selected from the group consisting of halo, C 1-4 alkyl, or C 1-4 haloalkyl (e.g., 144. The compound of any one of clauses 140-142, wherein R h is heterocyclyl or heterocycloalkenyl, wherein the heterocyclyl or heterocycloalkenyl has 3-10 (e.g., 5-6) ring atoms, wherein 1-3 (e.g,, 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl, such as . 145. The compound of any one of clauses 140-142, wherein R h is C3-8 (e.g., C3- 6) cycloalkyl or C3-8 (e.g., C3-6) cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of halo, C 1-4 alkyl, and C 1-4 haloalkyl (e.g., R h is cyclohexyl). 146. The compound of any one of clauses 129-145, wherein n1 is 0. 147. The compound of any one of clauses 129-145, wherein n1 is 1 or 2 (e.g., 1). 148. The compound of clause 147, wherein each occurrence of R cB is independently halo or C1-3 alkyl (e.g., halo). 149. The compound of clause 1, wherein the compound has the following formula: wherein n2 is 0, 1, or 2; each of R bA and R bB is an independently selected R b ; and R 7 is H or C 1-4 alkyl. 150. The compound of clause 1, wherein the compound has the following formula: wherein n2 is 0, 1, or 2; each of R bA and R bB is an independently selected R b ; and R 7 is H or C 1-4 alkyl. 151. The compound of clause 1, wherein the compound has the following formula: wherein B 2 is: bicyclic, tricyclic, or polycyclic C7-20 cycloalkyl or C7-20 cycloalkenyl, each optionally substituted with 1-2 R b ; and R 7 is H or C 1-4 alkyl. 152. The compound of clause 151, wherein B 2 is selected from the group consisting of: spiro[5.5]undecanyl (e.g., ), bicyclo[2.2.1]hept-2-enyl (e.g., ), bicyclo[2.2.1]heptanyl (e.g., ), spiro[2.5]octanyl (e.g., ), and adamantly (e.g., ). 153. The compound of any one of clauses 149-150, wherein R bA is C1-10 alkyl which is optionally substituted with 1-6 independently selected R a . 154. The compound of clause 153, wherein R bA is unsubstituted C1-10 alkyl (e.g., C2, C3, C4, C5, C6, or C7-10), such as ethyl, propyl (e.g., n-propyl), butyl (e.g., n-butyl; or sec-butyl; or tert-butyl; or iso-butyl), or octyl (e.g., n-octyl). 155. The compound of clause 153, wherein R bA is C1-10 alkyl which is substituted with 1-6 independently selected R a (e.g., each R a is selected from the group consisting of halo, OH, C 1-4 alkoxy, and C 1-4 haloalkoxy) (e.g., R bA is CF3). 156. The compound of any one of clauses 149-150, wherein R bA is –F or –Cl. 157. The compound of any one of clauses 149-150, wherein R bA is –L 1 -L 2 -R h (e.g., -R h or –CH2-R h such as benzyl); or wherein R bA is C 1-4 alkoxy or C 1-4 haloalkoxy (e.g., ). 158. The compound of any one of clauses 149-157, wherein n2 is 0. 159. The compound of any one of clauses 149-157, wherein n2 is 1 or 2. 160. The compound of clause 159, wherein each occurrence R bB is selected from the group consisting of –F, -Cl, and C1-3 alkyl. 161. The compound of any one of clauses 129-160, wherein n is 0. 162. The compound of any one of clauses 129-160, wherein n is 1. 163. The compound of clause 162, wherein Y A1 is C 1-6 alkylene, which is optionally substituted with 1-2 R a . 164. The compound of clause 163, wherein Y A1 is -CH 2 -, -CH 2 CH 2 -, - CH2CH2CH2-, -CH(CF3)-, -CH2CH(OH)-, (e.g., CH2), , or (e.g., Y A1 is -CH2- or -CH2CH2-). 165. The compound of clause 1, wherein the compound has the following formula: (I-9), E is a ring of 3-16 ring atoms, wherein 0-3 ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b . 166. The compound of clause 165, wherein E is a ring of 5-8 ring atoms, wherein 0-3 additional ring atoms are heteroatoms (in addition to the nitrogen atom that is present), each independently selected from the group consisting of N, N(H), N(R d ), O, and S(O) 0-2 , and wherein the ring is optionally substituted with 1-4 independently selected R b (e.g., E is piperidinyl which is optionally substituted with 1-2 independently selected R b (e.g., E is , wherein R b is C 1-6 alkyl)). 167. The compound of any one of clauses 129-166, wherein the moiety is (e.g., , , or ). 168. The compound of any one of clauses 129-166, wherein the moiety is (e.g., ). 169. The compound of any one of clauses 129-166, wherein the moiety is (e.g., ). 170. The compound of any one of clauses 129-166, wherein the moiety is (e.g., or ). 171. The compound of any one of clauses 129-170, wherein R 2 is H. 172. The compound of any one of clauses 129-171, wherein R 5 is H. 173. The compound of any one of clauses 129-164 and 167-172, wherein R 7 is H. 174. The compound of any one of clauses 129-173, wherein each R 1 is independently selected from the group consisting of: H; halo; cyano; C 1-6 alkyl optionally substituted with 1-2 R a ; C 2-6 alkenyl; C 2-6 alkynyl; C 1-4 haloalkyl; C 1-4 alkoxy; C 1-4 haloalkoxy; –L 3 -L 4 -R i ; -S(O)1-2(C 1-4 alkyl); -S(O)(=NH)(C 1-4 alkyl); SF5; -S(O)1- 2(NR’R’’); -C 1-4 thioalkoxy; -NO2; -C(=O)(C 1-4 alkyl); -C(=O)O(C 1-4 alkyl); -C(=O)OH; and -C(=O)N(R’)(R’’). 175. The compound of clause 174, wherein each occurrence of R 1 is H. 176. The compound of clause 174, wherein 1-2 occurrences of R 1 is other than H. 177. The compound of any one of clauses 174 and 176, wherein one occurrence of R 1 is halo (e.g., F). 178. The compound of any one of clauses 1-177, wherein R 6 is H. 179. The compound of clause 1, wherein the compound is selected from the group consisting of the compounds in Table C1, or a pharmaceutically acceptable salts thereof. 180. A pharmaceutical composition comprising a compound of clauses 1-179 and one or more pharmaceutically accetapble excipients. 181. A method for inhibiting STING activity, the method comprising contacting STING with a compound as defined in any one of clauses 1-179. 182. The method of clause 181, wherein the inhibiting comprises antagonizing STING. 183. The method of any one of clauses 181-182, which is carried out in vitro. 184. The method of clause 183, wherein the method comprises contacting a sample comprising one or more cells comprising STING with the compound. 185. The method of clause 183 or 184, wherein the one or more cells are one or more cancer cells. 186. The method of clause 184 or 185 wherein the sample further comprises one or more cancer cells (e.g., 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). 187. The method of clause 181, which is carried out in vivo. 188. The method of clause 187, 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. 189. The method of clause 188, wherein the subject is a human. 190. The method of clause 188, wherein the disease is cancer. 191. The method of clause 190, 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. 192. The method of clause 190 or 191, wherein the cancer is a refractory cancer. 193. The method of clause 188, wherein the compound is administered in combination with one or more additional cancer therapies. 194. The method of clause 193, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof. 195. The method of clause 194, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents. 196. The method of clause 195, 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, Muromonab-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 anti- helminthic 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-β (TGFβ), 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–4‑1BB ligand, OX40–OX40 ligand, GITR, GITR ligand – GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25–TL1A, CD40L, CD40– CD40 ligand, HVEM–LIGHT–LTA, HVEM, HVEM – BTLA, HVEM – CD160, 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). 197. The method of any one of clauses 188-196, wherein the compound is administered intratumorally. 198. 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- 179, or a pharmaceutical composition as defined in clause 180. 199. The method of clause 198, 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. 200. The method of clause 198 or 199, wherein the cancer is a refractory cancer. 201. The method of clause 198, wherein the compound is administered in combination with one or more additional cancer therapies. 202. The method of clause 201, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof. 203. The method of clause 202, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents. 204. The method of clause 203, 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, Muromonab-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 anti- helminthic 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-β (TGFβ), 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–4‑1BB ligand, OX40–OX40 ligand, GITR, GITR ligand – GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25–TL1A, CD40L, CD40– CD40 ligand, HVEM–LIGHT–LTA, HVEM, HVEM – BTLA, HVEM – CD160, 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). 205. The method of any one of clauses 198-204, wherein the compound is administered intratumorally. 206. 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 1-179, or a pharmaceutical composition as defined in clause 180. 207. The method of clause 206, wherein the subject has cancer. 208. The method of clause 207, wherein the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies. 209. The method of clause 207, 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 . 210. The method of clause 209, wherein the cancer is a refractory cancer. 211. The method of clause 206, wherein the immune response is an innate immune response. 212. The method of clause 211, wherein the at least one or more 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 213, 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, Muromonab-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 anti- helminthic 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-β (TGFβ), 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–4‑1BB ligand, OX40–OX40 ligand, GITR, GITR ligand – GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25–TL1A, CD40L, CD40– CD40 ligand, HVEM–LIGHT–LTA, HVEM, HVEM – BTLA, HVEM – CD160, 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. 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 1-179, or a pharmaceutical composition as defined in clause 180. 216. 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 1-179, or a pharmaceutical composition as defined in clause 180. 217. A method of treatment comprising administering to a subject a compound as defined in any one of clauses 1-179, or a pharmaceutical composition as defined in clause 180, 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. 218. The method of any one of clauses 215-217, wherein the disease is cancer. 219. The method of clause 218, 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. 220. The method of clause 218 or 219, wherein the cancer is a refractory cancer. 221. The method of any one of clauses 218-220, wherein the compound is administered in combination with one or more additional cancer therapies. 222. The method of clause 221, wherein the one or more additional 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 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, Muromonab-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 anti- helminthic 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-β (TGFβ), 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–4‑1BB ligand, OX40–OX40 ligand, GITR, GITR ligand – GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25–TL1A, CD40L, CD40– CD40 ligand, HVEM–LIGHT–LTA, HVEM, HVEM – BTLA, HVEM – CD160, 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. The method of any one of clauses 215-224, wherein the compound is administered intratumorally. 226. 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 1-179, or a pharmaceutical composition as defined in clause 180. 227. The method of clause 226, wherein the disease, disorder, or condition is selected from type I interferonopathies, Aicardi-Goutières Syndrome (AGS), genetic forms of lupus, inflammation-associated disorders, and rheumatoid arthritis. 228. The method of clause 227, wherein the disease, disorder, or condition is a type I interferonopathy (e.g., STING-associated vasculopathywith onset in infancy (SAVI)). 229. The method of clause 228, wherein the type I interferonopathy is STING- associated vasculopathy with onset in infancy (SAVI)). 230. The method of clause 227, wherein the disease, disorder, or condition is Aicardi-Goutières Syndrome (AGS). 231. The method of clause 227, wherein the disease, disorder, or condition is a genetic form of lupus. 232. The method of clause 227, wherein the disease, disorder, or condition is inflammation-associated disorder. 233. The method of clause 232, wherein the inflammation-associated disorder is systemic lupus erythematosus. 234. The method of any one of clauses 181-233, wherein the method further comprises identifying the subject. 235. A combination comprising a compounds defined in any one of clauses 1 to 179 or a pharmaceutically acceptable salt or tautomer thereof, and one or more therapeutically active agents. 236. A compound defined in any one of clauses 1 to 179 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 180, for use as a medicament. 237. A compound defined in any one of clauses 1 to 179 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 180, for use in the treatment of a disease, condition or disorder modulated by STING inhibition. 238. A compound defined in any one of clauses 1 to 179 or a pharmaceutically acceptable salt or tautomer thereof, or the pharmaceutical composition defined in clause 180, for use in the treatment of a disease mentioned in any one of clauses 181 to 234. 239. Use of a compound defined in any one of clauses 1 to 179 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 180, in the manufacture of a medicament for the treatment of a disease mentioned in in any one of clauses 181 to 234.