Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
BI-FUNCTIONAL COMPOUNDS AND METHODS FOR TARGETED UBIQUITINATION OF ANDROGEN RECEPTOR
Document Type and Number:
WIPO Patent Application WO/2024/102810
Kind Code:
A1
Abstract:
The present invention relates to bi-functional compounds which function to recruit endogenous proteins to an E3 ubiquitin ligase for degradation, and methods for using same. More specifically, the present disclosure provides specific proteolysis targeting chimera (PROTAC) molecules which find utility as modulators of targeted ubiquitination of a variety of polypeptides and other proteins, in particular the androgen receptor of a slice variant of AR which lacks the LBD, labelled as AR-V7, which are then degraded and/or otherwise inhibited by the compounds as described herein.

Inventors:
VAZ ROY J (US)
DESANTIS JENNY (IT)
ELEUTERI MICHELA (IT)
Application Number:
PCT/US2023/079072
Publication Date:
May 16, 2024
Filing Date:
November 08, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MONTELINO THERAPEUTICS INC (US)
International Classes:
C07D417/12; A61K31/5377; A61K31/5386; A61P35/00; C07D417/14; C07D498/08
Domestic Patent References:
WO2021058017A12021-04-01
WO2020160295A12020-08-06
WO2021236695A12021-11-25
WO2017197046A12017-11-16
WO2023001028A12023-01-26
WO2023139199A12023-07-27
WO2023093845A12023-06-01
WO2015120543A12015-08-20
WO2021236695A12021-11-25
WO2023039601A12023-03-16
WO2023039602A12023-03-16
WO2023039603A22023-03-16
WO2023039604A12023-03-16
WO2010045401A12010-04-22
WO2008113756A22008-09-25
Foreign References:
US20170327469A12017-11-16
US20180099940A12018-04-12
US20200239430A12020-07-30
US20200282068A12020-09-10
US20230132823A12023-05-04
US20230134817A12023-05-04
US11098025B22021-08-24
US11787778B22023-10-17
US11547759B22023-01-10
Other References:
TAPLIN, M. E. ET AL., J. CLIN. ONCOL., vol. 21, 2003, pages 2673 - 8
TILLEY, W. D. ET AL., CANCER RES., vol. 54, 1994, pages 4096 - 4102
HELZLSOUER, K.J. ET AL., JAMA, vol. 274, 1995, pages 1926 - 1930
EDMONDSON, R. J. ET AL., BR. J. CANCER, vol. 86, 2002, pages 879 - 885
RISCH, H. A., J. NATL. CANCER INST., vol. 90, 1998, pages 1774 - 1786
RAO, B. R. ET AL., ENDOCR. REV., vol. 12, 1991, pages 14 - 26
CLINTON, G. M. ET AL., CRIT. REV. ONCOL. HEMATOL., vol. 25, 1997, pages 1 - 9
GAO, W. Q. ET AL., CHEM. REV., vol. 105, 2005, pages 3352 - 3370
ZHOU, X. E. ET AL., J. BIOL. CHEM., vol. 285, 2010, pages 9161 - 9171
EORGET, V. ET AL., BIOCHEMISTRY, vol. 41, 2002, pages 11824 - 11831
FANG, Y. F. ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 28697 - 28702
WONG, C. I. ET AL., J. BIOL. CHEM., vol. 268, 1993, pages 19004 - 19012
ANTONARAKIS, E. S. ET AL., NEW ENGL. J. MED., vol. 37, 2014, pages 1028 - 1038
HUIFANG LI ET AL., J. MED. CHEM., vol. 57, 2014, pages 6458 - 6467
DALAL, K. ET AL., MOL. CANCER THER., vol. 16, 2017, pages 2281 - 2291
XU, R. ET AL., CHEM. BIOL. & DRUG DESIGN, vol. 91, no. 1, 2018, pages 172 - 180
CHOPRA, R.SADOK, A.COLLINS, I., DRUG DISC TODAY: TECHNOLOGIES, no. 31, 2019, pages 5 - 13
SHIBATA, N. ET AL., J. MED. CHEM., vol. 61, no. 2, 2018, pages 543 - 575
DESHALES, R.J., NATURE CHEM BIOL., November 2015 (2015-11-01), pages 634 - 635
SKALNIAK, L. ET AL., EXPERT OPIN. THER, PATENTS, vol. 29, 2019, pages 151 - 170
UNITED STATES PATENT OFFICE MANUAL OF PATENT EXAMINING PROCEDURES, SECTION 2111.03
T. W. GREENP. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
MIN J. ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 60, 2021, pages 26663 - 26670
BHUMIREDDY A ET AL., BIOORG. MED. CHEM. LETT., 2022, pages 128448
GEUN TAEK LEE ET AL., MOLECULAR CANCER THERAPEUTICS, vol. 20, 2021, pages 490 - 9
Attorney, Agent or Firm:
POKER, Cory (US)
Download PDF:
Claims:
CLAIMS What is claimed is: 1. A compound having a chemical structure ARB-L-E3LB, wherein ARB is an AR binding moiety that does not bind to a ligand binding domain, E3LB is an E3 ligase binding moiety, and L is a linker coupling the AR binding moiety to the E3 ligase binding moiety, and wherein: (A) the E3LB moiety has the following general structure: , wherein: “ ”represents a bond that may be stereospecific ((R) or (S)), or non- stereospecific; C is an aryl or heteroaryl ring, each optionally substituted by one or more substituents R9; Q is -CH- or -N-; each R2 and R3 is independently selected from H, halo, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy; R4 is selected from H, OH, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), C1-6 alkyl-OCOOR6, C1-6 alkyl-OCONR5R7, CH2-heterocycloalkyl optionally substituted with R5, or benzyl optionally substituted with R5; R5 and R7 are each independently a bond, H, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), or -C(=O)R6, wherein each of said alkyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); and R6 is independently selected from CONR5R7, OR5, NR5R7, SR5, SO2R5, SO2NR5R7, CR5R7, CR5NR5R7, aryl, heteroaryl, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), P(O)(OR5)R7, P(O)R5R7, OP(O)(OR5)R7, OP(O)R5R7, Cl, F, Br, I, CF3, CHF2, CH2F, CN, NR5SO2NR5R7, NR5CONR5R7, CONR5COR7, NR5C(=N-CN)NR5R7, C(=N- CN)NR5R7, NR5C(-N=CN)R7, NR5C(=C-NO2)NR5R7, SO2NR5COR7, NO2, CO2R5, C(C=N-OR5)R7, CR5, CR5R7, CCR5, S(C=O)(C=N-R5)R7, SF5, R5NR5R7, (R5O)nR7, or OCF3; wherein n is an integer from 1 to 10; and R9 is independently selected from H, halo (e.g., fluoro, chloro), hydroxy, -CONH2, - CONR2R3, -SONH2, -SONR2R3, -SO2NH2, -SO2NR2R3, -NHCOC1-3 alkyl (optionally substituted by 1 or more halo), -NR2COC1-3 alkyl (optionally substituted by 1 or more halo), -NR2SO2C1-3 alkyl (optionally substituted by 1 or more halo), -NR2SOC1-3 alkyl (optionally substituted by 1 or more halo), cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R9 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C3-6 cycloalkyl, C3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl). 2. The compound of claim 1, wherein the E3LB moiety has a general structure selected from the following:

wherein R4, R9, and L are as provided in Compound 1.1, and wherein each X is independently selected from C(R9) and -N-, each Y is independently selected from C(R9), -NH-, -N(R10)-, -O-, and -S-, and Z is -C- or -N-; wherein R10 is selected from C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), -C(=O)R6, - C(=O)OR6, or -C(=O)NR2R6, wherein each of said alkyl, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl). 3. The compound of claim 1 or 2, wherein ring C is a 6-membered aryl, optionally substituted by one or more group R9, or a 6-membered heteroaryl (e.g., pyridine, pyrimidine, pyridazine, or pyrazine), optionally substituted by one or more group R9, or a 5-membered heteroaryl, optionally substituted by one or more groups R9. 4. The compound of claim 3, wherein ring C is phenyl, pyridine, pyrimidine, pyridazine, or pyrazine, optionally substituted by one or more group R9. 5. The compound of claim 3, wherein ring C is selected from furan, thiophene, pyrrole, pyrazole, thiazole, oxazole, imidazole, isothiazole, isoxazole, triazole, thiadiazole, or oxadiazole, each optionally substituted by one or more groups R9.

6. The compound of claim 1, wherein the E3LB moiety is a moiety selected from any of the following:

7. The compound of claim 1, wherein the E3LB moiety is a moiety is selected from any of the following: 8. The compound of claim 1, wherein the E3LB moiety is: 9. The compound of any preceding claim, wherein each R9 is independently selected from H, halo (e.g., fluoro, chloro or bromo), hydroxy, CN, C1-6 alkyl, NH2, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl), and wherein R4 is H or C1-3 alkyl (e.g., methyl). 10. The compound of any preceding claim, wherein R4 and R9 are H. 11. The compound of claim 1, wherein the E3LB moiety is: . 12. The compound of claim 1, wherein the AR binding moiety is selected from: ,

, wherein: A is an aryl (e.g., phenyl), heteroaryl (e.g., pyridyl), C3-7 cycloalkyl, or 3-10 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as morpholine) or a bridged bicyclic 7-12 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as a bridged morpholine), each of which is optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); B is an aryl (e.g., phenyl) or heteroaryl (e.g., pyridyl or imidazolyl) ring, optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); each R1 is independently selected from H, halo (e.g., fluoro), hydroxy, -CONH2, - CONR2R3, -SONH2, -SONR2R3, -SO2NH2, -SO2NR2R3, -NHCOC1-3 alkyl (optionally substituted by 1 or more halo), -NR2COC1-3 alkyl (optionally substituted by 1 or more halo), -NR2SO2C1-3 alkyl (optionally substituted by 1 or more halo), -NR2SOC1-3 alkyl (optionally substituted by 1 or more halo), cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R1 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C3-6 cycloalkyl, C3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); and each R2 and R3 is independently selected from H, halo, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy. 13. The compound according to claim 12, wherein A is: wherein each of Q1, Q2, and Q3 are independently selected from O, S, NR2, C1-3 alkyl (e.g., CH2 or CH2CH2) optionally substituted by one or more R1, or wherein Q1 or Q2 is absent; and wherein B is selected from: 14. The compound according to claim 1, wherein the AR binding moiety is selected from:

16. The compound according to claim 1, wherein the AR binding moiety is:

17. The compound according to claim 1, wherein the linker (“L”) consists of a chemical structural unit represented by the formula -Aq-, in which q is an integer greater than 1, and each A is independently selected from the group consisting of: a bond, CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, , SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(=NCN)NRL4, NRL3C(=NCN), NRL3C(=CNO2)NRL4, C3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 RL1 and/or 0-6 RL2 groups; and wherein: RL1, RL2, RL3, RL4 and RL5 are each independently selected from the group consisting of H, halo, C1-8 alkyl, OC1-8 alkyl, SC1-8 alkyl, NHC1-8 alkyl, N(C1-8 alkyl)2, C3-11 cycloalkyl, aryl, heteroaryl, 3-6 membered heterocycloalkyl, OC1-8 cycloalkyl, SC1-8 cycloalkyl, NHC1-8 cycloalkyl, N(C1-8cycloalkyl)2, N(C1-8 cycloalkyl)(C1-8 alkyl), OH, NH2, SH, SO2C1-8 alkyl, P(O)(OC1-8 alkyl)(C1-8 alkyl), P(O)(OC1-8 alkyl)2, CC-C1-8 alkyl, CCH, CH=CH(C1-8 alkyl), C(C1-8 alkyl)=CH(C1-8 alkyl), C(C1-8 alkyl)=C(C1-8 alkyl)2, Si(OH)3, SiC(1-8 alkyl)3, Si(OH)(C1-8 alkyl)2, COC1-8 alkyl, CO2H, CN, haloC1-8 alkyl (e.g., CF3, CHF2, CH2F), NO2, SF5, SO2NHC1-8 alkyl, SO2NHC1-8 alkyl, SO2N(C1-8 alkyl)2, SONHC1-8 alkyl, SON(C1-8 alkyl)2, CONHC1-8 alkyl, CON(C1-8 alkyl)2, N(C1-8 alkyl)CONH(C1-8 alkyl), N(C1-8 alkyl)CON(C1-8 alkyl)2, NHCONH(C1-8 alkyl), NHCON(C1-8 alkyl)2, NHCONH2, N(C1-8 alkyl)SO2NH(C1-8 alkyl), N(C1-8 alkyl)SO2N(C1- 8 alkyl)2, NHSO2NH(C1-8 alkyl), NHSO2N(C1-8 alkyl)2 and NHSO2NH2; and wherein RL1 and RL2 each, independently may be linked to another A group to form a cycloalkyl and or heterocycloalkyl moiety that can be further substituted with 0-4 RL5 groups.

18. The compound according to claim 17, wherein q is an integer from 1 to 30, e.g., 5 to 25, or 5 to 20, or 5 to 15, or 10 to 20, or 5 to 10, or 10 to 15, or 7 to 12. 19. The compound according to claim 17 or 18, wherein the units A are selected from CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, , C3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3- 6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 RL1 and/or 0-6 RL2 groups. 20. The compound according to claim 1, wherein the linker L is selected from: , , . ., , , wherein m is from 0-10; , herein n is from 1-5 (e.g., 3 or 4); , wherein m is from 0-10; , wherein m is from 0-10;

, 0-10; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 0-6 (e.g., 3 or 4);

, wherein m is from 0-12 (e.g., 2-6); herein n is from 1-5 (e.g., 1 or 2); rein m is from 0-10; m is from 1-12 (e.g., 6-10); n m is from 2 to 8 (e.g., 4, 6, or 8); wherein m is from 2 to 8 (e.g., 4, 6, or 8); , herein n is from 1 to 4 (e.g., 2, 3, or 4);

, - .g., - .g., 3);

, , m is from 0-4 (e.g., 0); , , (e.g., 0); from 0-4 (e.g., 0);

, .., .., 0);

, 1 .g., 2 (e.g., 0); , .., from 0-4 (e.g., 0); , -5;

, , , .., ., 4, 5, or 6) and n is from 1-4 (e.g., 2);

, .g., , , from 1-4 (e.g., 2); or wherein the linker L is selected from: , wherein each F is a group selected from C3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3- 6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, pyridazinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with 0-6 RL1 and/or 0-6 RL2 groups, wherein RL1 and RL2 are each independently selected from the group consisting of H, halo, and C1-8 alkyl (e.g., CH3); and wherein a, b, and c are each independently integers from 0 to 10, e.g., 0 to 8, or 0 to 6, or any of 0, 1, 2, 3, 4, 5, 6, or 7. 21. The compound according to claim 20, wherein the linker L is selected from: erein m is from 0-10; m is from 1-12 (e.g., 6-10); wherein n is from 1-5 and m is from 1-6; wherein n is from 0-6 (e.g., 3 or 4); , wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-12 (e.g., 7-9); , wherein m1 is from 0-12 (e.g., 4, 5, or 6) and m2 wherein m1 is from 0-12 (e.g., 4, 5, or 6) and m2 , wherein m is from 0-12 (e.g., 4, 5, or 6) and n erein m is from 2-10, e.g., 4-8, or 2-4; , wherein n is from 1-5 (e.g., 3); 159

, w e e s o - ; or wherein F is independently selected from cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, pyridazinyl, and triazolyl, and wherein a, b and c are each independently integers selected from 0, 1, 2, 3 and 4. 22. A compound selected from the group consisting of: F F O O N S N O F F O O 3 N S N O

F S 23. A pharmaceutical composition comprising a compound according to any one of claims 1- 22, and a pharmaceutically acceptable carrier, additive and/or excipient. 24. A method of treating a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease or condition, said method comprising administering an effective amount of a compound according to any one of claims 1-22, to a patient in need thereof. 25. A method of degrading an androgen receptor in a cell, e.g., a mutated AR such as any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant, said method comprising administering an effective amount of a compound according to any one of claims 1-22, to such cell, e.g., a cancer cell. 26. A method of inducing apoptosis in a cell, e.g., a cancer cell, said method comprising administering an effective amount of a compound according to any one of claims 1-22, to such cell.

27. Use of a compound according to any one of claims 1-22 for the treatment a disease state or condition in a patient, wherein dysregulated protein activity is responsible for said disease or condition, or for degrading an androgen receptor in a cell, e.g., a mutated AR such as any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant, or for inducing apoptosis in a cell, e.g., a cancer cell.

AMENDED CLAIMS received by the International Bureau on 04 April 2024 (04.04.2024)

What is claimed is:

1. A compound having a chemical structure ARB-L-E3LB, wherein ARB is an AR binding moiety that does not bind to a ligand binding domain, E3LB is an E3 ligase binding moiety, and L is a linker coupling the AR binding moiety to the E3 ligase binding moiety, and wherein:

(A) the E3LB moiety has the following general structure: wherein: represents a bond that may be stereospecific ((R) or (S)), or non- stereospecific;

C is a 6-membered aryl or a 5- or 6-membered heteroaryl (e.g., pyridine, pyrimidine, pyridazine, or pyrazine) ring, each optionally substituted by one or more substituents R9;

Q is -CH- or -N-; each R2 and R3 is independently selected from H, halo, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy;

R4 is selected from H, OH, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), C1-6 alkyl-OCOOR6, C1-6 alkyl-OCONR5R7, CH2-heterocycloalkyl optionally substituted with R5, or benzyl optionally substituted with R5;

R5 and R7 are each independently a bond, H, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), or -C(=O)R6, wherein each of said alkyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); and R6 is independently selected from CONR5R7, OR5, NR5R7, SR5, SO2R5, SO2NR5R7, CR5R7, CR5NR5R7, aryl, heteroaryl, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), P(O)(OR5)R7, P(O)R5R7, OP(O)(OR5)R7, OP(O)R5R7, Cl, F, Br, I, CF3, CHF2, CH2F, CN, NR5SO2NR5R7, NR5CONR5R7, CONR5COR7, NR5C(=N-CN)NR5R7, C(=N- CN)NR5R7, NR5C(-N=CN)R7, NR5C(=C-NO2)NR5R7, SO2NR5COR7, NO2, CO2R5, C(C=N-OR5)R7, CR5, CR5R7, CCR5, S(C=O)(C=N-R5)R7, SF5, R5NR5R7, (R5O)nR7, or OCF3; wherein n is an integer from 1 to 10; and

R9is independently selected from H, halo (e.g., fluoro, chloro), hydroxy, -CONH2, - CONR2R3, -SONH2, -SONR2R3, -SO2NH2, -SO2NR2R3, -NHCOC1-3 alkyl (optionally substituted by 1 or more halo), -NR2COC1-3 alkyl (optionally substituted by 1 or more halo), -NR2SO2C1-3 alkyl (optionally substituted by 1 or more halo), -NR2SOC1-3 alkyl (optionally substituted by 1 or more halo), cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R9 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C3-6 cycloalkyl, C3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); and

(B) the AR binding moiety is selected from:

wherein:

A is an aryl (e.g., phenyl), heteroaryl (e.g., pyridyl), C3-7 cycloalkyl, or 3-10 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as morpholine) or a bridged bicyclic 7-12 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as a bridged morpholine), each of which is optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl);

B is an aryl (e.g., phenyl) or heteroaryl (e.g., pyridyl or imidazolyl) ring, optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); each R1 is independently selected from H, halo (e.g., fluoro), hydroxy, -CONH2, - CONR2R3, -SONH2, -SONR2R3, -SO2NH2, -SO2NR2R3, -NHCOCI-3 alkyl (optionally substituted by 1 or more halo), -NR2COC1-3 alkyl (optionally substituted by 1 or more halo), -NR2SO2C1-3 alkyl (optionally substituted by 1 or more halo), -NR2SOC1-3 alkyl (optionally substituted by 1 or more halo), cyano, C1-6 alkyl (e.g., methyl), C2-6 alkenyl (e.g., vinyl), C2-6 alkynyl (e.g., ethynyl), NR2R3, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), C3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R1 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C3-6 cycloalkyl, C3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl); and each R2 and R3 is independently selected from H, halo, C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy.

2. The compound of claim 1, wherein the E3LB moiety has a general structure selected from the following: wherein R4, R9, and L are as provided in Compound 1.1, and wherein each X is independently selected from C(R9) and -N-, each Y is independently selected from C(R9), -NH-, -N(R10)-, -O-, and -S-, and Z is -C- or -N-; wherein

R10 is selected from C1-6 alkyl (e.g., methyl), C3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), -C(=O)R6, - C(=O)OR6, or -C(=O)NR2R6, wherein each of said alkyl, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C1-6 alkyl (e.g., methyl), haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6 alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl).

3. The compound of claim 1, wherein ring C is a ring selected from phenyl, pyridine, pyrimidine, pyridazine, pyrazine, thiophene, thiazole, isothiazole, pyrrole, pyrazole, imidazole, furan, oxazole, isoxazole, triazole, oxadiazole, thiadiazole, and tetrazole, optionally substituted by one or more groups R9.

4. The compound of claim 3, wherein ring C is phenyl, pyridine, pyrimidine, pyridazine, or pyrazine, optionally substituted by one or more group R9.

5. The compound of claim 3, wherein ring C is selected from furan, thiophene, pyrrole, pyrazole, thiazole, oxazole, imidazole, isothiazole, isoxazole, triazole, thiadiazole, or oxadiazole, each optionally substituted by one or more groups R9.

6. The compound of claim 1, wherein the E3LB moiety is a moiety selected from any of the following: wherein X is -NH-, N(R10), -0-, or -S-.

7. The compound of claim 1, wherein the E3LB moiety is a moiety is selected from any of the following:

8. The compound of claim 1, wherein the E3LB moiety is:

9. The compound of any preceding claim, wherein each R9 is independently selected from

H, halo (e.g., fluoro, chloro or bromo), hydroxy, CN, C1-6 alkyl, NH2, haloC1-6 alkyl (e.g., CH2F, CHF2, CF3), C1-6alkoxy (e.g., methoxy), or C3-6 cycloalkyl (e.g., cyclopropyl), and wherein R4 is H or C1-3 alkyl (e.g., methyl).

10. The compound of any preceding claim, wherein R4 and R9 are H.

11. The compound of claim 1, wherein the E3LB moiety is:

12. The compound of claim 1, wherein the AR binding moiety is selected from: wherein A, B, R1, R2, and R3 are as defined in claim 1.

13. The compound according to claim 12, wherein A is: wherein X is -CH- or -N-, or wherein A is wherein each of Q1, Q2, and Q3 are independently selected from O, S, NR2, C1-3 alkyl (e.g., CH2 or CH2CH2) optionally substituted by one or more R1, or wherein Q1 or Q2 is absent; and wherein B is selected from:

14. The compound according to claim 1, wherein the AR binding moiety is selected from:

15. The compound according to claim 1, wherein the AR binding moiety is selected from:

16. The compound according to claim 1, wherein the AR binding moiety is:

17. The compound according to claim 1, wherein the linker (“L”) consists of a chemical structural unit represented by the formula -Aq-, in which q is an integer greater than 1, and each A is independently selected from the group consisting of: a bond, CRL1RL2, O, s, so, so2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, C≡C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(=NCN)NRL4, NRL3C(=NCN), NRL3C(=CNO2)NRL4, C3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 RL1 and/or 0-6 RL2 groups; and wherein: RL1 RL2, RL3, RL4 and RL5 are each independently selected from the group consisting of H, halo, Ci-8 alkyl, OC1-8 alkyl, SC1-8 alkyl, NHC1-8 alkyl, N(C1-8 alkyl)2, C3-11 cycloalkyl, aryl, heteroaryl, 3-6 membered heterocycloalkyl, OC1-8 cycloalkyl, SC1-8 cycloalkyl, NHC1-8 cycloalkyl, N(C1-8cycloalkyl)2, N(C1-8 cycloalkyl)(C1-8 alkyl), OH, NH2, SH, SO2C1-8 alkyl, P(O)(OC1-8 alkyl)(C1-8 alkyl), P(O)(OC1-8 alkyl)2, CC-C1-8 alkyl, CCH, CH=CH(C1-8 alkyl), C(C1-8 alkyl)=CH(C1-8 alkyl), C(C1-8 alkyl)=C(C1-8 alkyl)2, Si(OH)3, SiC(1-8 alkyl)3, Si(OH)(C1-8 alkyl)2, COC1-8 alkyl, C02H, CN, haloC1-8 alkyl (e.g., CF3, CHF2, CH2F), NO2, SF5, SO2NHC1-8 alkyl, SO2NHC1-8 alkyl, SO2N(C1-8 alkyl)2, SONHC1-8 alkyl, SON(C1-8 alkyl)2, CONHC1-8 alkyl, C0N(C1-8 alkyl)2, N(C1-8 alkyl)CONH(C1-8 alkyl), N(C1-8 alkyl)CON(C1-8 alkyl)2, NHC0NH(C1-8 alkyl), NHC0N(CI-8 alkyl)2, NHCONH2, N(C1-8 alkyl)SO2NH(C1-8 alkyl), N(C1-8 alkyl)SO2N(C1- 8 alkyl)2, NHSO2NH(C1-8 alkyl), NHSO2N(C1-8 alkyl)2 and NHSO2NH2; and wherein RL1 and RL2 each, independently may be linked to another A group to form a cycloalkyl and or heterocycloalkyl moiety that can be further substituted with 0-4 RL5 groups.

18. The compound according to claim 17, wherein q is an integer from 1 to 30, e.g., 5 to 25, or 5 to 20, or 5 to 15, or 10 to 20, or 5 to 10, or 10 to 15, or 7 to 12.

19. The compound according to claim 17 or 18, wherein the units A are selected from CRL1RL2, O, S, SO, SO2, NRL3, SO2NR1 3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, C≡C, C3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3- 6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 RL1 and/or 0-6 RL2 groups.

20. The compound according to claim 1 , wherein the linker L is selected from: wherein n is from 1-5; , wherein m is from 2 to 8 (e.g., 4, 6, or 8); wherein n is from 1 to 4 (e.g., 2, 3, or 4); , wherein n is from 1 to 4 (e.g., 2, 3, or 4); wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-4 (e.g., 2 or

3); wherein n is from 0-4 (e.g., 0, 1 or 2) and m is from 0-10

, wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-4 (e.g., 2 or , wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-4 (e.g., 2 or

3); wherein n is from 0-4 (e.g., 1) and m is from 0-4 (e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from

0-4 (e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from 0-4 wherein n is from 0-4 (e.g., 1) and m is from 0-4

, wherein n is from 0-4 (e.g., 1) and m is from 0-4 wherein n is from 0-4 (e.g., 1) and m is from 0-4

(e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from 0-4 (e.g., 0); wherein n is from 0-4 (e.g., 1) and m is from 0-4

(e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from 0-4

(e.g., 0); . . , wherein n is from 0-4 (e.g., 1) and m is from 0-4 (e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from 0-4 (e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from 0-4 (e.g., 0); , wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., 0); , wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from

0-4 (e.g., 0); , wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4

(e.g., 0); , wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g.,

0); wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4

(e.g., 0); , wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., 0); wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4

(e.g., 0); , wherein mi is from 0-12 (e.g., 4, 5, or 6) and m2 is from

0-4 (e.g., 2); wherein mi is from 0-12 (e.g., 4, 5, or 6) and m2 is from

0-4 (e.g., 2); wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4

(e.g., 0); , wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., 0); wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., 0);

O wherein mi is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., 0); , wherein n is from 1-5; , wherein m is from 1-12; wherein n is from 1-5; wherein n is from 1-5; , wherein m is from 0-12 (e.g., 8); wherein mi is from 0-12 (e.g., 4, 5, or 6) and m2 is from , wherein n is from 1-5 (e.g., 3 or 4); wherein n is from 1-5 (e.g., 3 or 4); , wherein m is from 0-10; wherein n is from 0-4 (e.g., 0, 1 or 2) and m is from 0-10

(e.g., 4, 6, or 8); herein n is from 0-6 (e.g., 3 or 4); , wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 and m is from 1-6; , wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e.g., 2); , wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e g 2); wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e.g., 2); wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e.g., 2); wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e.g., 2); and wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e.g., 2); or wherein the linker L is selected from:

wherein each F is a group selected from C3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3- 6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, pyridazinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with 0-6 RL1 and/or 0-6 RL2 groups, wherein RL1 and RL2 are each independently selected from the group consisting of H, halo, and C1-8 alkyl (e.g., CH3); and wherein a, b, and c are each independently integers from 0 to 10, e.g., 0 to 8, or 0 to 6, or any of 0, 1, 2, 3, 4, 5, 6, or 7.

21. The compound according to claim 20, wherein the linker L is selected from: wherein m is from 4-8; wherein n is from 1 to 4 (e.g., 2, 3, or 4); , wherein n is from 1-5; , wherein m is from 0-12 (e.g., 2-6); wherein n is from 1-5 (e.g., 1 or 2); , wherein m is from 0-10; , wherein m is from 1-12 (e.g., 6-10); wherein n is from 1-5 and m is from 1-6; wherein n is from 0-6 (e.g., 3 or 4); , wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-12

(e.g., 7-9); wherein mi is from 0-12 (e.g., 4, 5, or 6) and m2 is from 0-4 (e.g., 2); wherein mi is from 0-12 (e.g., 4, 5, or 6) and m2 is from 0-4 (e.g., 2); wherein m is from 0-12 (e.g., 4, 5, or 6) and n is from 1-4 (e.g., 2); wherein m is from 2-10, e.g., 4-8, or 2-4; , wherein n is from 1-5 (e.g., 3); , wherein n is from 1-5 (e.g., 3); wherein m is from 2-10 (e.g., 8 or 9); and , wherein m is from 1-12; or wherein F is independently selected from cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, pyridazinyl, and triazolyl, and wherein a, b and c are each independently integers selected from 0, 1, 2, 3 and 4.

22. A compound selected from the group consisting of:

23. A pharmaceutical composition comprising a compound according to any one of claims 1-

22, and a pharmaceutically acceptable earner, additive and/or excipient.

24. A method of treating a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease or condition, said method comprising administering an effective amount of a compound according to any one of claims 1 -22, to a patient in need thereof.

25. A method of degrading an androgen receptor in a cell, e.g., a mutated AR such as any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant, said method comprising administering an effective amount of a compound according to any one of claims 1 -22, to such cell, e.g., a cancer cell.

26. A method of inducing apoptosis in a cell, e.g., a cancer cell, said method comprising administering an effective amount of a compound according to any one of claims 1 -22, to such cell.

27. Use of a compound according to any one of claims 1-22 for the treatment a disease state or condition in a patient, wherein dysregulated protein activity is responsible for said disease or condition, or for degrading an androgen receptor in a cell, e.g., a mutated AR such as any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant, or for inducing apoptosis in a cell, e.g., a cancer cell.

Description:
BI-FUNCTIONAL COMPOUNDS AND METHODS FOR TARGETED UBIQUITINATION OF ANDROGEN RECEPTOR CROSS-REFERENCE TO RELATED APPLICATIONS This is a PCT international application which claims priority to, and the benefit of, U.S. Provisional Application Ser. No.63/382,832, filed on November 8, 2022, the contents of which are hereby incorporated by reference in its entirety. FIELD OF THE INVENTION [001] This invention relates to therapeutic compounds and compositions, and methods for their use in the treatment of various indications, including various cancers. In particular, the invention relates to therapies and methods of treatment for cancers such as prostate cancer. BACKGROUND OF THE INVENTION [002] Prostate cancer is the most commonly diagnosed malignancy in males in the United States and the second leading cause of male cancer mortality. Numerous studies have shown that the androgen receptor (AR) is central not only to the development of prostate cancer, but also the progression of the disease to the castration resistance state (Taplin, M. E. et al., J. Clin. Oncol. 200321:2673-8; and Tilley, W. D. et al., Cancer Res.199454:4096-4102). Thus, effective inhibition of human AR remains one of the most effective therapeutic approaches to the treatment of advanced, metastatic prostate cancer. [003] Androgens are also known to play a role in female cancers. One example is ovarian cancer where elevated levels of androgens are associated with an increased risk of developing ovarian cancer (Helzlsouer, K.J. et al., JAMA 1995274, 1926-1930; Edmondson, R. J. et al., Br. J. Cancer 200286, 879-885). Moreover, AR has been detected in a majority of ovarian cancers (Risch, H. A., J. Natl. Cancer Inst.199890, 1774-1786; Rao, B. R. et al., Endocr. Rev.199112, 14-26; Clinton, G. M. et al., Crit. Rev. Oncol. Hematol.199725, 1-9). [004] AR belongs to the nuclear hormone receptor family that is activated by androgens such as testosterone and dihydrotestosterone. These androgens, as well as antagonists such as enzalutamide, compete with the androgens that bind to the ligand binding domain (LBD). AR possesses a modular organization characteristic of all nuclear receptors. It is comprised of an N- terminal domain (NTD), a central DNA binding domain (DBD), a short hinge region, and a C- terminal domain that contains a hormone ligand binding pocket (the LBD, which also comprises the hormone binding site (HBS)) and the Activation Function-2 (AF2) site (Gao, W. Q. et al., Chem. Rev.2005105:3352-3370). The latter represents a hydrophobic groove on the AR surface which is flanked with regions of positive and negative charges – “charge clamps” – that are significant for binding AR activation factors (Zhou, X. E. et al., J. Biol. Chem.2010285:9161- 9171). [005] The activation of AR follows a well characterized pathway: in the cytoplasm, the receptor is associated with chaperone proteins that maintain agonist binding conformation of the AR (Georget, V. et al., Biochemistry 200241:11824-11831). Upon binding of an androgen, the AR undergoes a series of conformational changes, disassociation from chaperones, dimerization, and translocation into the nucleus (Fang, Y. F. et al., J. Biol. Chem.1996271:28697-28702; and Wong, C. I. et al., J. Biol. Chem.1993268:19004-19012) where it further interacts with co- activator proteins at the AF2 site (Zhou, X. E. et al. J. Biol. Chem.2010285:9161-9171). This event triggers the recruitment of RNA polymerase II and other factors to form a functional transcriptional complex with the AR. [006] In castration-resistant prostate cancer (CRPC), drug resistance can manifest through AR-LBD mutations that convert AR-antagonists into agonists or by expression of AR-variants lacking the LBD. AR is a major driver of prostate cancer and inhibition of its transcriptional activity using competitive antagonists such as enzalutamide and apalutamide remains a frontline therapy for prostate cancer management. Another therapy is abiraterone which is an inhibitor of cytochrome P45017A1 that impairs AR signaling by depleting adrenal and intratumoral testosterone and dihydrotestosterone. Recent work (Antonarakis, E.S. et al., New Engl. J. Med. 201437, 1028-1038) has shown that patients on enzalutamide and abiraterone with a splice variant of AR, labelled as AR-V7, had lower PSA response rates, shorter PSA progression-free survival, and shorter overall survival. [007] AR-V7 lacks the LBD, which is the target of enzalutamide and testosterone, but AR-V7 remains constitutively active as a transcription factor. Accordingly, it is desirable to investigate other approaches to antagxnize the AR receptor, in particular the AR-V7 splice variant. The common domain between these two proteins is the DBD and compounds have been identified as discussed in Li, H. et al., J. Med. Chem.201457, 6458-6467 (2014); Dalal, K. et al., Mol. Cancer Ther.2017 vol.16, 2281-2291; Xu, R. et al., Chem. Biol. & Drug Design 201891(1), 172-180; and WO 2015/120543. [008] Several methods are available for the manipulation of protein levels, including bi- functional proteolysis targeting chimeric molecules (PROTACs) which contain a ligand that recognizes the target protein that is linked to a ligand that binds to a specific E3 ubiquitin ligase. The ensuing bifunctional molecule binds to the target protein and the E3 ligase enabling the transfer of ubiquitin to the target protein from the ligase provided there is a suitable acceptor on the target protein. Another method is the “molecular glue” process whereby the molecule together with the E3 ligase recruit the target protein to the E3 ligase followed by the ubiquitin transfer and degradation of the target (Chopra, R., Sadok, A., Collins, I., Drug Disc Today: Technologies, 2019, 31, 5-13.) In the case of a compound acting as a “molecular glue,” the only requirement is the presence of an E3 ligase binding moiety. After binding to the E3 ligase, the ensuing moiety could recruit the protein to be degraded. The labelling of proteins with ubiquitin is implicated in the protein’s turnover by the 26S proteasome. [009] Protein ubiquitination is a multi-step process whereby a ubiquitin protein is successively relayed between different classes of enzymes (E1, E2, E3) in order to eventually tag a cellular substrate. Initially, the C-terminal carboxylate of ubiquitin is adenylated by the E1 activating enzyme in an ATP-dependent step. Subsequently, a conserved nucleophilic cysteine residue of the E1 enzyme displaces the AMP from the ubiquitin adenylate resulting in a covalent ubiquitin thioester conjugate. The binding and ensuing adenylation of a second ubiquitin molecule promotes the recruitment of an E2 conjugating enzyme to this ternary complex. An active site Cys on the E2 subsequently facilitates the transfer of the covalently linked ubiquitin from the E1 to a Cys residue on the E2 through a trans-thioesterification reaction. Concomitantly, an E3 ligase recruits a specific downstream target protein and mediates the transfer of the ubiquitin from the E2 enzyme to the terminal substrate through either a covalent or non-covalent mechanism. Each ubiquitin is ligated to a protein through either a peptide bond with the N-terminal amino group or an isopeptide bond formed between a side chain ε-amino group of a select Lys residue on the target protein and the ubiquitin. [0010] Deubiquitinating enzymes (DUBs) are enzymes that specifically cleave the ubiquitin protein from the substrate thereby offering additional mechanisms of regulation over the entire labeling pathway. In the current human proteome, there are eight known human E1s, about 40 E2s, over 600 E3s and over 100 DUBs. These enzymes are well described in Pavia, S. et al., J. Med. Chem.201861(2), 405-421. [0011] The E3 ligases originate in three major classes – the RING finger and U-box E3s, the HECT E3s, and the RING/HECT-hybrid type E3s. The E3 ligases are localized in various cell organelles and hence the effectiveness of the E3 ligase ligand depends at least in part on the location of the protein targeted for degradation, assuming that the full molecule is available within the appropriate location in the cell. In addition, for every combination of the target ligand and the ubiquitin recruiting ligand, the linker length and conformational flexibility also contributes to the effectiveness of the degradation molecule. The mechanism depends on the availability of a Lys residue on the surface of the protein close to the targeted protein ligand binding pocket. Ubiquitin binds at Lys residues and hence the “delivery” of ubiquitin for binding at the appropriate Lys influences the effectiveness of the degradation molecule. Crew et al. (US20170327469A1, US20180099940A1) proposed a treatment for castration-resistant prostate cancer based on bifunctional molecules coupling various E3 ligases to AR antagonists binding at the AR LBD site. The approach in the present application is different in that instead of targeting the LBD site of the AR, the DBD site of the AR is targeted, and correspondingly, the chemical matter is quite different. [0012] There exists a continuing need for effective treatments for diseases and conditions that are related to aberrant AR regulation or activity, for example, cancers such as prostate cancer, and Kennedy’s Disease. In developing such treatments, it would be desirable to have a molecule which can simultaneously bind AR and an E3 ubiquitin ligase and which also promotes ubiquitination of AR-V7 and perhaps AR, and leads to degradation of AR-V7 and AR by the proteasome. Some compounds of this type have been described, for example, by the present Applicant in: US 2020/0239430, US 2020/0282068, US 2023/0132823, US 2023/0134817, U.S. 11,098,025, U.S.11,787,778, U.S.11,547,759, WO 2021/236695, WO 2023//039601, WO 2023/039602, WO 2023/039603, and WO 2023/039604, the contents of each of which are hereby incorporated by reference in their entireties. [0013] Bi-functional compounds which function to recruit endogenous proteins to an E3 ubiquitin ligase for degradation, and methods for using same, are known. More specifically, specific proteolysis targeting chimera (PROTAC) molecules which find utility as modulators of targeted ubiquitination of a variety of polypeptides and other proteins, such as AR, which are then degraded and/or otherwise inhibited by the compounds as described herein have been described, for example, by the present Applicant in: US 2020/0239430, US 2020/0282068, US 2023/0132823, US 2023/0134817, U.S.11,098,025, U.S.11,787,778, U.S.11,547,759, WO 2021/236695, WO 2023//039601, WO 2023/039602, WO 2023/039603, and WO 2023/039604, the contents of each of which are hereby incorporated by reference in their entireties. There remains a need for improved bi-functional PROTAC compounds having superior pharmaceutical properties, such as activity, oral bioavailability, and metabolic stability. SUMMARY OF THE INVENTION [0014] The present disclosure provides new proteolysis targeting chimera (PROTAC) molecules which find utility as modulators of targeted ubiquitination of a variety of polypeptides and other proteins, such as AR, which are then degraded and/or otherwise inhibited by the compounds as described herein. [0015] These PROTAC molecules comprise an E3 ubiquitin ligase binding moiety (i.e., a ligand for an E3 ubiquitin ligase) linked to a moiety that binds the target protein (i.e., a protein/polypeptide targeting ligand) such that the target protein/polypeptide is placed in proximity to the ubiquitin ligase to effect degradation (and/or inhibition) of that protein, wherein the E3LB moiety is a novel heterocycloalkyl-substituted thalidomide-based IMiD moiety which binds to the Androgen Receptor. In addition, the description provides methods for using an effective amount of the compounds described herein for the treatment or amelioration of a disease condition including cancer, e.g., prostate cancer, and Kennedy’s Disease. [0016] Known ligands that bind to the E3 ubiquitin ligase include cereblon binders such as immunomodulatory imide drugs (IMiDs) including thalidomide, pomalidomide, and lenalidomide (Deshales, R.J., Nature Chem Biol.201511, 634-635), and some analogs or derivatives thereof. These IMiDs act as “molecular glues” and therefore have been shown to recruit a different set of proteins for degradation. Other known E3 ubiquitin ligase binders are the E3 CRL2 VHL compounds, also called Von-Hippel-Lindau or VHL ligands, the cellular inhibitor of apoptosis protein (IAP), as discussed in Shibata, N. et al., J. Med. Chem., 201861(2), 543- 575, and binders of the E3 ligase Mouse Double Minute 2 (MDM2), as discussed in Skalniak, L., et al., Expert Opin. Ther, Patents, 2019, 29, 151-170. [0017] The known IMiD moieties share a common isoindoline core, as shown in the parent immunomodulatory drugs: [0018] However, there remains a need for PROTAC compounds with improved IMiD moieties. [0019] In one aspect, the present disclosure provides new PROTAC compounds which function to recruit proteins, including AR-V7 and AR, for targeted ubiquitination and degradation, wherein the compounds have a structure which can be depicted as: ARB – E3LB or ARB – L – E3LB wherein ARB is an AR binding moiety, L is a chemical linker moiety, and E3LB is an E3-ligase binding moiety having the following general structure: , wherein R 4 , R 9 , C, Q, and L are as provided herein. This E3LB moiety is a non-indoline cereblon (CRBN) binding moiety. [0020] In an additional aspect, the present disclosure provides therapeutic compositions comprising an effective amount of a compound as described herein or pharmaceutically acceptable salt form thereof, and one or more pharmaceutically acceptable carriers. The therapeutic compositions modulate protein degradation in a patient or subject, for example, an animal such as a human, and can be used for treating or ameliorating disease states or conditions which are modulated through the degraded protein. In certain embodiments, the therapeutic compositions as described herein may be used to effectuate the degradation and/or inhibition of proteins of interest for the treatment or amelioration of a disease, e.g., cancer. [0021] In another aspect, the present disclosure provides a method of ubiquitinating/degrading a target protein (i.e., AR, such as splice variant AR-V7) in a cell. In certain embodiments, the method comprises administering a bi-functional compound, or pharmaceutically acceptable salt form thereof, as described herein, such that degradation of the target protein (i.e., AR, such as splice variant AR-V7) occurs when the target protein is placed in proximity to the ubiquitin ligase, thus resulting in degradation/inhibition of the effects of the target protein and the control of protein levels. The control of protein levels afforded by the present disclosure provides treatment of a disease state or condition, which is modulated through the target protein by lowering the level of that protein in the cells of a patient. [0022] In another aspect, the present disclosure provides methods for treating or ameliorating a disease, disorder or symptom thereof in a subject or a patient, e.g., an animal such as a human, comprising administering to a subject in need thereof a pharmaceutical composition comprising an effective amount, e.g., a therapeutically effective amount, of a compound as described herein or pharmaceutically acceptable salt form thereof, and a pharmaceutically acceptable carrier, wherein the composition is effective for treating or ameliorating the disease or disorder or symptom thereof in the subject. [0023] In another aspect, the present disclosure provides methods for identifying the effects of the degradation of proteins of interest in a biological system using compounds according to the present disclosure. [0024] The preceding general areas of utility are given by way of example only and are not intended to be limiting on the scope of the present disclosure and appended claims. Additional objects and advantages associated with the compositions, methods, and processes of the present disclosure will be appreciated by one of ordinary skill in the art in light of the instant claims, description, and examples. For example, the various aspects and embodiments of the invention may be utilized in numerous combinations, all of which are expressly contemplated by the present description. These additional advantages objects and embodiments are expressly included within the scope of the present disclosure. The publications and other materials used herein to illuminate the background of the invention, and in particular cases, to provide additional details respecting the practice, are incorporated by reference. Where applicable or not specifically disclaimed, any one of the embodiments described herein are contemplated to be able to combine with any other one or more embodiments, even though the embodiments are described under different aspects of the disclosure. DETAILED DESCRIPTION [0025] The following is a detailed description provided to aid those skilled in the art in practicing the invention of the present disclosure. Those of ordinary skill in the art may make modifications and variations in the embodiments described herein without departing from the spirit or scope of the present disclosure. All publications, patent applications, patents, figures and other references mentioned herein are expressly incorporated by reference in their entirety. [0026] The present disclosure relates to bi-functional PROTAC compounds which facilitate E3 ubiquitin ligase-mediated ubiquitination of an androgen receptor target protein, such as, in particular, the androgen receptor of a splice variant of AR which lacks the LBD, known as AR- V7. Accordingly, the present description provides compounds, compositions comprising the same, and associated methods of use for ubiquitination and degradation of a chosen target protein, e.g., androgen receptor, including AR-V7. [0027] In one aspect, the present disclosure provides new PROTAC compounds comprising an E3 ubiquitin ligase binding moiety (“E3LB”), a moiety that binds a target protein (i.e., a protein/polypeptide targeting ligand) that is an AR binding moiety (“ARB”), and optionally a chemical linker (“L”) between the E3LB and the ARB. The structure of the bi-functional compound can be depicted as: ARB-E3LB or ARB–L–E3LB where ARB is an AR binding moiety as described herein, L is a chemical linker moiety as described herein, and E3LB is an E3 ligase binding moiety having the following general structure: , wherein R 4 , R 9 , C, Q, and L are as provided hereinbelow. [0028] The respective positions of the ARB and E3LB moieties as well as their number as illustrated herein is provided by way of example only and is not intended to limit the compounds in any way. As would be understood by the skilled artisan, the bi-functional compounds as described herein can be synthesized such that the number and position of the respective functional moieties can be varied as desired. [0029] It will be understood that the general structures are exemplary and the respective moieties can be arranged spatially in any desired order or configuration, e.g., ARB–L–E3LB, and E3LB–L–ARB, respectively. The E3LB group and ARB group may be covalently linked to the linker group through any covalent bond which is appropriate and stable to the chemistry of the linker. It will be further understood that for all compounds described herein, one or more hydrogen atoms may be replaced with an equivalent number of deuterium atoms. [0030] In certain embodiments, the ARB may be selected from the following structures, wherein L is the optional linker (e.g., in the formula ARB—L—E3LB): [0031] In each of ARB-a, ARB-b, and ARB-c, the following definitions apply: [0032] L is the linker moiety, as provided hereinbelow; [0033] A is an aryl, heteroaryl, C 3-7 cycloalkyl, 3-10 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as morpholine), or a bridged bicyclic 7-12 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as a bridged morpholine), each of which is optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 (e.g., N,N-dimethyl), halo C 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl (e.g., phenyl), heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); [0034] B is an aryl or heteroaryl ring, each optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 , haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); and [0035] each R 1 is independently selected from H, halo (e.g., fluoro), hydroxy, -CONH 2 , - CONR 2 R 3 , -SONH 2 , -SONR 2 R 3 , -SO 2 NH 2 , -SO 2 NR 2 R 3 , -NHCOC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 COC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SO 2 C 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SOC 1-3 alkyl (optionally substituted by 1 or more halo), cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 , haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R 1 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); and [0036] each R 2 and R 3 is independently selected from H, halo, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy. [0037] In some embodiments, A is selected from the following: wherein R 1 is as described above and X is selected from -CH- or -N-. [0038] In some embodiments, A has the general structure: ; wherein R 1 is as described above, and wherein each of Q 1 , Q 2 , and Q 3 are independently selected from O, S, NR 2 , C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 ) optionally substituted by one or more R 1 , or wherein Q 1 or Q 2 is absent (in such case the missing valences of the ring atoms is filled by H atoms). [0039] In some embodiments, B is selected from the following:

wherein L is the chemical linker and R 1 is as described above. [0040] The chemical linker group (L) comprises a chemical structural unit represented by the formula -A q- , in which q is an integer greater than 1; and A is independently selected from the group consisting: of a bond, CR L1 R L2 , O, S, SO, SO 2 , NR L3 , SO 2 NR L3 , SONR L3 , CONR L3 , NR L3 CONR L4 , NR L3 SO 2 NR L4 , CO, CR L1 =CR L2 , , SiR L1 R L2 , P(O)R L1 , P(O)OR L1 , NR L3 C(=NCN)NR L4 , NR L3 C(=NCN), NR L3 C(=CNO 2 )NR L4 , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups; wherein R L1 , R L2 , R L3 , R L4 and R L5 are each independently selected from the group consisting of H, halo, C 1-8 alkyl, OC 1-8 alkyl, SC 1-8 alkyl, NHC 1-8 alkyl, N(C 1-8 alkyl) 2 , C 3-11 cycloalkyl, aryl, heteroaryl, 3-6 membered heterocycloalkyl, OC 1-8 cycloalkyl, SC 1-8 cycloalkyl, NHC 1-8 cycloalkyl, N(C 1-8 cycloalkyl) 2 , N(C 1-8 cycloalkyl)(C 1-8 alkyl), OH, NH 2 , SH, SO 2 C 1-8 alkyl, P(O)(OC 1-8 alkyl)(C 1-8 alkyl), P(O)(OC 1-8 alkyl) 2 , CC-C 1-8 alkyl, CCH, CH=CH(C 1-8 alkyl), C(C 1-8 alkyl)=CH(C 1-8 alkyl), C(C 1-8 alkyl)=C(C 1-8 alkyl) 2 , Si(OH) 3 , SiC( 1-8 alkyl) 3 , Si(OH)(C 1-8 alkyl) 2 , COC 1-8 alkyl, CO 2 H, CN, haloC 1-8 alkyl (e.g., CF 3 , CHF 2 , CH 2 F), NO 2 , SF 5 , SO 2 NHC 1-8 alkyl, SO 2 NHC 1-8 alkyl, SO 2 N(C 1-8 alkyl) 2 , SONHC 1-8 alkyl, SON(C 1-8 alkyl) 2 , CONHC 1-8 alkyl, CON(C 1-8 alkyl) 2 , N(C 1-8 alkyl)CONH(C 1-8 alkyl), N(C 1-8 alkyl)CON(C 1-8 alkyl) 2 , NHCONH(C 1-8 alkyl), NHCON(C 1-8 alkyl) 2 , NHCONH 2 , N(C 1-8 alkyl)SO 2 NH(C 1-8 alkyl), N(C 1-8 alkyl)SO 2 N(C 1- 8 alkyl) 2 , NHSO 2 NH(C 1-8 alkyl), NHSO 2 N(C 1-8 alkyl) 2 and NHSO 2 NH 2 . [0041] In some embodiments, R L1 and R L2 each, independently can be linked to another A group to form a cycloalkyl and or heterocycloalkyl moiety that can be further substituted with 0- 4 R L5 groups. [0042] In some embodiments, q is an integer from 1 to 30, e.g., 5 to 25, or 5 to 20, or 5 to 15, or 10 to 20, or 5 to 10, or 10 to 15, or 7 to 12. [0043] In some embodiments, the linker moiety is preferably a flexible linker moiety, as opposed to a semi-rigid or rigid linker moiety. In other embodiments, the linker moiety is preferably a semi-rigid or rigid linker moiety. [0044] It is understood that an essentially linear chain, including a linear chain with branches, such as a linear chain comprising saturated and optionally substituted -C-, -O-, -N-, and -S- atoms is highly flexible when all bonds forming the chain are sp 2 -hybridized atoms. For example, the following are highly flexible examples of linker chains: [0045] Such linker chains lose their flexibility, becoming more rigid, by the incorporation of either rings or sp- or sp 2 -hybridized atoms, or combinations thereof. Rings (aromatic and non- aromatic) and double and triple bonds, when provided within the linker chain (not as a substituent attached to the chain) are rigid groups, which inhibit free rotation and flexibility of the linker chain L. Thus, for example, the following are examples of rigid and semi-rigid linker chains:

. [0046] Therefore, in some embodiments wherein the linker is a semi-rigid or rigid linker, the linker L comprises a chemical structural unit represented by the formula -A q- , in which q is an integer greater than 1; and L comprises at least one moiety A selected from CR L1 =CR L2 , , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups, as provided above. [0047] In some embodiments wherein the linker moiety L is a flexible linker, the linker L comprises a structural unit represented by the formula -A q -, in which q is an integer greater than 1; and L does not comprise any moieties selected from CR L1 =CR L2 , , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl. [0048] Ring C is an aryl or heteroaryl ring, each optionally substituted by one or more substituents R 9 , for example, a phenyl, pyridine, pyrimidine, pyridazine, pyrazine, thiophene, thiazole, isothiazole, pyrrole, pyrazole, imidazole, furan, oxazole, isoxazole, triazole, oxadiazole, thiadiazole, or tetrazole. Preferably, ring C is a phenyl ring or a 6-membered heteroaromatic ring (e.g., pyridine, pyrimidine, pyridazine, or pyrazine). [0049] Q is -CH- or -N-. [0050] In some embodiments, the E3LB moiety is a moiety having the following general structure (E3LB-y1): [0051] In some embodiments, the E3LB moiety is a moiety having the following general structure (E3LB-y2): wherein each X is independently selected from C(R 9 ) and -N-. [0052] In some embodiments, the E3LB moiety is a moiety having the following general structure (E3LB-y3): wherein each Y is independently selected from C(R 9 ), -NH-, -N(R 10 )-, -O-, and -S-, and Z is -C- or -N-. [0053] In particular embodiments, ring C may be a C-linked 6-membered heterocycle or 5- membered heterocycle (i.e., the linker L attaches to ring C via a carbon atom of ring C), for example, the E3LB moiety is a moiety selected from:

wherein X is -NH-, N(R 10 ), -O-, or -S-. In some embodiments said ring C may be substituted by 0 groups R 9 , 1 group R 9 , or two groups R 9 , or three groups R 9 , or four groups R 9 , as permitted by the structure of ring C. For example, the skilled artisan would recognize that, in the above structures, if ring C is a phenyl, it may be substituted by 0, 1, 2, 3 or 4 groups R 9 ; whereas, if ring C is a pyridine, it may be substituted by 0, 1, 2 or 3 groups R 9 ; whereas, if ring C is a pyrimidine, pyridazine, pyrazine, furan, thiophene, or pyrrole, it may be substituted by 0, 1, or 2 groups R 9 ; whereas, if ring C is a pyrazole, thiazole, oxazole, imidazole, isothiazole, or isoxazole, it may be substituted by 0, or 1 groups R 9 ; and, whereas, if ring C is a triazole, thiadiazole, or oxadiazole, it must be unsubstituted by groups R 9 . [0054] In particular embodiments, ring C may be an N-linked 6-membered heterocycle or 5- membered heterocycle (i.e., the linker L attaches to ring C via a nitrogen atom of ring C), for example, the E3LB moiety is a moiety selected from: In some embodiments said ring C may be substituted by 0 groups R 9 , 1 group R 9 , or two groups R 9 , or three groups R 9 , or four groups R 9 , as permitted by the structure of ring C. For example, the skilled artisan would recognize that, in the above structures, if ring C is a pyrrole, it may be substituted by 0, 1, 2 or 3 groups R 9 ; whereas, if ring C is a pyrazole or imidazole, it may be substituted by 0, 1, or 2 groups R 9 ; whereas, if ring C is a triazole, it may be substituted by 0, or 1 groups R 9 ; and, whereas, if ring C is a tetrazole, it must be unsubstituted by groups R 9 . [0055] In each of the above E3LB structures, L is the optional linker, and the following definitions apply: [0056] “---” represents a bond that may be a single bond or a double bond, as permitted by the valencies of the atoms attached to the bond; [0057] “ ”represents a bond that may be stereospecific ((R) or (S)), or non- stereospecific; [0058] R 4 is selected from H, OH, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), C 1-6 alkyl-OCOOR 6 , C 1-6 alkyl-OCONR 5 R 7 , CH 2 -heterocycloalkyl optionally substituted with R 5 , or benzyl optionally substituted with R 5 ; [0059] Each R 9 is independently selected from H, halo (e.g., fluoro, chloro), hydroxy, - CONH 2 , -CONR 2 R 3 , -SONH 2 , -SONR 2 R 3 , -SO 2 NH 2 , -SO 2 NR 2 R 3 , -NHCOC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 COC 1-3 alkyl (optionally substituted by 1 or more halo), - NR 2 SO 2 C 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SOC 1-3 alkyl (optionally substituted by 1 or more halo), cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 , haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R 9 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl). [0060] R 10 is selected from H, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), -C(=O)R 6 , - C(=O)OR 6 , or -C(=O)NR 2 R 6 , wherein each of said alkyl, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); [0061] R 5 and R 7 are each independently a bond, H, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), or -C(=O)R 6 , wherein each of said alkyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); [0062] R 6 is independently selected from CONR 5 R 7 , OR 5 , NR 5 R 7 , SR 5 , SO 2 R 5 , SO 2 NR 5 R 7 , CR 5 R 7 , CR 5 NR 5 R 7 , aryl, heteroaryl, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), P(O)(OR 5 )R 7 , P(O)R 5 R 7 , OP(O)(OR 5 )R 7 , OP(O)R 5 R 7 , Cl, F, Br, I, CF 3 , CHF 2 , CH 2 F, CN, NR 5 SO 2 NR 5 R 7 , NR 5 CONR 5 R 7 , CONR 5 COR 7 , NR 5 C(=N-CN)NR 5 R 7 , C(=N-CN)NR 5 R 7 , NR 5 C(-N=CN)R 7 , NR 5 C(=C- NO 2 )NR 5 R 7 , SO 2 NR 5 COR 7 , NO 2 , CO 2 R 5 , C(C=N-OR 5 )R 7 , CR 5 , CR 5 R 7 , CCR 5 , S(C=O)(C=N- R 5 )R 7 , SF 5 , R 5 NR 5 R 7 , (R 5 O) n R 7 , or OCF 3 ; wherein n is an integer from 1 to 10; [0063] In preferred embodiments, the Linker moiety “L” is a linker which consists of a chemical structural unit represented by the formula -A q- , in which q is an integer greater than 1, and each A is independently selected from the group consisting of CH 2 , O, NH, NCH 3 , CONH, CO, C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl. Preferably the Linker does not comprise any alkyl chain having more than 3 carbons (e.g., L may comprise subunits selected from -CH 2 -, -CH 2 CH 2 -, and -CH 2 CH 2 CH 2 -,), and preferably the Linker comprises one or more ethyleneoxy subunits (e.g., -CH 2 CH 2 O-). [0064] In certain embodiments, the compound is selected from the group consisting of the exemplary compounds as described below, and salts and polymorphs thereof:

[0065] In one aspect, the disclosure provides compounds of formula (I): . [0066] Such a compound is referred to as Androgen Receptor Binder-Linker-E3 Ligase Binder (I). It is understood that the terms “Androgen Receptor Binder,” “Androgen Receptor Binding Moiety” and “AR Binding Moiety” refers to a molecular structure which generally binds successfully to androgen receptor protein, recognizing that in different people androgen receptors will not have the identical amino acid sequence, and thus, the strength of binding may vary across different particular AR sequences. [0067] In further embodiments of this aspect, the present disclosure provides: 1.1 A compound (Compound 1) having a chemical structure ARB-E3LB or ARB-L-E3LB, wherein ARB is an AR binding moiety that does not bind to a ligand binding domain, E3LB is an E3 ligase binding moiety, and L is a linker coupling the AR binding moiety to the E3 ligase binding moiety, and wherein the E3LB moiety has the following general structure: , wherein: “ ”represents a bond that may be stereospecific ((R) or (S)), or non- stereospecific; C is an aryl or heteroaryl ring, each optionally substituted by one or more substituents R 9 ; Q is -CH- or -N-; each R 2 and R 3 is independently selected from H, halo, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy R 4 is selected from H, OH, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), C 1-6 alkyl-OCOOR 6 , C 1-6 alkyl-OCONR 5 R 7 , CH 2 -heterocycloalkyl optionally substituted with R 5 , or benzyl optionally substituted with R 5 ; R 5 and R 7 are each independently a bond, H, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), or -C(=O)R 6 , wherein each of said alkyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); and R 6 is independently selected from CONR 5 R 7 , OR 5 , NR 5 R 7 , SR 5 , SO 2 R 5 , SO 2 NR 5 R 7 , CR 5 R 7 , CR 5 NR 5 R 7 , aryl, heteroaryl, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), P(O)(OR 5 )R 7 , P(O)R 5 R 7 , OP(O)(OR 5 )R 7 , OP(O)R 5 R 7 , Cl, F, Br, I, CF 3 , CHF 2 , CH 2 F, CN, NR 5 SO 2 NR 5 R 7 , NR 5 CONR 5 R 7 , CONR 5 COR 7 , NR 5 C(=N-CN)NR 5 R 7 , C(=N- CN)NR 5 R 7 , NR 5 C(-N=CN)R 7 , NR 5 C(=C-NO 2 )NR 5 R 7 , SO 2 NR 5 COR 7 , NO 2 , CO 2 R 5 , C(C=N-OR 5 )R 7 , CR 5 , CR 5 R 7 , CCR 5 , S(C=O)(C=N-R 5 )R 7 , SF 5 , R 5 NR 5 R 7 , (R 5 O)nR 7 , or OCF 3 ; wherein n is an integer from 1 to 10; and R 9 is independently selected from H, halo (e.g., fluoro, chloro), hydroxy, - CONH 2 , -CONR 2 R 3 , -SONH 2 , -SONR 2 R 3 , -SO 2 NH 2 , -SO 2 NR 2 R 3 , -NHCOC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 COC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SO 2 C 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SOC 1-3 alkyl (optionally substituted by 1 or more halo), cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 (e.g., N,N-dimethyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R 9 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl). 1.2 Compound 1.1, wherein the AR binding moiety binds to one more of AR splice variants V1 to V15, for example, to AR splice variant V7 (AR-V7). 1.3 Compound 1.1 or 1.2, wherein the E3LB moiety has a general structure selected from the following: wherein R 4 , R 9 , and L are as provided in Compound 1.1, and wherein each X is independently selected from C(R 9 ) and -N-, each Y is independently selected from C(R 9 ), -NH-, -N(R 10 )-, -O-, and -S-, and Z is -C- or -N-; wherein R 10 is selected from C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), -C(=O)R 6 , - C(=O)OR 6 , or -C(=O)NR 2 R 6 , wherein each of said alkyl, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl). 1.4 Any of Compounds 1.1-1.3, wherein the E3LB moiety has the general structure: wherein R 4 , R 9 , and L are as provided in Compound 1.1. 1.5 Any of Compounds 1.1-1.3, wherein the E3LB moiety has the general structure: wherein R 4 , R 9 , and L are as provided in Compound 1.1, and wherein each X is independently selected from C(R 9 ) and -N-. 1.6 Any of Compounds 1.1-1.3, wherein the E3LB moiety has the general structure:

wherein R 4 , R 9 , and L are as provided in Compound 1.1, and wherein each Y is independently selected from C(R 9 ), -NH-, -N(R 10 )-, -O-, and -S-, and Z is -C- or -N-, and R 10 is as provided hereinabove. 1.7 Compound 1, or any of 1.1-1.6, wherein ring C is a 6-membered aryl (e.g., phenyl), optionally substituted by one or more group R 9 . 1.8 Compound 1, or any of 1.1-1.6, wherein ring C is a 6-membered heteroaryl (e.g., pyridine, pyrimidine, pyridazine, or pyrazine), optionally substituted by one or more groups R 9 . 1.9 Compound 1, or any of 1.1-1.6, wherein ring C is a 5-membered heteroaryl, optionally substituted by one or more groups R 9 . 1.10 Compound 1.9, wherein ring C is selected from furan, thiophene, pyrrole, pyrazole, thiazole, oxazole, imidazole, isothiazole, isoxazole, triazole, thiadiazole, oxadiazole, or tetrazole, each optionally substituted by one or more groups R 9 . 1.11 Compound 1, or any of 1.1-1.10, wherein the E3LB moiety is a moiety selected from any of the following:

wherein X is -NH-, N(R 10 ), -O-, or -S-. 1.12 Compound 1, or any of 1.1-1.10, wherein the E3LB moiety is a moiety is selected from any of the following:

1.13 Compound 1, or any of 1.1-1.12, wherein ring C is substituted by 0 groups R 9 , 1 group R 9 , or two groups R 9 , or three groups R 9 , or four groups R 9 , as permitted by the structure of ring C. 1.14 Compound 1, or any of 1.1-1.10, wherein the E3LB binding moiety is: 1.15 Compound 1.14, wherein Ring C is substituted by 0, 1 or 2 groups R 9 . 1.16 Compound 1.14, wherein Ring C is substituted by 1 or 2 groups R 9 . 1.17 Compound 1.14, wherein Ring C is substituted by 1 group R 9 . 1.18 Compound 1.14, wherein Ring C is substituted by 0 groups R 9 . 1.19 Any preceding compound, wherein each R 9 is independently selected from H, halo (e.g., fluoro, chloro or bromo), hydroxy, CN, C 1-6 alkyl (e.g., methyl), NH 2 , haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl), and wherein R 4 is H or C 1-3 alkyl (e.g., methyl), and optionally wherein R 4 and each R 9 are H. 1.20 Any preceding compound, wherein the E3LB binding moiety is: 1.21 Any preceding compound, wherein the AR binding moiety is selected from: ,

, wherein: A is an aryl (e.g., phenyl), heteroaryl (e.g., pyridyl), C 3-7 cycloalkyl, or 3-10 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as morpholine), or a bridged bicyclic 7-12 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as a bridged morpholine), each of which is optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 (e.g., N,N-dimethyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); B is an aryl (e.g., phenyl) or heteroaryl (e.g., pyridyl or imidazolyl) ring, optionally substituted by one or more groups selected from halo (e.g., fluoro), hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 (e.g., N,N-dimethyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), aryl, heteroaryl, and 3-6 membered heterocycloalkyl (e.g., with 1-4 heteroatoms), wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); and each R 1 is independently selected from H, halo (e.g., chloro or fluoro), hydroxy, - CONH 2 , -CONR 2 R 3 , -SONH 2 , -SONR 2 R 3 , -SO 2 NH 2 , -SO 2 NR 2 R 3 , -NHCOC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 COC 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SO 2 C 1-3 alkyl (optionally substituted by 1 or more halo), -NR 2 SOC 1-3 alkyl (optionally substituted by 1 or more halo), cyano, C 1-6 alkyl (e.g., methyl), C 2-6 alkenyl (e.g., vinyl), C 2-6 alkynyl (e.g., ethynyl), NR 2 R 3 (e.g., N,N-dimethyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), C 3-6 cycloalkyl (e.g., cyclopropyl), or, if applicable, taken together with an R 1 on an adjacent atom, together with the atoms they are attached to, form a 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkenyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, aryl, or heteroaryl ring system, wherein each of said alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heteroaryl and heterocycloalkyl are independently optionally substituted by one or more halo, hydroxy, nitro, cyano, C 1-6 alkyl (e.g., methyl), haloC 1-6 alkyl (e.g., CH 2 F, CHF 2 , CF 3 ), C 1-6 alkoxy (e.g., methoxy), or C 3-6 cycloalkyl (e.g., cyclopropyl); and each R 2 and R 3 is independently selected from H, halo, C 1-6 alkyl (e.g., methyl), C 3-6 cycloalkyl (e.g., cyclopropyl), or taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms, wherein said alkyl or cycloalkyl is optionally substituted with one or more halo or hydroxy. 1.22 Compound 1.21, wherein A is: wherein X is CH or N, or wherein A is , wherein each of Q 1 , Q 2 , and Q 3 are independently selected from O, S, NR 2 , C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 ) optionally substituted by one or more R 1 , or wherein Q 1 or Q 2 is absent. 1.23 Compound 1.21 or 1.22, wherein B is: 1.24 Any preceding compound, wherein the AR binding moiety is: A is a 3-10 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as morpholine) or a bridged bicyclic 7-12 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as a bridged morpholine), B is aryl (e.g., phenyl) or heteroaryl (e.g., imidazolyl) optionally substituted by one or more halo, and R 1 is H, hydroxy, cyano, NH 2 , methoxy, halo (e.g., fluoro), or C 1-6 alkyl (e.g., methyl); or the AR binding moiety i A is a 3-10 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as morpholine) or a bridged bicyclic 7-12 membered heterocycloalkyl ring (e.g., with 1-4 heteroatoms, such as a bridged morpholine), and R 1 is H, hydroxy, cyano, NH 2 , methoxy, halo (e.g., fluoro), or C 1-6 alkyl (e.g., methyl). 1.25 Compound 1.21, wherein A is selected from morpholinyl, piperazinyl, N- methylpiperazinyl, piperidinyl, and pyrrolidinyl. 1.26 Compound 1.25, wherein A is morpholinyl (e.g., 1-morpholinyl). 1.27 Any of Compounds 1.21 to 1.24, wherein A is , wherein each of Q 1 , Q 2 , and Q 3 are independently selected from O, S, NR 2 , C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ), optionally substituted by one or more R 1 , or wherein Q 1 or Q 2 is absent. 1.28 Compound 1.27, wherein Q 2 is absent, and Q 1 and Q 3 are independently selected from O, S, NR 2 , C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ), optionally substituted by one or more R 1 . 1.29 Compound 1.28, wherein Q 1 is C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ), optionally substituted by one or more R 1 , and Q 3 is selected from O, S, NR 2 , and C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ) optionally substituted by one or more R 1 . 1.30 Compound 1.28, wherein Q 1 is C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ), optionally substituted by one or more R 1 and Q 3 is selected from O, S, and NR 2 . 1.31 Compound 1.28, wherein Q 1 is CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 , and Q 3 is selected from O, S, and NR 2 . 1.32 Compound 1.28, wherein Q 1 is CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 , and Q 3 is selected from O, S, and NH. 1.33 Compound 1.28, wherein Q 1 is CH 2 and Q 3 is O. 1.34 Compound 1.28, wherein Q 1 is CH 2 CH 2 , and Q 3 is O. 1.35 Compound 1.27, wherein Q 1 is absent, and Q 2 and Q 3 are independently selected from O, S, NR 2 , C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ) optionally substituted by one or more R 1 . 1.36 Compound 1.35, wherein Q 2 is C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ) optionally substituted by one or more R 1 , and Q 3 is selected from O, S, NR 2 , and C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ) optionally substituted by one or more R 1 . 1.37 Compound 1.35, wherein Q 2 is C 1-3 alkyl (e.g., CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 ), optionally substituted by one or more R 1 and Q 3 is selected from O, S, and NR 2 . 1.38 Compound 1.35, wherein Q 2 is CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 , and Q 3 is selected from O, S, and NR 2 . 1.39 Compound 1.35, wherein Q 2 is CH 2 or CH 2 CH 2 or CH 2 CH 2 CH 2 , and Q 3 is selected from O, S, and NH. 1.40 Compound 1.35, wherein Q 2 is CH 2 and Q 3 is O. 1.41 Compound 1.35, wherein Q 2 is CH 2 CH 2 , and Q 3 is O 1.42 Any of Compounds 1.21 to 1.41, wherein each R 1 is H or halo (e.g., F, Cl, or Br). 1.43 Any of Compounds 1.21 to 1.41, wherein each R 1 is H. 1.44 Any of Compounds 1.21 to 1.43, wherein B is phenyl optionally substituted by one or two halo (e.g., fluoro, chloro or bromo) or B is imidazolyl optionally substituted by one or two halo (e.g., fluoro, chloro or bromo). 1.45 Compound 1.44, wherein B is: wherein each R 1 is independently H, F, or Cl. 1.46 Any preceding compound, wherein the AR binding moiety is selected from: 1.47 Any preceding compound, wherein the AR binding moiety is: 1.48 Any preceding com R binding moiety is selected from:

1.49 Any preceding compound, wherein the ARB moiety is: 1.50 Any preceding compound, wherein the ARB moiety is: 1.51 Any preceding compound, wherein the linker (“L”) consists of a chemical structural unit represented by the formula -A q- , in which q is an integer greater than 1, and each A is independently selected from the group consisting of: a bond, CR L1 R L2 , O, S, SO, SO 2 , NR L3 , SO 2 NR L3 , SONR L3 , CONR L3 , NR L3 CONR L4 , NR L3 SO 2 NR L4 , CO, CR L1 =CR L2 , , SiR L1 R L2 , P(O)R L1 , P(O)OR L1 , NR L3 C(=NCN)NR L4 , NR L3 C(=NCN), NR L3 C(=CNO 2 )NR L4 , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups; and wherein: R L1 , R L2 , R L3 , R L4 and R L5 are each independently selected from the group consisting of H, halo, C 1-8 alkyl, OC 1-8 alkyl, SC 1-8 alkyl, NHC 1-8 alkyl, N(C 1-8 alkyl) 2 , C 3-11 cycloalkyl, aryl, heteroaryl, 3-6 membered heterocycloalkyl, OC 1-8 cycloalkyl, SC 1-8 cycloalkyl, NHC 1-8 cycloalkyl, N(C 1-8 cycloalkyl) 2 , N(C 1-8 cycloalkyl)(C 1-8 alkyl), OH, NH 2 , SH, SO 2 C 1-8 alkyl, P(O)(OC 1-8 alkyl)(C 1-8 alkyl), P(O)(OC 1-8 alkyl) 2 , CC-C 1-8 alkyl, CCH, CH=CH(C 1-8 alkyl), C(C 1-8 alkyl)=CH(C 1-8 alkyl), C(C 1-8 alkyl)=C(C 1-8 alkyl) 2 , Si(OH) 3 , SiC(1-8 alkyl) 3 , Si(OH)(C 1-8 alkyl) 2 , COC 1-8 alkyl, CO 2 H, CN, haloC 1-8 alkyl (e.g., CF 3 , CHF 2 , CH 2 F), NO 2 , SF 5 , SO 2 NHC 1-8 alkyl, SO 2 NHC 1-8 alkyl, SO 2 N(C 1-8 alkyl) 2 , SONHC 1-8 alkyl, SON(C 1-8 alkyl) 2 , CONHC 1-8 alkyl, CON(C 1-8 alkyl) 2 , N(C 1-8 alkyl)CONH(C 1-8 alkyl), N(C 1-8 alkyl)CON(C 1-8 alkyl) 2 , NHCONH(C 1 - 8 alkyl), NHCON(C 1-8 alkyl) 2 , NHCONH 2 , N(C 1-8 alkyl)SO 2 NH(C 1-8 alkyl), N(C 1-8 alkyl)SO 2 N(C 1-8 alkyl) 2 , NHSO 2 NH(C 1-8 alkyl), NHSO 2 N(C 1-8 alkyl) 2 and NHSO 2 NH 2 ; and wherein R L1 and R L2 each, independently may be linked to another A group to form a cycloalkyl and or heterocycloalkyl moiety that can be further substituted with 0-4 R L5 groups. 1.52 Compound 1.51, wherein q is an integer from 1 to 30, e.g., 5 to 25, or 5 to 20, or 5 to 15, or 10 to 20, or 5 to 10, or 10 to 15, or 7 to 12. 1.53 Compound 1.51, wherein q is an integer from 5 to 15. 1.54 Compound 1.51, wherein q is an integer from 10 to 20. 1.55 Compound 1.51, wherein q is an integer from 5 to 10. 1.56 Any of compounds 1.51 to 1.55, wherein R L1 , R L2 , R L3 , R L4 and R L5 are each independently selected from the group consisting of H, halo, C 1-8 alkyl, OC 1-8 alkyl, NHC 1-8 alkyl, N(C 1-8 alkyl) 2 , C 3-11 cycloalkyl, aryl, heteroaryl, 3-6 membered heterocycloalkyl, OC 1-8 cycloalkyl, NHC 1-8 cycloalkyl, N(C 1-8 cycloalkyl) 2 , N(C 1-8 cycloalkyl)(C 1-8 alkyl), OH, NH 2 , haloC 1-8 alkyl (e.g., CF 3 , CHF 2 , CH 2 F), and CONHC 1-8 alkyl. 1.57 Any of compounds 1.51 to 1.55, wherein R L1 , R L2 , R L3 , R L4 and R L5 are each independently selected from the group consisting of H, halo, C 1-8 alkyl, OC 1-8 alkyl, NHC 1-8 alkyl, N(C 1-8 alkyl) 2 , C 3-11 cycloalkyl, aryl, heteroaryl, 3-6 membered heterocycloalkyl, OC 1-8 cycloalkyl, and haloC 1-8 alkyl (e.g., CF 3 , CHF 2 , CH 2 F). 1.58 Any of compounds 1.51 to 1.55, wherein R L1 , R L2 , R L3 , R L4 and R L5 are each independently selected from the group consisting of H, halo, C 1-8 alkyl (e.g., methyl), OC 1-8 alkyl (e.g., methoxy), and C 3-11 cycloalkyl (e.g., cyclopropyl). 1.59 Any of compounds 1.51 to 1.58, wherein the units A are selected from CR L1 R L2 , O, S, SO, SO 2 , NR L3 , SO 2 NR L3 , SONR L3 , CONR L3 , NR L3 CONR L4 , NR L3 SO 2 NR L4 , CO, CR L1 =CR L2 , , C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups. 1.60 Compound 1.59, wherein the linker L is a flexible linker, e.g., wherein the units A are selected from CR L1 R L2 , O, S, SO, SO 2 , NR L3 , SO 2 NR L3 , SONR L3 , CONR L3 , NR L3 CONR L4 , NR L3 SO 2 NR L4 , and CO. 1.61 Compound 1.60, wherein the units A are selected from CR L1 R L2 , O, NR L3 , SO 2 NR L3 , SONR L3 , CONR L3 , NR L3 CONR L4 , and CO. 1.62 Compound 1.60, wherein the units A are selected from CH 2 , O, NH, CONH, and CO. 1.63 Any preceding compound, wherein the linker L is selected from: , wherein n is from 1-5; , wherein n is from 1-5; , wherein n is from 1-5; , wherein m is from 0-12 (e.g., 8); , wherein m is from 0-12; , wherein m is from 0-12; , wherein m is from 2-4; , wherein m is from 0-12; wherein m is from 0-10; 46 herein n is from 1-5 (e.g., 3 or 4); , wherein n is from 1-5 (e.g., 3 or 4); , ; , wherein m is from 0-10; ,

, ; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 0-6 (e.g., 3 or 4); , herein n is from 0-6 (e.g., 3 or 4); , wherein m is from 0-12 (e.g., 2-6); herein n is from 1-5 (e.g., 1 or 2); erein m is from 0-10; m is from 1-12 (e.g., 6-10); in m is from 2 to 8 (e.g., 4, 6, or 8); , wherein m is from 2 to 8 (e.g., 4, 6, or 8); , wherein n is from 1 to 4 (e.g., 2, 3, or 4); , .., .., 3);

(e.g., 0); m is from 0-4 (e.g., 0);

, , m is from 0-4 (e.g., 0); , , m 0-4 (e.g., 0); ., 0);

0); , .., , , 0-4 (e.g., 2); , wherein m1 is from 0-12 (e.g., 4, 5, or 6) and m2 is from 0-4 (e.g., 2); (e.g., 0);

, ;

, .g., , ., 4, 5, or 6) and n is from 1-4 (e.g., 2);

from 1-4 (e.g., 2). 1.64 Any preceding compound, wherein the linker L is selected from:

, wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 and m is from 1-6; , wherein n is from 1-5 (e.g., 1 or 2); , .g., , (e.g., 4, 6, or 8); , wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-4 (e.g., 2 or m is from 4-6; , rein m is from 2-4;

(e.g., 0); m is from 0-4 (e.g., 0);

, , m is from 0-4 (e.g., 0); , , m 0-4 (e.g., 0); ., 0);

(e.g., 0); , wherein m1 is from 0-12 (e.g., 4, 5, or 6) and m2 is from 0-4 (e.g., 2); (e.g., 0); , wherein m1 is from 0-6 (e.g., 0 or 1) and m2 is from 0-4 (e.g., 0);

, . ., , wherein m1 is from 0-12 (e.g., 4, 5, or 6) and m2 is from 0-4 (e.g., 2); and from 1-4 (e.g., 2). 1.65 Any preceding compound, wherein the linker L is selected from:

(e.g., 0); , wherein n is from 0-4 (e.g., 1) and m is from 0-4 (e.g., 0); (e.g., 0);

, .g., s from 0-4 (e.g., 0); , , , 0);

(e.g., 0); , wherein m1 is from 0-12 (e.g., 4, 5, or 6) and m2 is from 0-4 (e.g., 2); (e.g., 0);

, , , 5, or 6) and n is from 1-4 (e.g., 2). 1.66 Any preceding compound, wherein the linker L is selected from:

, .g., , .., in m is from 2 to 8 (e.g., 4, 6, or 8); , wherein m is from 2 to 8 (e.g., 4, 6, or 8); , wherein n is from 1 to 4 (e.g., 2, 3, or 4); and

, , , , from 1-4 (e.g., 2). 1.67 Any preceding compound, wherein the linker L is selected from:

, ,

the above structures “N” is understood to be -NH- or -N(C 1-3 alkyl). 1.68 Any preceding compound, wherein the linker L is selected from: , .g., 2, 3, or 4); , wherein m is from 0-10; , wherein m is from 1-12 (e.g., 6-10);

, .., , wherein m is from 1-12. 1.69 Any preceding compound, wherein the linker L is a structure selected from the group consisting of: , .g., 2, 3, or 4); , . ., . 1.70 Any preceding compound, wherein the linker L is a structure selected from the group consisting of:

, w e e s o - e.g., o . 1.71 Any of Compounds 1.51-1.59, wherein the linker L is a rigid or semi-rigid linker, e.g., wherein the units A comprising the linker L comprise at least one unit A selected from CR L1 =CR L2 , , C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups. 1.72 Compound 1.71, wherein L comprises a single unit A selected from CR L1 =CR L2 , , C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups; and all other units A are as otherwise provided. 1.73 Compound 1.71 or 1.72, wherein the units A are selected from CR L1 R L2 , O, S, SO, SO 2 , NR L3 , SO 2 NR L3 , SONR L3 , CONR L3 , NR L3 CONR L4 , NR L3 SO 2 NR L4 , CO, CR L1 =CR L2 , , C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups. 1.74 Compound 1.71 or 1.72, wherein the units A are selected from CR L1 R L2 , O, NR L3 , SO 2 NR L3 , CONR L3 , NR L3 CONR L4 , CO, CR L1 =CR L2 , , C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups. 1.75 Compound 1.71 or 1.72, wherein the units A are selected from CR L1 R L2 , O, NR L3 , CONR L3 , CO, C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups, and wherein R L1 , R L2 , and R L3 are each independently selected from the group consisting of H, halo, and C 1-8 alkyl (e.g., methyl), and wherein L comprises at least one unit A selected from C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl). 1.76 Compound 1.75, wherein L consists of a set of moieties A selected from CH 2 , O, NH, NCH3, CONH, CO, cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, phenyl, pyridinyl, pyrimidinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiophenyl, furanyl, and pyrrolyl, optionally, with at least one of cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, phenyl, pyridinyl, pyrimidinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiophenyl, furanyl, and pyrrolyl. 1.77 Compound 1.70, wherein the linker L has a structure selected from the group consisting of: ,

wherein each F is a group selected from C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3- 6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, pyridazinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups, wherein R L1 and R L2 are each independently selected from the group consisting of H, halo, and C 1-8 alkyl (e.g., CH 3 ); and wherein a, b, and c are each independently integers from 0 to 10, e.g., 0 to 8, or 0 to 6, or any of 0, 1, 2, 3, 4, 5, 6, or 7. 1.78 Compound 1.77, wherein each F is independently selected from cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, pyridazinyl, and triazolyl, and wherein a, b and c are each independently integers selected from 0, 1, 2, 3 and 4. 1.79 Compound 1.77, wherein the linker L is a structure selected from the group consisting of:

1.80 Any preceding compound, wherein the compound comprises:

H and each R 9 is H, and a linker selected from: , , , , 3, or 4); m is from 1-12 (e.g., 6-10); herein n is from 1-5 and m is from 1-6; herein n is from 0-6 (e.g., 3 or 4); , wherein n is from 0-4 (e.g., 0 or 1) and m is from 0-12 (e.g., 7-9); , wherein m 1 is from 0-12 (e.g., 4, 5, or 6) and m 2 is from 0-4 (e.g., 2); , 12 (e.g., 4, 5, or 6) and m 2 is from 0-4 (e.g., 2). 1.81 Compound 1.80, wherein the linker L is selected from: , 1 12 (e.g., 4, 5, or 6) and m 2 is from 0-4 (e.g., 2). 1.82 Any preceding compound, wherein L consists of a set of moieties A selected from CH 2 , O, NH, NCH3, CONH, CO, cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, phenyl, pyridinyl, pyrimidinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiophenyl, furanyl, and pyrrolyl, optionally wherein at least one moiety A is selected from cyclopropyl, cyclobutyl, morpholinyl, piperidinyl, piperazinyl, azetidinyl, phenyl, pyridinyl, pyrimidinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiophenyl, furanyl, and pyrrolyl. 1.83 Compound 1.82, wherein L consists of a set of moieties A selected from CH 2 , O, NH, CONH, and CO. 1.84 Any preceding compound, wherein L does not comprise any alkyl chain having more than 3 carbons (e.g., L may comprise subunits selected from -CH 2 -, -CH 2 CH 2 -, and - CH 2 CH 2 CH 2 -), optionally wherein L comprises one or more ethyleneoxy subunits (e.g., - CH 2 CH 2 O-). 1.85 Any preceding compound, wherein the compound comprises: the AR binding moiety

H and each R 9 is H, and a linker selected from: wherein each F is selected from C 3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl), 3-6 membered heterocycloalkyl (e.g., piperidinyl, piperazinyl, azetidinyl), aryl (e.g., phenyl), and heteroaryl (e.g., pyridinyl, pyrimidinyl, pyridazinyl, triazolyl, oxadiazolyl, imidazolyl, pyrazolyl, isoxazolyl, pyrrolyl), wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with 0-6 R L1 and/or 0-6 R L2 groups, wherein R L1 , R L2 , and R L2 are each independently selected from the group consisting of H, halo, and C 1-8 alkyl (e.g., CH3); and wherein a, b and c are each independently integers from 0 to 10, e.g., 0 to 8, 0 to 6, or any of 0, 1, 2, 3, 4, 5, 6, or 7. 1.86 Compound 1.85, wherein the linker is selected from: wherein: each F is selected from 3-6 membered heterocycloalkyl (e.g., morpholinyl, piperidinyl, piperazinyl, azetidinyl), and heteroaryl (e.g., pyridinyl, pyrimidinyl, pyridazinyl), and wherein a is an integer from 0-4 (e.g., 0, 1 or 4) and b is an integer from 0-4 (e.g., 0 or 1). 1.87 Any preceding compound, wherein the compound comprises:

H and each R 9 is H, and the linker is: 1.88 Any preceding compound, wherein the compound comprises:

H and each R 9 is H, and the linker is: , wherein m is from 2-10 (e.g., 8 or 9). 1.89 Any preceding compound, wherein the compound is selected from any one or more of Examples 1 to 39 in the table above. 1.90 Any of Compounds 1.1-1.89, wherein the compound is effective in causing or promoting the degradation of the androgen receptor (AR) in a cell, or of causing or promoting apoptosis in a cell. 1.91 Compound 1.90, wherein the cell is a cancer cell (e.g., a prostate cancer cell or ovarian cancer cell, for example, castration-resistant prostate cancer (CRPC) cell). 1.92 Compound 1.90 or 1.91, wherein the cell overexpresses the AR or expresses a mutated AR, such as an AR having a truncated ligand binding domain or absent ligand binding domain. 1.93 Compound 1.92, wherein the mutant AR is any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant. 1.94 A pharmaceutical composition comprising any of Compounds 1.1-1.93 (e.g., an effective amount of any of Compounds 1.1-1.93), and a pharmaceutically acceptable carrier, additive and/or excipient. 1.95 Pharmaceutical Composition 1.94, further comprising at least one additional anticancer agent. 1.96 Any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, for use in the treatment of a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease or condition. 1.97 Use of any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, in the treatment of a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease or condition. 1.98 A Method of treating a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease or condition, said method comprising administering an effective amount of any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, to a patient in need thereof. 1.99 Any of the Uses or Methods according to 1.96 to 1.98, wherein the disease or condition is a cancer. 1.100 Any of the Uses or Methods according to 1.96 to 1.99, wherein the disease or condition is a cancer identified as having a mutation resulting, or expected to result in, overexpression of the androgen receptor. 1.101 Use or Method 1.100, wherein the cell expresses a mutated androgen receptor, e.g., one in which there is a mutation in the ligand binding domain of the AR. 1.102 Use or Method 1.101, wherein the ligand binding domain of the AR is truncated or absent. 1.103 Any of Uses or Methods 1.98-1.102, wherein the cell expresses or overexpresses any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant. 1.104 Any of the uses or methods according to 1.100 to 1.103, wherein the cancer is a prostate cancer or ovarian cancer. 1.105 Use or Method 1.104, wherein the cancer is a prostate cancer, for example, castration- resistant prostate cancer (CRPC). 1.106 Any of the uses or methods according to 1.96 to 1.105, wherein the disease or condition is not responsive to, or no longer responsive to, treatment with an androgen receptor antagonist (e.g., abiraterone, apalutamide, enzalutamide, or darolutamide). 1.107 Any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, for use in the degradation of an androgen receptor in a cell, e.g., a mutated AR such as any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant. 1.108 Use of any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, in the degradation of an androgen receptor (AR) in a cell, e.g., a mutated AR such as any AR- V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant. 1.109 A Method of degrading an androgen receptor in a cell, e.g., a mutated AR such as any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant, said method comprising administering an effective amount of any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, to such cell. 1.110 Any of Uses or Methods 1.107-1.109, wherein the cell is a cancer cell (e.g., a prostate cancer cell or ovarian cancer cell, for example, castration-resistant prostate cancer (CRPC) cell). 1.111 Any of Uses or Methods 1.107-1.110, wherein the cell overexpresses the AR or expresses a mutated AR, such as an AR having a truncated ligand binding domain or absent ligand binding domain. 1.112 Use or Method 1.111, wherein the mutant AR is any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant. 1.113 Any of Uses or Methods 1.107-1.112, wherein the AR is resistant to inhibition by an AR antagonist (e.g., abiraterone, apalutamide, enzalutamide, or darolutamide). 1.114 Any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, for use in inducing apoptosis in a cell, e.g., a cancer cell. 1.115 Use of any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, in the induction of apoptosis in a cell, e.g., a cancer cell. 1.116 A Method of inducing apoptosis in a cell, e.g., a cancer cell, said method comprising administering an effective amount of any of Compounds 1.1-1.93, or pharmaceutical composition 1.94 or 1.95, to such cell. 1.117 Any of Uses or Methods 1.114-1.116 wherein the cell is a prostate cancer cell or ovarian cancer cell (for example, castration-resistant prostate cancer (CRPC) cell). 1.118 Any of Uses or Methods 1.114-1.117, wherein the cell overexpresses the androgen receptor (AR) or expresses a mutated AR, such as an AR having a truncated ligand binding domain or absent ligand binding domain. 1.119 Use or Method 1.118, wherein the mutant AR is any AR-V1 to AR-V15 splice variant, e.g., the AR-V7 splice variant. 1.120 Any of Uses or Methods 1.107-1.119, wherein the cell is from a patient suffering from or diagnosed with cancer. 1.121 Any of Uses or Methods 1.107-1.119, wherein the cell is in a patient suffering from or diagnosed with cancer. DEFINITIONS [0068] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The terminology used in the description is for describing particular embodiments only and is not intended to be limiting of the invention. [0069] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise (such as in the case of a group containing a number of carbon atoms in which case each carbon atom number falling within the range is provided), between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention. [0070] The following terms are used to describe the present disclosure. In instances where a term is not specifically defined herein, that term is given an art-recognized meaning by those of ordinary skill applying that term in context to its use in describing the present invention. [0071] The articles “a” and “an” as used herein and in the claims are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article unless the context clearly indicates otherwise. By way of example, “an element” means one element or more than one element. [0072] The phrase “and/or” as used herein and in the claims should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc. [0073] As used herein in the specification and in the claims, the term “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of” or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” [0074] The term “about” and the like, as used herein, in association with numeric values or ranges, reflects the fact that there is a certain level of variation that is recognized and tolerated in the art due to practical and/or theoretical limitations. For example, minor variation is tolerated due to inherent variances in the manner in which certain devices operate and/or measurements are taken. In accordance with the above, the term “about” is normally used to encompass values within the standard deviation or standard error. For example, “about” may be used to indicate a value above or below the stated value by up to 10% (i.e., ± 10%). [0075] In the claims, as well as in the specification, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean “including without limitation”. Only the transitional phrases “consisting of” and “consisting essentially of” shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03. [0076] It should also be understood, that although various compounds, compositions, and methods are described in “open” terms of “comprising,” “including,” or “having” various components or steps (interpreted as meaning “including without limitation”), the compounds, compositions, methods, and devices can also “consist essentially of” or “consist of” the various components and steps, and such terminology should be interpreted as defining essentially closed- member groups. This paragraph is not meant in any way to limit the meaning of “comprising,” “having,” or “including” (and other verb forms thereof) which are to be interpreted as open- ended phrases meaning “including without limitation” consistent with patent law and custom. The intent of this paragraph is merely to indicate that the closed-member groups defined by the “consisting of” or “consisting essentially of” language are lesser included groups within the open-ended descriptions and to provide support for claims employing the “consisting of” or “consisting essentially of” language. [0077] As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from anyone or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc. [0078] It should also be understood that, in certain methods described herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited unless the context indicates otherwise. [0079] The terms “co-administration” and “co-administering” or “combination therapy” can refer to both concurrent administration (administration of two or more therapeutic agents at the same time) and time varied administration (administration of one or more therapeutic agents at a time different from that of the administration of an additional therapeutic agent or agents), as long as the therapeutic agents are present in the patient to some extent, preferably at effective amounts, at the same time. In some embodiments, one or more of the present compounds described herein, are co-administered in combination with at least one additional bioactive agent, such as another anticancer agent. In some embodiments, the co-administration of compounds results in synergistic activity and/or therapy, including anticancer activity. [0080] The term “effective” can mean, but is in no way limited to, that amount/dose of the active pharmaceutical ingredient, which, when used in the context of its intended use, effectuates or is sufficient to prevent, inhibit the occurrence, ameliorate, delay or treat (alleviate a symptom to some extent, preferably all) the symptoms of a condition, disorder or disease state in a subject in need of such treatment or receiving such treatment. The term effective subsumes all other effective amount or effective concentration terms, e.g., “effective amount/dose,” “pharmaceutically effective amount/dose” or “therapeutically effective amount/dose,” which are otherwise described or used in the present application. [0081] The effective amount depends on the type and severity of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors which those skilled in the medical arts will recognize. [0082] The term “pharmacological composition,” “therapeutic composition,” “therapeutic formulation” or “pharmaceutically acceptable formulation” can mean, but is in no way limited to, a composition or formulation that allows for the effective distribution of an agent provided by the present disclosure, which is in a form suitable for administration to the physical location most suitable for their desired activity, e.g., systemic administration. [0083] The term “pharmaceutically acceptable” can mean, but is in no way limited to, entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a patient or subject. [0084] The term “pharmaceutically acceptable carrier” can mean, but is in no way limited to, any and all solvents, excipients, coatings, and the like, compatible with pharmaceutical administration to a patient or subject. [0085] The term “systemic administration” refers to a route of administration that is, e.g., enteral or parenteral, and results in the systemic distribution of an agent leading to systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body. Suitable forms, in part, depend upon the use or the route of entry, for example oral (enteral) administration or parenteral (e.g., intravenous injection). Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged polymer is desired to be delivered to). For example, pharmacological compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms which prevent the composition or formulation from exerting its effect. [0086] The terms “patient” and “subject” are used throughout the specification to describe a cell, tissue, or animal, preferably a mammal, e.g., a human or a domesticated animal, to whom treatment, including prophylactic treatment, with the compositions according to the present disclosure is provided. For treatment of those infections, conditions or disease states which are specific for a specific animal such as a human patient, the term patient refers to that specific animal, including a domesticated animal such as a dog or cat or a farm animal such as a horse, cow, sheep, etc. In general, in the present disclosure, the term patient refers to a human patient unless otherwise stated or implied from the context of the use of the term. [0087] The term “compound,” as used herein, unless otherwise indicated, refers to any specific chemical compound or genus of compounds disclosed herein and includes tautomers, regioisomers, geometric isomers, and where applicable, stereoisomers, including optical isomers (enantiomers) and other stereoisomers (diastereomers) thereof, as well as pharmaceutically acceptable salts, polymorphs, and derivatives thereof where applicable, in context. Within its use in context, the term compound generally refers to a single compound, but also may include other compounds such as stereoisomers, regioisomers and/or optical isomers (including racemic mixtures) as well as specific enantiomers or enantiomerically enriched mixtures of disclosed compounds. The term also refers, in context to prodrug forms of compounds which have been modified to facilitate the administration and delivery of compounds to a site of activity. The term also refers to any specific chemical compound in which one or more atoms have been replaced with one or more different isotopes of the same element. It is noted that in describing the present compounds, numerous substituents and variables associated with same, among others, are described. [0088] It is understood by those of ordinary skill that molecules which are described herein are stable compounds as generally described hereunder. When the bond is shown, both a double bond and single bond are represented or understood within the context of the compound shown and well-known rules for valence interactions. [0089] As used herein, “derivatives” can mean compositions formed from the native compounds either directly, by modification, or by partial substitution. As used herein, “analogs” can mean compositions that have a structure similar to, but not identical to, the native compound. [0090] The term “ubiquitin ligase” refers to a family of proteins that facilitate the transfer of ubiquitin to a specific substrate protein, targeting the substrate protein for degradation. For example, cereblon is an E3 ubiquitin ligase protein that alone or in combination with an E2 ubiquitin-conjugating enzyme causes the attachment of ubiquitin to a lysine on a target protein, and subsequently targets the specific protein substrates for degradation by the proteasome. Thus, E3 ubiquitin ligase alone or in complex with an E2 ubiquitin conjugating enzyme is responsible for the transfer of ubiquitin to targeted proteins. In general, the ubiquitin ligase may be involved in polyubiquitination such that a second ubiquitin may be attached to the first; a third may be attached to the second, and so forth. Polyubiquitination marks proteins for degradation by the proteasome. However, there are some ubiquitination events that are limited to mono- ubiquitination, in which only a single ubiquitin is added by the ubiquitin ligase to a substrate molecule. Mono-ubiquitinated proteins may not be targeted to the proteasome for degradation, but may instead be altered in their cellular location or function, for example, via binding other proteins that have domains capable of binding ubiquitin. Further, different lysine residues on ubiquitin can be targeted by an E3 to make chains. The most common lysine is Lys48 on the ubiquitin chain. This is the lysine used to make polyubiquitin, which is recognized by the proteasome. [0091] As used herein, the terms “halo” or “halogen” means fluoro (F), chloro (Cl), bromo (Br) or iodo (I). [0092] As used herein, the term “alkyl” means a linear or branched fully saturated hydrocarbon radical or group, preferably a C 1 -C 10 , more preferably a C 1 -C 6 , alternatively a C 1 -C 3 group, which may be optionally substituted. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, or any other acyclic hydrocarbon group having the general formula -C n H 2n+1 . [0093] As used herein, the term “alkenyl” refers to a linear or branched unsaturated hydrocarbon radical group having at least one carbon-carbon double bond, preferably a C 2 -C 10 , more preferably a C 2 -C 6 , which may be optionally substituted. Preferably, said group is mono- unsaturated (having a single carbon-carbon double bond). Examples of alkenyl groups include, but are not limited to, vinyl and allyl, or any other hydrocarbon acyclic group having the general formula -C n H 2n-1 . [0094] As used herein, the term “alkynyl” refers to a linear or branched hydrocarbon radical group having at least one carbon-carbon triple bond, preferably a C 2 -C 10 , more preferably a C 2 - C 6 , which may be optionally substituted. Preferably, said group is mono-unsaturated (having a single triple bond). Examples of alkynyl groups include, but are not limited to, ethynyl and propargyl, or any other acyclic group having at least one triple bond and the general formula - C n H 2n-3 . As used herein, hydrocarbon radicals having both a double bond and a triple bond are considered alkynyl radicals. [0095] As used herein, the term “alkylene” refers to a -(CH 2 ) n - group (wherein n is an integer generally from 1-10, such as 1-6), which may be optionally substituted. When substituted, the alkylene group preferably is substituted on one or more of the methylene groups with a C 1 - C 6 alkyl group (including a cyclopropyl group or a t-butyl group), more preferably a methyl group, but may also be substituted with one or more halo groups, preferably from 1 to 3 halo groups or one or two hydroxyl groups, O-(C 1 -C 6 alkyl) groups or amino acid sidechains as otherwise disclosed herein. In certain embodiments, an alkylene group may be substituted with a urethane or alkoxy group (or other group) which is further substituted with a polyethylene glycol chain (of from 1 to 10, preferably 1 to 6, often 1 to 4 ethylene glycol units) to which is substituted (preferably, but not exclusively on the distal end of the polyethylene glycol chain) an alkyl chain substituted with a single halogen group, preferably a chlorine group. In still other embodiments, the alkylene (often, a methylene) group, may be substituted with an amino acid sidechain group such as a sidechain group of a natural or unnatural amino acid, for example, alanine, (3-alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, praline, serine, threonine, valine, tryptophan, or tyrosine. [0096] As used herein, a range of carbon atoms which includes C 0 means that carbon may be absent and is replaced with H (or deuterium). Thus, a range of carbon atoms which is C 0 -C 6 includes carbons atoms of 1, 2, 3, 4, 5 and 6 and for C 0 , H (or deuterium) stands in place of carbon. [0097] As used herein, the term “unsubstituted” means that the indicated structure or group is further substituted only with hydrogen atoms. [0098] As used herein, it is understood that each of the independent groups, moieties, or units (these terms are synonymous) “A” which form the linker moiety L are bivalent groups having two “open” valences for attachment to other atoms. It is understood that the unit “CO” refers to a carbonyl group, -C(=O)-, and thus any reference to “CO” as a bivalent radical is understood as indicated the attachment of two atoms to the carbon atom of the “CO” unit. Similarly, when used in combination with other hetero atoms, such as in the term “NR L3 CONR L4 ” it is understood that the carbonyl carbon atom is attached to each of the two nitrogen atoms, each of which has a single attached R-group (R L3 or R L4 ), and wherein the entire bivalent radical “NR L3 CONR L4 ” is attached two additional atoms at each of the nitrogen atoms. Similarly, the term “NR L3 SO 2 NR L4 ” refers to a bivalent radical having attachment point on each nitrogen atom with the two nitrogen atoms bridge by the sulfur atom of a sulfonyl group, -S(O)(O)- or -S(O 2 )-. It is further understood that the terms “CO” and “-C(O)-” are interchangeable. More generally, in complex functional groups such as SiR L1 R L2 , P(O)R L1 , and P(O)OR L1 , it is understood that the connections of these bivalent groups to other atoms is on the central atom, i.e., the silicon atom or the phosphorus atom, and that the R-groups or OR-groups are also attached to the central atom. It is further understood that in the bivalent radicals SO, and SO 2 , the oxygen atom or atoms are bonded to the sulfur atom, and the open valences are both on the sulfur atom in each instance. [0099] As used herein, the term “substituted” or “optionally substituted” means that one or more hydrogen atoms of a group or radical is independently (i.e., where more than a single substitution occurs, each substituent is independent of another substituent) replaced by one or more non-hydrogen substituents, up to the maximum permissible number of substituents for the chemical structure, substructure, group or radical, for example, up to five substituents, preferably up to three substituents, often 1 or 2 substituents on a moiety. Substituents may themselves be further substituted. Optional substituents, unless otherwise indicated, include hydroxy (-OH), thiol (-SH), carboxy (-COOH), cyano (-CN), nitro (-NO 2 ), halogen (preferably, F or Cl), C 1 - C 20 alkyl (e.g., C 1 -C 10 alkyl, C 1 -C 6 alkyl), C 2 - C 20 alkenyl (e.g., C 2 -C 10 alkenyl, C 2 -C 6 alkenyl), C 2 - C 20 alkynyl (e.g., C 2 -C 10 alkynyl, C 2 -C 6 alkynyl), aryl (e.g., phenyl, naphthyl), heteroaryl (e.g., 5- to 10-membered ring heteroaryls, such as azoles, diazoles, triazoles, pyridine, diazines, triazines, and benzo-fused derivatives thereof), 5-10 membered heterocycloalkyl (containing at least one heteroatom N, S, or O), C 1 -C 20 alkoxy (e.g., C 1 -C 20 alkoxy, C 1 -C 6 alkoxy), C 3 -C 20 cycloalkyl (e.g., C 3 -C 10 cycloalkyl, C 3 -C 6 cycloalkyl), C 3 -C 20 cycloalkoxy (e.g., C 3 -C 10 cycloalkoxy, C 3 - C 6 cycloalkoxy), aryloxy (e.g., phenoxy), thioether (thioC 1 -C 6 alkyl or aryl), keto, acyl (preferably, C 1 -C 6 acyl), ester or thioester (preferably, C 1 -C 6 alkyl or aryl; including alkylene ester, such that attachment is on the alkylene group, rather than at the ester function which is preferably substituted with a C 1 -C 6 alkyl or aryl group), amine (including amino (-NH 2 ), mono- C 1 -C 6 alkylamino, di-C 1 -C 6 alkylamino, five- or six-membered N-containing heterocycloalkyl), carbamate or urethane (such as optionally substituted N(C 0 -C 6 alkyl)C(O)(OC 1 -C 6 alkyl) group), hydrazine or hydrazide, amido (N-C(O), preferably substituted with one or two C 1 -C 6 alkyl groups; including a carboxamido which is optionally substituted with one or two C 1 -C 6 alkyl groups), sulfone (SO 2 ), sulfoxide (S(O)), sulfonamide, alkanol (preferably, C 1 -C 6 alkyl or aryl, such as (CH 2 ) n OH), or alkanoic acid (preferably, C 1 -C 6 alkyl or aryl, such as (CH 2 ) n COOH), wherein n is an integer from 1-10, e.g., 1-6. Substituents according to the present invention may also include SiR 1 R 2 R 3 groups wherein each of R 1 and R 2 is as otherwise described herein, and R 3 is H or a C 1 -C 6 alkyl group, preferably R 1 , R 2 , R 3 in this context is a C 1 -C 3 alkyl group (including an isopropyl or t-butyl group). Additional optional substituents include: NHC(O)NH, (CH 2 )nSH, ,(CH 2 ) n O(C 1 -C 6 alkyl), (CH 2 ) n C(O)(C 1 -C 6 alkyl), (CH 2 ) n OC(O)(C 1 -C 6 alkyl), (CH 2 ) n C(O)O(C 1 - C 6 alkyl), (CH 2 )nNHC(O)R 1 , (CH 2 )nC(O)NR 1 R 2 , (OCH 2 )nOH, (CH 2 0)nCOOH, C 1 -C 6 alkyl, (OCH 2 )nO(C 1 -C 6 alkyl), (CH 2 O)nC(O)(C 1 -C 6 alkyl), (OCH 2 )nNHC(O)R 1 , (CH 2 O)nC(O)NR 1 R 2 , S(O) 2 R s , and S(O)R s (R s is C 1 -C 6 alkyl or a (CH 2 ) m NR 1 R 2 group). Each of the above-described groups may be linked directly to the substituted moiety or alternatively, the substituent may be linked to the substituted moiety (preferably in the case of an aryl or heteroaryl moiety) through an optionally substituted -(CH 2 ) m - or, alternatively, an optionally substituted -(OCH 2 ) m -, - (OCH 2 CH 2 ) m - or -(CH 2 CH 2 O) m - group, which may be substituted with any one or more of the above-described substituents, wherein m is an integer from 1 to 20, e.g., 1 to 10 or 1 to 6. Alkylene groups -(CH 2 )m- or -(CH 2 )n- groups or other chains such as ethylene glycol chains, as identified above, may be substituted anywhere on the chain. As substituents can themselves be substituted, these groups also include, for example, such compound groups as arylC 1 -C 6 alkyl (e.g., benzyl), haloC 1 -C 6 alkyl (e.g., trifluoromethyl), hydroxyC 1 -C 6 alkyl (e.g., 2-hydroxy-2- methylbutyl), and C 1 -C 6 alkyl-aryl (e.g., tolyl). [00100] In some embodiments, the substituents on alkylene groups include halogen or C 1 -C 6 (preferably C 1 -C 3 ) alkyl groups, which may be optionally substituted with one or two hydroxyl groups, one or two ether groups (OC 1 -C 6 groups), up to three halo groups (preferably F), or a sidechain of an amino acid as otherwise described herein and optionally substituted amide (preferably carboxamide substituted as described above) or urethane groups (often with one or two C 0 -C 6 alkyl substituents, which group(s) may be further substituted). In certain embodiments, the alkylene group (often a single methylene group) is substituted with one or two optionally substituted C 1 -C 6 alkyl groups, preferably C 1 -C 4 alkyl group, most often methyl or O- methyl groups or a sidechain of an amino acid as otherwise described herein. In the present invention, a moiety in a molecule may be optionally substituted with up to five substituents, preferably up to three substituents. Most often, in the present invention moieties which are substituted are substituted with one or two substituents. [00101] As used herein, the terms “aryl” means any carbocyclic aromatic ring system, i.e., any aromatic ring system comprising only carbon atoms as ring atoms. This includes 6-membered monocyclic aryl ring systems and 9-membered or 10-membered fused bicyclic aryl ring systems, and larger fused ring systems, as long as such ring systems comprise at least one 6-membered aromatic carbocyclic ring (i.e., a benzene ring) within the fused ring system, and as long as no ring-atoms are heteroatoms. Thus, the term aryl includes, but is not limited to, phenyl, naphthyl, phenanthryl, and anthracenyl. [00102] As used herein, “heteroaryl” means any cyclic heteroaromatic ring system, i.e., any aromatic ring system comprising at least one heteroatom (e.g., N, S, or O) ring atom. This includes 5-membered and 6-membered monocyclic heteroaryl ring systems (e.g., azoles, azines) and 9-membered or 10-membered fused bicyclic heteroaryl ring systems, and larger fused ring systems, as long as such ring systems comprise at least one aromatic carbocyclic or aromatic heterocyclic ring within the fused ring system and at least one heteroatom (e.g., N, S or O) ring- atom within the fused ring system (either in an aromatic ring or non-aromatic ring). Heteroaryl therefore includes, but is not limited to, bicyclic fused ring systems selected from aromatic- heteroaromatic, aromatic-heterocyclic, heteroaromatic-carbocyclic, heterocyclic-aromatic, and heteroaromatic-heteroaromatic, as well as larger fused ring systems comprising some combination of benzene, cycloalkane, heterocycloalkane and heteroaromatic rings. Exemplary heteroaryl groups include furyl, thienyl, thiazolyl, pyrazolyl, isothiazolyl, oxazolyl, isoxazolyl, pyrrolyl, triazolyl, tetrazolyl, imidazolyl, 1,3,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-oxadiazolyl, 1,3,5-thiadiazolyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyridonyl, 1,2,4-triazinyl, 1,2,3-triazinyl, 1,3,5-triazinyl, pyrazolo[3,4-b]pyridinyl, cinnolinyl, pteridinyl, purinyl, 6,7-dihydro-5H-[1]pyridinyl, benzo[b]thiophenyl, 5,6,7,8- tetrahydro-quinolin-3-yl, benzoxazolyl, benzofuranyl, isobenzofuranyl, benzothiazolyl, benzisothiazolyl, benzisoxazolyl, benzimidazolyl, benzothiadiazolyl, thianaphthenyl, isothianaphthenyl, benzofuranyl, isobenzofuranyl, isoindolyl, indolyl, indolizinyl, indazolyl, isoquinolinyl, quinolinyl, phthalazinyl, quinoxalinyl, quinazolinyl, naphthyridyl, phenanthridinyl, acridinyl, carbazolyl, carbazolinyl, permidinyl, phenanthrolinyl, phenacenyl, pyrrolopyrimidinyl, pyrrolopyridinyl, pyridopyrimidinyl, theinopyrimidinyl, furopyrimidinyl, furopyridyl, furopyrrolyl, pyrazoloxazolyl, thienofuranyl, imidazothiazolyl, imidazopyridyl, imidazotriazyl, imidazopyrimidinyl, pyrazinopyridazinyl, phenothiazinyl, furazanyl, phenoxazinyl, pyrazo benzoxazinyl, azaindolizinyl, dihydroquinolinyl, dihydroisoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyridoxazinyl. It is understood that for heteroaryl systems in which both the ring carbon atoms and ring heteroatoms have open valencies, bonds can be formed to either such atom types (e.g., C-linked or N-linked). For example, where a pyrazolyl ring is one of the group A in the Linker moiety L, the adjacent A groups can be connected to the pyrazolyl ring at either a ring nitrogen atom or at a ring carbon atom. [00103] As used herein, “heterocycloalkyl” means any cyclic nonaromatic ring system comprising at least one heteroatom (e.g., N, S, or O) ring atom. This includes 3- to 12-membered monocyclic and fused bicyclic ring systems, and any larger multi-ring fused ring systems, as long as such ring systems do not comprise any aromatic carbocyclic or aromatic heterocyclic ring. Exemplary heterocycloalkyl groups include pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydropyranyl, pyranyl, thiopyranyl, aziridinyl, azetidinyl, oxiranyl, methylenedioxyl, chromenyl, barbituryl, isoxazolidinyl, 1,3-oxazolidin-3-yl, isothiazolidinyl, 1,3-thiazolidin-3-yl, 1,2-pyrazolidin-2-yl, 1,3-pyrazolidin-1-yl, piperidinyl, thiomorpholinyl, 1,2-tetrahydrothiazin-2- yl, 1,3-tetrahydrothiazin-3-yl, tetrahydrothiadiazinyl, morpholinyl, 1,2-tetrahydrodiazin-2-yl, 1,3-tetrahydrodiazin-1-yl, tetrahydroazepinyl, piperazinyl, piperizin-2-onyl, piperizin-3-onyl, chromanyl, 2-pyrrolinyl, 3-pyrrolinyl, imidazolidinyl, 2-imidazolidinyl, 1,4-dioxanyl, 8- azabicyclo[3.2.1]octanyl, 3-azabicyclo[3.2.1]octanyl, 3,8-diazabicyclo[3.2.1]octanyl, 2,5- diazabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.2]octanyl, octahydro-2H-pyrido[1 ,2- a]pyrazinyl, 3-azabicyclo[4.1.0]heptanyl, 3-azabicyclo[3.1.0]hexanyl, 2-azaspiro[4.4]nonanyl, 7- oxa-1-aza-spiro[4.4]nonanyl, 7-azabicyclo[2.2.2]heptanyl, octahydro-1H-indolyl, etc. In general, the heterocycloalkyl group typically is attached to the main structure via a carbon atom or a nitrogen atom. Of course, other heterocycloalkyl groups will be readily apparent to those of skill in the art given the benefit of the present disclosure. [00104] It is understood that the term “bridged bicyclic 7-12 membered heterocycloalkyl ring” has its common understanding, which is a heterocyclic ring system formed by two rings wherein the bridgehead ring atoms are separated by bridges having at least one atom (either a carbon atom or a heteroatom). The entire bicyclic system contains from 7 to 12 ring atom members, for example, wherein 1 to 4 of these atoms are heteroatoms (e.g., N, O, or S). Such bicyclic systems include bridged morpholine rings, bridged piperidine rings, bridged piperazine rings, and the like. Preferably, at least one of the rings is a 5, 6, or 7-membered ring. [00105] As used herein, the term “bridged morpholine” refers to a morpholine having one or two bridges between carbon atoms on opposite sides of the morpholine ring, wherein said bridges consists of either one, two or three bridge atoms with any attached hydrogen atoms or substituents. Preferably, the bridges are saturated bridges, but alternatively, one or more bridges may be unsaturated. For example, at least the following bridged morpholine rings are contemplated, wherein each carbon may be optionally substituted as provided herein: wherein each G is independently O, S, or NR 2 as defined herein. [00106] As used herein, “cycloalkyl” means a nonaromatic saturated or unsaturated free radical forming at least one ring consisting essentially of 3 to 10 carbon atoms and a corresponding number of hydrogen atoms. The term “cycloalkyl” therefore includes cycloalkenyl groups, as further defined below. As such, cycloalkyl groups can be monocyclic or polycyclic. Individual rings of such polycyclic cycloalkyl groups can have different connectivities, e.g., fused, bridged, spiro, etc., in addition to covalent bond substitution. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornanyl, bicyclo[3.2.1]octanyl, octahydro-pentalenyl, spiro[4.5]decanyl, cyclopropyl, adamantyl, substituted with cyclobutyl, cyclobutyl substituted with cyclopentyl, cyclohexyl substituted with cyclopropyl, etc. [00107] It is understood that when describing the substituents attached in various positions to the core structure described herein, including substituents attached to substituents, in some cases, the substituent may be referred to using the name of the corresponding chemical compound, especially in the case of rings, whereas in some cases the same substituent may be referred to using the name of the corresponding chemical radical (e.g., having an “-yl” suffix), but these terms are interchangeable. For example, when referring to a heteroaryl ring substituent, the terms “pyridine” and “pyridyl” are equivalent, as are the terms “morpholine” and “morpholinyl.” The skilled artisan will recognize that such terms are used to denote attachment of, for example, pyridine or morpholine ring at the designated position, thus converting said ring to a pyridyl or morpholinyl substituent (radical) respectively. Absent an indication otherwise, such attachments may be made at any chemically permissible location of the attached ring. EXAMPLES [00108] Unless otherwise noted, starting materials, reagents, and solvents were obtained from commercial suppliers (e.g., Acros Organics, Sigma-Aldrich, Alfa Aesar, Fluorochem, and Merck) and were used without further purification. Reactions were routinely monitored by thin- layer chromatography (TLC) performed on silica gel 60 F 2 54 (layer 0.2 mm) pre-coated aluminum foil (with fluorescent indicator UV254) (Sigma-Aldrich). Developed plates were air- dried and visualized under UV light (254/365 nm) or by using KMnO 4 or ninhydrin solutions. Flash column chromatography was performed on Merck silica gel 60 (mesh 230-400). Automated flash chromatographic purifications were performed using Biotage® Selekt (Cartridge: Sfär Silica HC Duo 5g or 10g). Preparative TLC purification was performed on Merck silica gel 60 F254 (0.5 mm) pre-coated glass plates (20x20 cm) (Sigma-Aldrich) [00109] 1 H NMR and 13 C NMR spectra were recorded at room temperature at 400 and 101 MHz, respectively, on a Bruker Avance 400 spectrometer by using TMS or residual solvent peak as internal standard. Chemical shifts are reported in ppm (δ) and the coupling constants (J) are given in Hertz (Hz). Peak multiplicities are abbreviated as follow: s (singlet), bs (broad singlet), d (doublet), dd (double doublet), t (triplet), dt (double triplet), q (quartet), p (pentet), and m (multiplet). [00110] High-Resolution Mass Spectroscopy (HRMS) spectra were registered on Agilent Technologies 6540 UHD Accurate Mass Q-TOF LC-MS system or on Agilent 1290 Infinity Series U-HPLC system (Agilent Technologies, Santa Clara, CA, USA) coupled with a Q-TOF 6540 high-resolution mass spectrometer and 1290 Infinity Series DAD/UV-Vis detector (Agilent Technologies). The purity of all final compounds that were evaluated in biological assays was assessed as >95%, using LC-MS. The analyses were carried out according to the method listed below. The mobile phase was a mixture of water (solvent A) and acetonitrile (solvent B), both containing formic acid at 0.1%. Method: Acquity UPLC BEH C181.7 µm ( C18, 150 x 2.1 mm) column at 40° C using a flow rate of 0.65 mL/min in a 10 min gradient elution. Gradient elution was as follows: 99.5:0.5 (A/B) to 5:95 (A/B) over 8 min, 5:95 (A/B) for 2 min, and then reversion back to 99.5:0.5 (A/B) over 0.1 min. The UV detection is an averaged signal from wavelength of 190 nm to 640 nm and mass spectra are recorded on a mass spectrometer using positive mode electro spray ionization. [00111] Compounds described herein may be synthesized as described herein, using modified methods described herein or by methods known to a person of skill in the art. [00112] Chemistry abbreviations: ACN, acetonitrile; Boc, tert-butoxycarbonyl; CD3OD, deuterated methanol; CDCl3, deuterated chloroform; DCE, dichloroethane; DCM, dichloromethane; DEE, diethyl ether; DIPEA, N,N’-diisopropylethylamine; DMA, dimethylacetamide; DMF, dimethylformamide; DMSO, dimethylsulfoxide; DMSO-d6, deuterated dimethylsulfoxide; EA, ethyl acetate; h, hour; EtOH, absolute ethanol; Et3N, triethylamine; HATU, 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate; min, minutes; HRMS, high-resolution mass spectroscopy; MeOH, methanol; NMR, nuclear magnetic resonance; tBu, tert-butyl; tBuOH, tert-butanol; TLC, thin- layer chromatography; PE, petroleum ether; rt, room temperature. Chemical Synthesis [00113] Compounds of general formula (I) may be prepared by the general synthetic approaches described below (General Scheme 1 and 2), together with synthetic methods known in the art of organic chemistry. In all methods, it is well-understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts (1999) Protective Groups in Organic Synthesis, 3' edition, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of Formula (I). [00114] General Scheme 1: [00116] ARB: Androgen Receptor (AR) Binder; E3LB: E3 Ligase Binder. [00117] The compounds of the present disclosure can generally be made by adapting the procedures known in the art for making related compounds according to General Scheme 1 and General Scheme 2, above. [00118] Specific detailed synthetic procedures for a variety of intermediates and related compounds to those within the scope of the present disclosure can be found in patents and patent application publications US 2020/0239430, US 2020/0282068, US 2023/0132823, US 2023/0134817, U.S. 11,098,025, U.S. 11,787,778, U.S. 11,547,759, WO 2021/236695, WO 2023//039601, WO 2023/039602, WO 2023/039603, and WO 2023/039604, the contents of each of which are hereby incorporated by reference in their entireties. These references provide specific intermediates and procedures for formation of a variety of ARB moieties, E3LB moieties, and linker moieties, useful for preparing the compounds disclosed herein. [00119] Synthetic Examples [00120] Scheme 1. Synthesis of ARB-1 moiety: [00121] 2,3-Difluorophenyl acetate (A) [00122] The title compound can be prepared according to the process described by Huifang et al., J. Med. Chem.2014, 57, 6458-6467. [00123] Acetyl chloride (6.01 mL, 6.63 g, 84.55 mmol) is slowly added at room temperature to a stirred solution of 2,3-difluorophenol (10.0 g, 76.87 mmol) and pyridine (6.83 mL, 6.68 g, 84.55 mmol) in dry DCM (60.0 mL). After 2 h, the mixture is diluted with 2N HCl (60 mL) and the aqueous layer is separated and extracted with DCM (30 mLx3). The reunited organic phases are washed with brine (20 mLx2), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure to afford the title compound (13.26 g, 99% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.16-7.03 (m, 2H), 6.99-6.83 (m, 1H), 2.37 (s, 3H); 13 C NMR (101 MHz, CDCl 3 ): δ 167.96, 151.26 (dd, J = 10.9, 249.4 Hz), 143.29 (dd, J = 14.3, 251.5 Hz), 139.55 (dd, J = 2.3, 10.1 Hz), 123.36 (dd, J = 5.0, 7.9 Hz), 118.78 (d, J = 3.5 Hz), 114.71 (d, J = 17.3 Hz), 20.44. [00124] 1-(3,4-Difluoro-2-hydroxyphenyl)ethan-1-one (B). [00125] The title compound can be prepared according to the process described by Gomtsyan, et al., WO2010/045401. [00126] AlCl 3 (1.55 g, 11.62 mmol) is added under nitrogen at 0 °C in small portions to a stirred solution of 2,3-difluorophenyl acetate (A) (2.00 g, 11.62 mmol) in DCE (3.0 mL). After the addition is completed, the mixture is refluxed for 14 h. After cooling at rt, the solvent is evaporated and the residue diluted with DCM (20 mL).2N HCl (10 mL) is added, and the mixture is stirred for 20 min. The organic phase is separated, the water phase is extracted with DCM (10 mLx2), and the reunited organic phases are dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to give the title compound (1.95 g, 97% yield) as a brownish solid. 1 H NMR (400 MHz, CDCl 3 ): δ 12.59 (d, J = 1.4 Hz, 1H), 7.55 (ddd, J = 2.3, 5.5, 9.1 Hz, 1H), 6.86-6.63 (m, 1H), 2.65 (s, 3H); 13 C NMR (101 MHz, CDCl 3 ): δ 203.66, 157.02, 154.88 (dd, J = 9.7, 257.9 Hz), 139.98 (dd, J = 13.7, 249.7 Hz), 125.92 (dd, J = 4.5, 10.0 Hz), 117.77, 107.24 (d, J = 18.7 Hz), 26.78 [00127] 2-Bromo-1-(3,4-difluoro-2-hydroxyphenyl)ethan-1-one (C). [00128] The title compound can be prepared according to the process described by Huifang Li et al., J. Med. Chem.2014, 57, 6458-6467 [00129] A solution of 1-(3,4-difluoro-2-hydroxyphenyl)ethan-1-one (B) (1.95 g, 11.33 mmol) in EA (40.0 mL) is added dropwise at rt to a stirred suspension of CuBr 2 (3.03 g, 13.59 mmol) in EA (40 mL). After 24 h of reflux, the mixture is allowed to cool to rt, filtered over Celite, and the filtrate is evaporated to dryness. The crude residue is purified by flash column chromatography on SiO 2 (PE/EA, 95:5) to give the title compound (1.55 g, 69% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ): δ 12.10-11.84 (m, 1H), 7.58 (ddd, J = 2.2, 5.4, 8.9 Hz, 1H), 6.93-6.65 (m, 1H), 2.65 (s, 2H) ; 13 C NMR (101 MHz, CDCl 3 ): δ 196.41, 155.36 (dd, J = 9.7, 259.7 Hz), 154.11 (dd, J = 5.6, 9.6 Hz), 140.29 (dd, J = 13.9, 251.0 Hz), 125.92 (dd, J = 4.6, 10.1 Hz), 115.13, 107.90 (d, J = 19.1 Hz), 29.31. [00130] 2,3-Difluoro-6-(2-morpholinothiazol-4-yl)phenol (ARB-1). [00131] The title compound can be prepared according to the process described by Huifang Li et al., J. Med. Chem.2014, 57, 6458-6467. [00132] Morpholine-4-carbothioamide (0.330 g, 2.26 mmol) is added in small portions to a stirred solution of compound 2-bromo-1-(3,4-difluoro-2-hydroxyphenyl)ethan-1-one (C) (0.568 g, 2.26 mmol) in absolute EtOH (10 mL) at 0 °C. When addition is completed, the mixture is refluxed for 4 h. After cooling to rt, the mixture is evaporated to dryness and NaHCO 3 saturated solution (20 mL) is added to pH 8. The aqueous phase is extracted with EA (10 mLx3), the reunited organic phases are dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to give a solid which is triturated with DEE and filtered, affording the title compound (0.590 g, 87% yield) as a light-yellow solid. 1 H NMR (400 MHz, CDCl 3 ): δ 12.43 (s, 1H), 7.24 (ddd, J = 2.3, 5.6, 8.5 Hz, 1H), 6.76 (s, 1H), 6.66 (td, J = 7.2, 9.3 Hz, 1H), 4.00-3.84 (m, 4H), 3.63-3.43 (m, 4H); 13 C NMR (101 MHz, CDCl 3 ): δ 171.13, 151.23 (dd, J = 10.4, 247.9 Hz), 148.09, 147.25-146.05 (m), 140.72 (dd, J = 14.2, 244.7 Hz), 119.63 (dd, J = 4.5, 8.7 Hz), 115.71, 106.97 (d, J = 18.4 Hz), 100.25 (d, J = 1.7 Hz), 65.90 (2C), 48.31 (2C). [00133] Tert-butyl 2-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)acetate (D). [00134] The title compound can be prepared according to the process described in US 2020/0282068. [00135] Tert-butyl bromoacetate (0.434 mL, 0.575 g, 2.95 mmol) is added to a stirred suspension of 2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenol (ARB-1) (0.800 g, 2.68 mmol) and K2CO3 (0.927 g, 6.71 mmol) in ACN (10.0 mL). The suspension is stirred for 18 h at rt, filtered, and the filtrate is evaporated to dryness. The residue is purified by flash column chromatography on SiO 2 (PE/EA, 95:5 to 90:10) to afford the title compound (0.800 g, 73% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ): δ 7.92 (ddd, J = 2.3, 6.2, 8.8 Hz, 1H), 7.68 (s, 1H), 7.10-6.87 (m, 1H), 4.67 (d, J = 1.7 Hz, 2H), 4.00-3.79 (m, 4H), 3.68-3.47 (m, 4H), 1.53 (s, 9H); 13 C NMR (101 MHz, CDCl 3 ): δ 169.77, 167.40, 150.32 (dd, J = 11.8, 249.8 Hz), 145.30, 144.64-144.23 (m), 143.98 (dd, J = 14.4, 246.1 Hz), 124.35, 124.04 (dd, J = 3.9, 7.8 Hz), 111.42 (d, J = 17.0 Hz), 107.71, 82.55, 70.00 (d, J = 7.5 Hz), 66.22 (2C), 48.61 (2C), 28.09 (3C). [00136] 2-(2,3-Difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)acetic acid (E). [00137] The title compound can be prepared according to the process described in US 2020/0282068. [00138] A solution of 4N HCl in dioxane (15 mL) is added to tert-butyl 2-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)acetate (D) (0.780 g, 1.89 mmol) and the resulting suspension is stirred at rt for 16 h. The solvent is evaporated to dryness and the residue is triturated with DEE. The solids are collected by filtration and dried under vacuo to afford the title compound (0.672 g, 91% yield) as a light-yellow solid. 1 H NMR (400 MHz, CD3OD): δ 7.48 (ddd, J = 2.3, 5.7, 8.1 Hz, 1H), 7.27 (s, 1H), 7.18 (td, J = 7.5, 9.2 Hz, 1H), 5.01 (d, J = 1.5 Hz, 2H), 4.02-3.86 (m, 4H), 3.86- 3.67 (m, 4H); 13 C NMR (101 MHz, CD 3 OD): δ 171.72, 169.44, 152.57 (dd, J = 11.6, 252.2 Hz), 144.55 (d, J = 1.7 Hz), 143.30 (dd, J = 15.0, 248.0 Hz), 136.41 (d, J = 13.3 Hz), 124.82 (dd, J = 3.9, 8.6 Hz), 118.47, 111.66 (d, J = 18.2 Hz), 106.15, 69.24 (d, J = 9.0 Hz), 65.05 (2C), 49.17 (2C). [00139] Scheme 2. Synthesis of E3LB-1 moiety.

[00140] 4-(2,6-Bis(benzyloxy)pyridin-3-yl)phenol (G). [00141] The title compound can be prepared according to the process described by Min. et al., Angew. Chem. Int. Ed. Engl.2021, 60, 26663-26670. [00142] 2,6-Bis(benzyloxy)-3-bromopyridine (F) (0.100 g, 0.270 mmol), (4- hydroxyphenyl)boronic acid (0.745 g, 0.540 mmol) and K3PO4 (0.123 g, 0.581 mmol) in a mixture (6:1) of dry dioxane (3.0 ml) and water (0.5 ml) are degassed under nitrogen for 10 min and then 1,1'-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (0.022 g, 0.027 mmol) is added. The reaction mixture is stirred at 110° C for 16 hours. After cooling to room temperature, the reaction mixture is filtered through a short pad of celite. The filtrate is diluted with EA (20.0 mL), washed with water (10 mLx2), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The crude product is purified by automated flash chromatography on SiO 2 cartridge (PE/EA, 93:7) to afford the title compound as a white solid (82 mg, 80% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.57 (d, J = 8.0 Hz, 1H), 7.50 – 7.42 (m, 4H), 7.42 – 7.27 (m, 8H), 6.86 (d, J = 8.6 Hz, 2H), 6.46 (d, J = 8.0 Hz, 1H), 5.43 (s, 2H), 5.36 (s, 2H), 4.78 (s, 1H). [00143] Tert-butyl 2-(4-(2,6-bis(benzyloxy)pyridin-3-yl)phenoxy)acetate (H). [00144] The title compound can be prepared according to the process described by Min J. et al., Angew. Chem. Int. Ed. Engl.2021, 60, 26663-26670. [00145] To the solution of 4-(2,6-bis(benzyloxy)pyridin-3- yl)phenol (G) (0.074 g, 0.193 mmol) and K 2 CO 3 (0.040 g, 0.212 mmol) in DMF (1.8 ml) is added tert-butyl 2-bromoacetate (0.031 mL, 0.212 mmol) and the reaction mixture is stirred at room temperature for 18 h. The reaction mixture is diluted with water (20.0 mL) and then extracted with EA (10mLx3). The organic layer is washed with brine (10mLx2), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford the title compound as a colorless oil (0.088 g, 92% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.57 (d, J = 8.0 Hz, 1H), 7.52 – 7.47 (m, 2H), 7.45 – 7.41 (m, 2H), 7.39 – 7.27 (m, 8H), 6.94 – 6.88 (m, 2H), 6.46 (d, J = 8.1 Hz, 1H), 5.42 (s, 2H), 5.36 (s, 2H), 4.54 (s, 2H), 1.49 (s, 9H). [00146] Tert-butyl 2-(4-(2,6-dioxopiperidin-3-yl)phenoxy)acetate (I). [00147] The title compound can be prepared according to the process described by Min et al., Angew. Chem. Int. Ed. Engl.2021, 60, 26663-26670. [00148] Under nitrogen atmosphere, to a suspension of tert-butyl 2-(4-(2,6- bis(benzyloxy)pyridin-3-yl)phenoxy)acetate (H) (0.084 g, 0.169 mmol) in absolute EtOH (1.8 ml) is added Pd/C (10% Pd, 0.0072 g, 0.675 mmol) and then the reaction mixture is stirred under hydrogen atmosphere at room temperature for 18 h. The reaction mixture is then filtered through a short pad of celite and washed with EA. The filtrate is concentrated to dryness to provide the title compound as a pale-pink oil (0.055 g, 65% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.29 (s, 1H), 7.12 (d, J = 8.7 Hz, 2H), 6.88 (d, J = 8.7 Hz, 2H), 4.49 (s, 2H), 3.73 (dd, J = 9.5, 5.2 Hz, 1H), 2.76 – 2.54 (m, 2H), 2.30 – 2.14 (m, 2H), 1.48 (s, 9H). [00149] 2-(4-(2,6-Dioxopiperidin-3-yl)phenoxy)acetic acid (E3LB-1). [00150] The title compound can be prepared according to the process described by Min J. et al., Angew. Chem. Int. Ed. Engl.2021, 60, 26663-26670. [00151] To a solution of tert-butyl 2-(4-(2,6-dioxopiperidin-3- yl)phenoxy)acetate (I) (0.050 g, 0.157 mmol) in dry DCM (1.6 ml) is added 2,2,2-trifluoroacetic acid (0.48 mL, 6.28 mmol) and the reaction mixture is stirred at room temperature for 4 h. Then, the solvent is concentrated under vacuum and the crude is triturated with DEE to give the title compound as a white solid (0.041 g, 100% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.97 (bs, 1H), 10.79 (s, 1H), 7.13 (d, J = 8.5 Hz, 2H), 6.86 (d, J = 8.5 Hz, 2H), 4.65 (s, 2H), 3.79 (dd, J = 11.4, 4.7 Hz, 1H), 2.72 – 2.58 (m, 1H), 2.48 – 2.41 (m, 1H), 2.23 – 2.08 (m, 1H), 2.06 – 1.94 (m, 1H). [00152] Scheme 3. Synthesis of Example 1.

[00153] Tert-butyl (8-(2-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy) acetamido)octyl)carbamate (J). [00154] Under a nitrogen atmosphere, to a stirred solution of 2-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)acetic acid (E) (0.235 g, 0.659 mmol), tert-butyl (8- aminooctyl)carbamate (0.161 g, 0.659 mmol) and DIPEA (0.35 mL, 1.977 mmol) in dry DMF (2.0 mL) at 0° C is added HATU (0.313 g, 0.823 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water and then extracted with EA (10 mLx3) and the reunited organic phases are washed with water (10mLx3), brine (10mLx3), dried over Na 2 SO 4 , and evaporated to dryness. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 98:2) to afford the title compound as a white solid (0.241 mg, 63% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.58 (ddd, J = 8.5, 5.9, 2.3 Hz, 1H), 7.02 – 6.93 (m, 2H), 6.91 (s, 1H), 4.57 (s, 2H), 4.52 (s, 1H), 3.88 – 3.77 (m, 4H), 3.59 – 3.49 (m, 4H), 3.35 – 3.24 (m, 2H), 3.16 – 3.03 (m, 2H), 1.54 – 1.40 (m, 13H), 1.34 – 1.25 (m, 8H). [00155] N-(8-Aminooctyl)-2-(2,3-difluoro-6-(2-morpholinothiazol-4- yl)phenoxy)acetamide hydrochloride (K). [00156] To a solution of tert-butyl (8-(2-(2,3-difluoro-6-(2-morpholinothiazol-4- yl)phenoxy)acetamido)octyl)carbamate (J) (0.230 g, 0.395 mmol) in dry DCM (1.0 ml) is added 4.0 N HCl in dioxane (2.3 mL) and the reaction is stirred at room temperature for 3 h. The solvent is evaporated to dryness and the solid is triturated with DEE and collected by filtration, yielding the title compound (0.202 g, 99% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.23 (t, J = 5.8 Hz, 1H), 7.89 (bs, 3H), 7.83 – 7.77 (m, 1H), 7.65 (s, 1H), 7.28 – 7.19 (m, 1H), 4.58 (s, 2H), 3.80 – 3.68 (m, 4H), 3.48 – 3.37 (m, 4H), 3.19 – 3.08 (m, 2H), 2.80 – 2.70 (m, 2H), 1.60 – 1.48 (m, 2H), 1.48 – 1.36 (m, 2H), 1.36 – 1.17 (m, 8H). [00157] 2-(2,3-Difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)-N-(8-(2 -(4-(2,6- dioxopiperidin-3-yl)phenoxy)acetamido)octyl)acetamide (Example 1). [00158] Under a nitrogen atmosphere, to a stirred solution of 2-(4-(2,6-dioxopiperidin-3- yl)phenoxy)acetic acid (E3LB-1) (0.045 g, 0.171 mmol), N-(8-aminooctyl)-2-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)acetamide hydrochloride (K) (0.089 g, 0.171 mmol) and DIPEA (0.12 mL, 0.684 mmol) in dry DMF (2.0 mL) at 0° C is added HATU (0.081 g, 0.214 mmol) and the reaction mixture is stirred at room temperature for 3 h. The reaction mixture is poured into ice- water yielding a precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 95:5) followed by preparative TLC eluting with DCM:MeOH 95:5 to afford the title compound as a white solid (0.033 g, 27% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.05 (bs, 1H), 7.63 – 7.54 (m, 1H), 7.16 (d, J = 8.7 Hz, 2H), 7.03 – 6.89 (m, 5H), 6.60 – 6.50 (m, 1H), 4.56 (s, 2H), 4.48 (s, 2H), 3.89 – 3.78 (m, 4H), 3.73 (dd, J = 9.8, 5.3 Hz, 1H), 3.55 – 3.47 (m, 4H), 3.39 – 3.24 (m, 4H), 2.78 – 2.61 (m, 2H), 2.31 – 2.16 (m, 2H), 1.58 – 1.45 (m, 4H), 1.36 – 1.23 (m, 8H); 13 C NMR (101 MHz, CDCl 3 ) δ 173.06, 172.03, 170.80, 167.87, 167.86, 156.77, 150.69 (dd, J = 251.1 Hz and 11.5 Hz), 146.52-146.42 (m), 144.63 (dd, J = 9.1 and 1.6 Hz), 144.21 (dd, J = 247.3 Hz and 14.0 Hz), 130.65, 129.54 (2C), 125.31 (d, J = 3.5 Hz), 124.50 (dd, J = 7.7 Hz and 4.0 Hz), 115.15 (2C), 112.45 (d, J = 17.1 Hz), 106.08, 72.41 (d, J = 5.1 Hz), 67.53, 66.14 (2C), 48.55 (2C), 47.24, 39.07, 39.05, 31.09, 29.50, 29.46, 29.12 (2C), 26.75 (2C), 26.38. HRMS (ESI) m/z [M+H]+ calcd for C 36 H 43 F 2 N 5 O 7 S 728.29240, found 728.29292. [00159] Scheme 4. Synthesis of Examples 2-3.

[00160] Tert-butyl 4-(2-(2-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy) ethoxy)ethyl)piperazine-1-carboxylate (L). [00161] Under a nitrogen atmosphere, DIAD (0.150 mL, 0.737 mmol) is slowly added dropwise to a stirred ice-cold solution of 2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenol (ARB-1) (0.200 g, 0.670 mmol), tert-butyl 4-(2-(2-hydroxyethoxy)ethyl)piperazine-1-carboxylate (e.g., prepared as described by Rossello et al. WO 2008/113756) (0.202 g, 0.737 mmol), and PPh 3 (0.193 g, 0.737 mmol) in dry THF (4.0 mL). The solution is stirred at 0 °C for 30 min, then at rt for 16 h. The reaction mixture is evaporated to dryness and the crude residue is directly purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 95:5) to give the title compound as a yellow oil (0.255 g, 69% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.87 (t, J = 6.7 Hz, 1H), 7.57 – 7.44 (m, 1H), 6.93 (dd, J = 16.7, 9.1 Hz, 1H), 4.31 – 4.23 (m, 2H), 3.88 – 3.77 (m, 6H), 3.73 – 3.60 (m, 2H), 3.54 – 3.41 (m, 8H), 2.79 – 2.32 (m, 6H), 1.46 (s, 9H). [00162] Tert-butyl (2-(2-(2-(2-(2,3-difluoro-6-(2-morpholinothiazol-4- yl)phenoxy)ethoxy)ethoxy)ethoxy)ethyl)carbamate (M). [00163] Under a nitrogen atmosphere, DIAD (0.087 mL, 0.442 mmol) is slowly added dropwise to a stirred ice-cooled solution of 2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenol (ARB-1) (0.120 g, 0.402 mmol), tert-butyl (2-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)ethyl)carbamate (0.130 g, 0.442 mmol), and PPh3 (0.116 g, 0.442 mmol) in dry THF (5.0 mL). The solution is stirred at 0 °C for 30 min, then at rt for 16 h. The reaction mixture is quenched with water (20 mL) and extracted with EA (10 mLx3). The reunited organic phases are dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford a crude residue which is purified by flash column chromatography on SiO 2 (DCM/EA, 7:3) to give the title compound as yellow oil, which solidified upon standing at rt (0.148 g, 93% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.90 (ddd, J = 8.9, 6.3, 2.3 Hz, 1H), 7.57 (s, 1H), 6.99-6.90 (m, 1H), 5.02 (s, 1H), 4.36-4.26 (m, 2H), 3.93-3.82 (m, 6H), 3.75-3.59 (m, 8H), 3.58-3.49 (m, 6H), 3.39-3.25 (m, 2H), 1.46 (s, 9H). [00164] 4-(4-(3,4-Difluoro-2-(2-(2-(piperazin-1-yl)ethoxy)ethoxy)phe nyl)thiazol-2- yl)morpholine dihydrochloride (N). [00165] A solution of 4N HCl in dioxane (2.40 mL) is added to a solution of tert-butyl 4-(2-(2- (2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)ethoxy)eth yl)piperazine-1-carboxylate (L) (0.240 g, 0.433 mmol) in dry DCM (1.0 mL) and the resulting solution is stirred at rt for 3 h. The solvent is evaporated to dryness and the residue is triturated with DEE, yielding a solid which is collected by filtration and dried under vacuo to afford the title compound (0.227 g, 99% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.99 (bs, 1H), 9.95 – 9.74 (m, 2H), 7.85 (ddd, J = 8.8, 6.4, 2.1 Hz, 1H), 7.48 (s, 1H), 7.22 (dd, J = 17.1, 9.4 Hz, 1H), 4.32 – 4.25 (m, 2H), 3.93 – 3.86 (m, 2H), 3.85 – 3.79 (m, 2H), 3.77 – 3.70 (m, 4H), 3.70 – 3.60 (m, 2H), 3.51 – 3.30 (m, 12H). [00166] 2-(2-(2-(2-(2,3-Difluoro-6-(2-morpholinothiazol-4- yl)phenoxy)ethoxy)ethoxy)ethoxy)ethan-1-amine hydrochloride (O). [00167] A solution of 4N HCl in dioxane (2.0 mL) is added to tert-butyl (2-(2-(2-(2-(2,3- difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)ethoxy)ethoxy)e thoxy)ethyl)carbamate (M) (0.140 g, 0.244 mmol) and the resulting solution is stirred at rt overnight. The solvent is evaporated to dryness and the residue is triturated with DEE, yielding a solid which is collected by filtration and dried under vacuo to afford the title compound (0.115 g, 93% yield) as white solid. 1 H NMR (400 MHz, MeOD) δ 7.59 (s, 1H), 7.43 (s, 1H), 7.15 (dd, J = 16.8, 8.8 Hz, 1H), 4.39 (s, 2H), 3.91 (s, 4H), 3.82 (s, 2H), 3.77-3.55 (m, 14H), 3.15 (s, 2H). [00168] 3-(4-(2-(4-(2-(2-(2,3-Difluoro-6-(2-morpholinothiazol-4- yl)phenoxy)ethoxy)ethyl)piperazin-1-yl)-2-oxoethoxy)phenyl)p iperidine-2,6-dione (Example 2). [00169] Under a nitrogen atmosphere, to a solution of 2-(4-(2,6-Dioxopiperidin-3- yl)phenoxy)acetic acid (E3LB-1) (0.014 g, 0.053 mmol), 4-(4-(3,4-difluoro-2-(2-(2-(piperazin-1- yl)ethoxy)ethoxy)phenyl)thiazol-2-yl)morpholine dihydrochloride (N) (0.028 g, 0.053 mmol), and DIPEA (0.037 µL, 0.212 mmol) in dry DMF (2.0 mL) is added HATU (0.025 g, 0.066 mmol) at 0 °C. The resulting mixture is allowed to warm to rt and stirred for 3 h. The reaction mixture is poured into ice-water yielding a white precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 95:5) to give the title product as a white solid (0.018 g, 49% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.02 (bs, 1H), 7.87 (ddd, J = 8.8, 6.3, 2.2 Hz, 1H), 7.51 (s, 1H), 7.12 (d, J = 8.6 Hz, 2H), 6.99 – 6.87 (m, 3H), 4.67 (s, 2H), 4.30 – 4.22 (m, 2H), 3.88 – 3.76 (m, 6H), 3.72 (dd, J = 9.8, 5.3 Hz, 1H), 3.69 – 3.60 (m, 4H), 3.61 – 3.54 (m, 2H), 3.53 – 3.47 (m, 4H), 2.78 – 2.58 (m, 4H), 2.57 – 2.45 (m, 4H), 2.31 – 2.14 (m, 2H); 13 C NMR (101 MHz, CDCl 3 ) δ 173.16, 172.12, 169.68, 166.12, 157.40, 150.36 (dd, J = 249.8 and 11.6 Hz), 145.63 (dd, J = 3.3 and 1.7 Hz), 145.46 (dd, J = 9.0 and 1.4 Hz), 144.64 (dd, J = 246.2 and 13.7 Hz), 130.03, 129.30 (2C), 124.99 (d, J = 3.9 Hz), 123.87 (dd, J = 7.8 and 4.0 Hz), 115.10 (2C), 111.49 (d, J = 17.0 Hz), 107.19, 72.53 (d, J = 5.8 Hz), 70.21, 69.00, 67.64, 66.20 (2C), 57.74, 53.70, 53.18, 48.60 (2C), 47.21, 45.24, 42.01, 31.01, 26.39. HRMS (ESI) m/z [M+H]+ calcd for C 34 H 39 F 2 N 5 O 7 S 700.26110, found 700.26124. [00170] N-(2-(2-(2-(2-(2,3-Difluoro-6-(2-morpholinothiazol-4- yl)phenoxy)ethoxy)ethoxy)ethoxy)ethyl)-2-(4-(2,6-dioxopiperi din-3-yl)phenoxy)acetamide (Example 3). [00171] Under a nitrogen atmosphere, to a solution of 2-(4-(2,6-Dioxopiperidin-3- yl)phenoxy)acetic acid (E3LB-1) (0.030 g, 0.114 mmol), 2-(2-(2-(2-(2,3-Difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)ethoxy)ethoxy)ethoxy)ethan-1- amine hydrochloride (O) (0.058 g, 0.114 mmol), and DIPEA (0.079 µL, 0.456 mmol) in dry DMF (2.0 mL) is added HATU (0.054 g, 0.143 mmol) at 0 °C. The resulting mixture is allowed to warm to rt and stirred for 2 h. The reaction mixture is dropped into ice-water (30 mL) and extracted with EA (15 mLx3). The combined organic phases are washed with water (15 mLx3), brine (20 mLx2), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The crude product is then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 95:5) to give the title product as a white solid (0.046 g, 56% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.01 (bs, 1H), 7.87 (ddd, J = 8.9, 6.2, 2.3 Hz, 1H), 7.53 (s, 1H), 7.17 – 7.10 (m, 2H), 7.01 – 6.96 (m, 1H), 6.95 – 6.87 (m, 3H), 4.48 (s, 2H), 4.29 – 4.22 (m, 2H), 3.87 – 3.77 (m, 6H), 3.70 (dd, J = 10.0, 5.3 Hz, 1H), 3.68 – 3.48 (m, 16H), 2.81 – 2.57 (m, 2H), 2.30 – 2.12 (m, 2H); 13 C NMR (101 MHz, CDCl 3 ) δ 173.00, 172.02, 169.71, 168.00, 156.77, 150.34 (dd, J = 249.6 and 11.6 Hz), 145.59-145.49 (m), 145.44 (d, J = 1.4 Hz), 144.63 (dd, J = 246.3 and 13.6 Hz) 130.57, 129.48 (2C), 125.01 (d, J = 3.4 Hz), 123.85 (dd, J = 7.8 and 4.0 Hz), 115.17 (2C), 111.42 (d, J = 17.0 Hz), 107.41, 72.59 (d, J = 5.9 Hz), 70.63, 70.60, 70.58, 70.38, 70.28, 69.68, 67.48, 66.21 (2C), 48.60 (2C), 47.23, 38.80, 31.10, 26.35. HRMS (ESI) m/z [M+H]+ calcd for C 34 H 40 F 2 N 4 O 9 S 719.25568, found 719.25635. [00172] Scheme 5. Synthesis of E3LB-2 moiety and linker connection. [00173] 2-(2-(2-hydroxyethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (P). [00174] Under a nitrogen atmosphere, to a solution of 2,2'-(ethane-1,2-diylbis(oxy))diethanol (1.5 g, 9.988 mmol) in dry DCM (10.0 mL) at 0° C, Et 3 N (0.208 mL, 1.498 mmol) and 4- methylbenzene-1-sulfonyl chloride (0.190 g, 0.999 mmol) are added, and the mixture is stirred at room temperature for 18 h. The reaction mixture is diluted with water and extracted with DCM (3x20mL). The combined organic layers are washed with 5% citric acid (3x0.5mL), dried over Na 2 SO 4 , filtered and evaporated to dryness to afford the title compound as a colorless oil (258 mg, 85%). 1 H NMR (400 MHz, CDCl 3 ) δ 7.80 (d, J = 8.1 Hz, 2H), 7.34 (d, J = 8.0 Hz, 2H), 4.20 – 4.14 (m, 2H), 3.74 – 3.67 (m, 4H), 3.61 (s, 4H), 3.60 – 3.55 (m, 2H), 2.45 (s, 3H). [00175] 2-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (Q). [00176] Under a nitrogen atmosphere, to a solution of 2,2'-((oxybis(ethane-2,1- diyl))bis(oxy))diethanol (1.5 g, 7.723 mmol) in dry DCM (10.0 mL) at 0° C, Et3N (0.160 mL, 1.158 mmol) and 4-methylbenzene-1-sulfonyl chloride (0.147 g, 0.772 mmol) are added, and the mixture is stirred at room temperature for 18 h. The reaction mixture is diluted with water and extracted with DCM (3x20mL). The combined organic layers are washed with 5% citric acid (3x0.5mL), dried over Na 2 SO 4 , filtered and evaporated to dryness to afford the title compound as a colorless oil (204 mg, 55%). 1 H NMR (400 MHz, CDCl 3 ) δ 7.80 (d, J = 8.3 Hz, 2H), 7.37 – 7.29 (m, 2H), 4.19 – 4.11 (m, 2H), 3.74 – 3.53 (m, 14H), 2.44 (s, 3H). [00177] 2-(2-(2-(4-(2,6-bis(benzyloxy)pyridin-3-yl)phenoxy)ethoxy)et hoxy)ethyl 4- methylbenzenesulfonate (R). [00178] Under a nitrogen atmosphere, DIAD (0.100 mL, 0.498 mmol) is slowly added to a stirred ice-cold solution of 4-(2,6-bis(benzyloxy)pyridin-3-yl)phenol (G) (0.120 g, 0.312 mmol), 2-(2-(2-hydroxyethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (P) (0.152 g, 0.499 mmol), and PPh3 (0.123 g, 0.468 mmol) in dry THF (4.0 mL). The solution is stirred at 0 °C for 30 min, then at rt for 18 h. The solvent is evaporated to dryness and the crude product is purified by automated flash column chromatography on SiO 2 cartridge (PE/EA, 9:1 to 5:5) to give the title compound as a pink solid (0.076 g, 36% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.79 (d, J = 8.3 Hz, 2H), 7.57 (d, J = 8.0 Hz, 1H), 7.50 – 7.46 (m, 2H), 7.45 – 7.41 (m, 2H), 7.40 – 7.27 (m, 10H), 6.95 – 6.90 (m, 2H), 6.46 (d, J = 8.1 Hz, 1H), 5.42 (s, 2H), 5.36 (s, 2H), 4.18 – 4.12 (m, 4H), 3.86 – 3.82 (m, 2H), 3.72 – 3.65 (m, 4H), 3.63 – 3.60 (m, 2H), 2.41 (s, 3H). [00179] 2-(2-(2-(2-(4-(2,6-bis(benzyloxy)pyridin-3-yl)phenoxy)ethoxy )ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (S). [00180] Under a nitrogen atmosphere, DIAD (0.160 mL, 0.790 mmol) is slowly added to a stirred ice-cold solution of 4-(2,6-bis(benzyloxy)pyridin-3-yl)phenol (G) (0.140 g, 0.494 mmol), 2-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (Q) (0.204 g, 0.790 mmol), and PPh 3 (0.191 g, 0.739 mmol) in dry THF (2.0 mL). The solution is stirred at 0 °C for 30 min, then at rt for 48 h. The solvent is evaporated to dryness and the crude is purified by automated flash column chromatography on SiO 2 cartridge (PE/EA, 9:1 to 5:5) to give the title compound as a clear yellow oil (0.178 g, 51% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.82 (d, J = 8.1 Hz, 2H), 7.60 (d, J = 8.0 Hz, 1H), 7.55 – 7.27 (m, 14H), 6.97 (d, J = 8.6 Hz, 2H), 6.49 (d, J = 8.1 Hz, 1H), 5.45 (s, 2H), 5.39 (s, 2H), 4.23 – 4.14 (m, 4H), 3.94 – 3.86 (m, 2H), 3.78 – 3.66 (m, 6H), 3.62 (s, 4H), 2.45 (s, 3H). [00181] 2-(2-(2-(4-(2,6-dioxopiperidin-3-yl)phenoxy)ethoxy)ethoxy)et hyl 4- methylbenzenesulfonate (T). [00182] Under a nitrogen atmosphere, to a suspension of 2-(2-(2-(4-(2,6-bis(benzyloxy)pyridin- 3-yl)phenoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (R) (0.075 g, 0.112 mmol) in dry THF (1.0 ml) is added Pd/C (10% Pd, 0.012 g, 0.112 mmol) and then the reaction mixture is stirred under hydrogen atmosphere at room temperature for 4 h. The reaction mixture is then filtered through a pad of celite and washed with EA. The filtrate is concentrated to dryness to provide a residue which is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 97:3) to afford the title compound as colorless oil (0.054 g, 96% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.25 (s, 1H), 7.78 (d, J = 8.3 Hz, 2H), 7.32 (d, J = 8.2 Hz, 2H), 7.14 – 7.07 (m, 2H), 6.93 – 6.86 (m, 2H), 4.18 – 4.12 (m, 2H), 4.12 – 4.06 (m, 2H), 3.83 – 3.78 (m, 2H), 3.75 – 3.57 (m, 7H), 2.75 – 2.56 (m, 2H), 2.42 (s, 3H), 2.29 – 2.13 (m, 2H). [00183] 2-(2-(2-(2-(4-(2,6-dioxopiperidin-3-yl)phenoxy)ethoxy)ethoxy )ethoxy)ethyl 4- methylbenzenesulfonate (U). [00184] Under a nitrogen atmosphere, to a suspension of 2-(2-(2-(2-(4-(2,6- bis(benzyloxy)pyridin-3-yl)phenoxy)ethoxy)ethoxy)ethoxy)ethy l 4-methylbenzenesulfonate (S) (0.228 g, 0.319 mmol) in dry THF (2.3 ml) is added Pd/C (10% Pd, 0.034 g, 0.319 mmol) and then the reaction mixture is stirred under a hydrogen atmosphere at room temperature for 3 h. The reaction mixture is then filtered through a pad of celite and washed with EA. The filtrate is concentrated to dryness to afford the title compound as light-yellow oil (0.112 g, 100% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.93 (bs, 1H), 7.79 (d, J = 8.1 Hz, 2H), 7.33 (d, J = 8.1 Hz, 2H), 7.12 (d, J = 8.6 Hz, 2H), 6.91 (d, J = 8.7 Hz, 2H), 4.20 – 4.08 (m, 4H), 3.88 – 3.81 (m, 2H), 3.77 – 3.62 (m, 7H), 3.59 (s, 4H), 2.77 – 2.57 (m, 2H), 2.44 (s, 3H), 2.33 – 2.15 (m, 2H). [00185] 3-(4-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)phenyl)piperidine-2, 6-dione (V). [00186] Under a nitrogen atmosphere, to a solution of 2-(2-(2-(4-(2,6-dioxopiperidin-3- yl)phenoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (T) (0.051 g, 0.104 mmol) in dry DMF (2.0 mL) is added NaN 3 (0.014 g, 0.207 mmol) and the reaction is stirred at 65°C for 4 h. The reaction mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford the title compound as a clear yellow oil (36 mg, 95% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.94 (bs, 1H), 7.12 (d, J = 8.7 Hz, 2H), 6.92 (d, J = 8.5 Hz, 2H), 4.16 – 4.09 (m, 2H), 3.89 – 3.82 (m, 2H), 3.75 – 3.65 (m, 9H), 3.43 – 3.34 (m, 2H), 2.77 – 2.59 (m, 2H), 2.33 – 2.15 (m, 2H). [00187] 3-(4-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethoxy)phenyl)pip eridine-2,6-dione (W). [00188] Under a nitrogen atmosphere, to a solution of 2-(2-(2-(2-(4-(2,6-dioxopiperidin-3- yl)phenoxy)ethoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (U) (0.060 g, 0.112 mmol) in dry DMF (2.0 mL) is added NaN 3 (0.014 g, 0.224 mmol) and the reaction is stirred at 65°C for 4 h. The reaction mixture is poured into ice-water (20 mL) and then extracted with EA (x3). The reunited organic phases are washed with brine (x3), dried over Na 2 SO 4 , filtered and evaporated to dryness to afford the title compound as a clear yellow oil (40 mg, 89% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.94 (bs, 1H), 7.12 (d, J = 8.7 Hz, 2H), 6.92 (d, J = 8.5 Hz, 2H), 4.16 – 4.09 (m, 2H), 3.89 – 3.82 (m, 2H), 3.75 – 3.65 (m, 11H), 3.43 – 3.34 (m, 2H), 2.77 – 2.59 (m, 2H), 2.33 – 2.15 (m, 2H). [00189] Scheme 6. Synthesis of Examples 10 and 11. [00190] 4-(4-(2-(but-3-yn-1-yloxy)-3,4-difluorophenyl)thiazol-2-yl)m orpholine (X). [00191] Under nitrogen atmosphere, DIAD (0.108 mL, 0.553 mmol) is slowly added to a stirred ice-cooled solution of 2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenol (ARB-1) (0.150 g, 0.503 mmol), but-3-yn-1-ol (0.042 mL, 0.553 mmol), and PPh 3 (0.145 g, 0.553 mmol) in dry THF (3.0 mL). The solution is stirred at 0 °C for 30 min, then at rt for 18 h. The reaction mixture is evaporated to dryness and the crude residue is purified by automated flash chromatography on SiO 2 cartridge (PE/EA 90:10 to 80:20) to afford the title compound as a clear oil (102 mg, 50% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.81 (ddd, J = 8.9, 6.2, 2.4 Hz, 1H), 7.33 (s, 1H), 6.94 (td, J = 9.2, 7.5 Hz, 1H), 4.23 (td, J = 6.8, 1.1 Hz, 2H), 3.88 – 3.82 (m, 4H), 3.59 – 3.50 (m, 4H), 2.69 (td, J = 6.8, 2.7 Hz, 2H), 2.04 (t, J = 2.7 Hz, 1H). [00192] 3-(4-(2-(2-(2-(4-(2-(2,3-difluoro-6-(2-morpholinothiazol-4-y l)phenoxy)ethyl)-1H- 1,2,3-triazol-1-yl)ethoxy)ethoxy)ethoxy)phenyl)piperidine-2, 6-dione (Example 10). [00193] Under nitrogen atmosphere, to a solution of 4-(4-(2-(but-3-yn-1-yloxy)-3,4- difluorophenyl)thiazol-2-yl)morpholine (X) (0.038 g, 0.107 mmol) and 3-(4-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)phenyl)piperidine-2,6-dione (V) (0.039 g, 0.107 mmol) in a mixture of DMF:tBuOH:H 2 O 1:1:1 (3.0 mL) is added CuSO4 pentahydrate (0.013 g, 0.054 mmol) and sodium ascorbate (0.064 g, 0.321 mmol) and the reaction mixture is stirred at room temperature for 3 h. The reaction mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered and evaporated to dryness yielding a crude product which is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 97:3) to afford the title compound as a white solid (26 mg, 34% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.00 (bs, 1H), 7.79 (ddd, J = 8.4, 6.4, 2.0 Hz, 1H), 7.63 (s, 1H), 7.09 (d, J = 8.5 Hz, 2H), 7.03 (s, 1H), 6.97 – 6.83 (m, 3H), 4.51 (t, J = 5.0 Hz, 2H), 4.34 (t, J = 6.2 Hz, 2H), 4.11 – 4.04 (m, 2H), 3.91 – 3.74 (m, 8H), 3.73 – 3.58 (m, 5H), 3.53 – 3.46 (m, 4H), 3.19 (t, J = 6.2 Hz, 2H), 2.78 – 2.56 (m, 2H), 2.30 – 2.13 (m, 2H). HRMS (ESI) m/z [M+H]+ calcd for C 3 4H38F 2 N6O7S 713.25635, found 713.25648. [00194] 3-(4-(2-(2-(2-(2-(4-(2-(2,3-difluoro-6-(2-morpholinothiazol- 4-yl)phenoxy)ethyl)- 1H-1,2,3-triazol-1-yl)ethoxy)ethoxy)ethoxy)ethoxy)phenyl)pip eridine-2,6-dione (Example 11). [00195] Under nitrogen atmosphere, to a solution of 4-(4-(2-(but-3-yn-1-yloxy)-3,4- difluorophenyl)thiazol-2-yl)morpholine (X) (0.051 g, 0.148 mmol) and 3-(4-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethoxy)phenyl)piperidine-2,6-dione (W) (0.060 g, 0.148 mmol) in a mixture of DMF:tBuOH:H 2 O 1:1:1 (3.0 mL) is added CuSO4 pentahydrate (0.018 g, 0.074 mmol) and sodium ascorbate (0.088 g, 0.444 mmol) and the reaction mixture is stirred at room temperature for 3 h. The reaction mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered and evaporated to dryness yielding a crude product which is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 97:3) to afford the title compound as a white solid (46 mg, 42% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 7.93 (bs, 1H), 7.80 (t, J = 7.5 Hz, 1H), 7.63 (s, 1H), 7.10 (d, J = 8.5 Hz, 2H), 7.04 (s, 1H), 6.98 – 6.86 (m, 3H), 4.50 (t, J = 4.9 Hz, 2H), 4.36 (t, J = 6.5 Hz, 2H), 4.15 – 4.05 (m, 2H), 3.93 – 3.76 (m, 8H), 3.74 – 3.64 (m, 3H), 3.64 – 3.55 (m, 6H), 3.54 – 3.45 (m, 4H), 3.22 (t, J = 6.5 Hz, 2H), 2.77 – 2.58 (m, 2H), 2.31 – 2.14 (m, 2H). HRMS (ESI) m/z [M+Na]+ calcd for C 3 6H42F 2 N6O8S 779.26396, found 779.26505. [00196] Scheme 7. Synthesis of E3LB-3 and linker connection.

[00197] 2,6-bis(benzyloxy)-3-(4-nitrophenyl)pyridine (Y). [00198] Under nitrogen atmosphere, to a solution of 2,6-bis(benzyloxy)-3-bromopyridine (0.400 g, 1.080 mmol) in dioxane (12.0 mL) is added (4-nitrophenyl)boronic acid (0.361 g, 2.160 mmol), K 3 PO 4 (0.493 g, 2.322 mmol), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II ) complex with dichloromethane (0.088 g, 0.108 mmol) and then water (2.0 mL), and then the reaction mixture is stirred at 110° C for 16 h. After cooling, the mixture is filtered over a pad of celite and the filtrate is extracted with EA (x3). The reunited organic phases are then washed with brine, dried over Na 2 SO 4 , filtered and evaporated to dryness yielding a crude product which is purified by automated flash chromatography on SiO 2 cartridge (PE/EA, 9:1) to afford the title compound as a yellow oil (421 mg, 94% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.23 (d, J = 8.7 Hz, 2H), 7.73 (d, J = 8.5 Hz, 2H), 7.66 (d, J = 8.2 Hz, 1H), 7.49 – 7.28 (m, 10H), 6.53 (d, J = 8.0 Hz, 1H), 5.44 (s, 2H), 5.40 (s, 2H). [00199] 3-(4-aminophenyl)piperidine-2,6-dione (E3LB-3). [00200] Under nitrogen atmosphere, to a mixture of 2,6-bis(benzyloxy)-3-(4- nitrophenyl)pyridine (Y) (0.100 g, 0.242 mmol) in absolute EtOH (2.0 mL) is added Pd/C (10% Pd, 0.015 g, 0.145 mmol) and then the reaction mixture is stirred under a hydrogen atmosphere at room temperature for 5 h. The reaction mixture is then filtered through a pad of celite and washed with EtOH. The filtrate is concentrated to dryness to afford a yellow oil which solidified upon standing at low temperature (0.421 mg, 94% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 6.98 (d, J = 8.3 Hz, 2H), 6.67 (d, J = 8.3 Hz, 2H), 3.95 – 3.53 (m, 3H), 2.77 – 2.55 (m, 2H), 2.31 – 2.11 (m, 2H), 1.31 – 1.20 (m, 1H). [00201] tert-butyl (5-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-5-oxopentyl)ca rbamate (AA). [00202] Under nitrogen atmosphere, to a stirred solution of 5-((tert- butoxycarbonyl)amino)pentanoic acid (0.050 g, 0.230 mmol), 3-(4-aminophenyl)piperidine-2,6- dione (E3LB-3) (0.047 g, 0.230 mmol) and HATU (0.109 g, 0.287 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.120 mL, 0.690 mmol) and the reaction mixture is stirred at room temperature for 3 h. The mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 90:10) to afford the title compound as a white solid (56 mg, 60% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.85 (s, 1H), 7.53 (d, J = 8.5 Hz, 2H), 7.13 (d, J = 8.4 Hz, 2H), 6.80 (t, J = 5.5 Hz, 1H), 3.78 (dd, J = 11.3, 4.8 Hz, 1H), 2.98 – 2.86 (m, 2H), 2.70 – 2.60 (m, 1H), 2.48 – 2.42 (m, 1H), 2.35 – 2.24 (m, 2H), 2.22 – 2.09 (m, 1H), 2.05 – 1.97 (m, 1H), 1.61 – 1.50 (m, 2H), 1.46 – 1.28 (m, 11H). [00203] tert-butyl (7-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-7-oxoheptyl)ca rbamate (AB). [00204] Under nitrogen atmosphere, to a stirred solution of 7-((tert- butoxycarbonyl)amino)heptanoic acid (0.040 g, 0.161 mmol), 3-(4-aminophenyl)piperidine-2,6- dione (E3LB-3) (0.033 g, 0.161 mmol) and HATU (0.077 g, 0.201 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.084 mL, 0.483 mmol) and the reaction mixture is stirred at room temperature for 3 h. The mixture is poured into ice-water yielding a white precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 95:5) to afford the title compound as a white solid (44 mg, 64% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.84 (s, 1H), 7.53 (d, J = 8.4 Hz, 2H), 7.12 (d, J = 8.4 Hz, 2H), 6.81 – 6.69 (m, 1H), 3.78 (dd, J = 11.3, 4.8 Hz, 1H), 2.93 – 2.85 (m, 2H), 2.70 – 2.59 (m, 1H), 2.47 – 2.40 (m, 1H), 2.33 – 2.24 (m, 2H), 2.23 – 2.08 (m, 1H), 2.07 – 1.95 (m, 1H), 1.61 – 1.51 (m, 2H), 1.46 – 1.17 (m, 15H). [00205] tert-butyl (2-(2-(2-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-2-oxoeth oxy) ethoxy)ethyl)carbamate (AC). [00206] Under nitrogen atmosphere, to a stirred solution of 2,2-dimethyl-4-oxo-3,8,11-trioxa-5- azatridecan-13-oic acid (0.036 g, 0.137 mmol), 3-(4-aminophenyl)piperidine-2,6-dione (E3LB-3) (0.028 g, 0.137 mmol) and HATU (0.065 g, 0.171 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.095 mL, 0.548 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The crude is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 97:3) to afford the title compound as a colorless oil (36 mg, 58% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.63 (s, 1H), 8.38 (s, 1H), 7.58 (d, J = 8.5 Hz, 2H), 7.16 (d, J = 8.5 Hz, 2H), 4.93 (bs, 1H), 4.10 (s, 2H), 3.79 – 3.66 (m, 5H), 3.59 (t, J = 5.4 Hz, 2H), 3.38 – 3.29 (m, 2H), 2.76 – 2.55 (m, 2H), 2.31 – 2.12 (m, 2H), 1.41 (s, 9H). [00207] 5-amino-N-(4-(2,6-dioxopiperidin-3-yl)phenyl)pentanamide hydrochloride (AD). [00208] To the solution of tert-butyl (5-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-5- oxopentyl)carbamate (AA) (0.050 g, 0.123 mmol) in dry DCM (1.0 ml) is added 4.0 N HCl in dioxane (0.5 mL) and the reaction is stirred at room temperature for 3 h. The solvent is evaporated to dryness and the solid is triturated with DEE and collected by filtration, yielding the title compound as a white solid (0.034 g, 86% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.81 (s, 1H), 10.01 (s, 1H), 7.83 (bs, 3H), 7.55 (d, J = 8.3 Hz, 2H), 7.13 (d, J = 8.4 Hz, 2H), 3.79 (dd, J = 11.4, 4.9 Hz, 1H), 2.86 – 2.74 (m, 2H), 2.72 – 2.60 (m, 1H), 2.48 – 2.42 (m, 1H), 2.34 (t, J = 6.4 Hz, 2H), 2.23 – 2.09 (m, 1H), 2.07 – 1.96 (m, 1H), 1.70 – 1.52 (m, 4H). [00209] 7-amino-N-(4-(2,6-dioxopiperidin-3-yl)phenyl)heptanamide hydrochloride (AE). [00210] To the solution of tert-butyl (7-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-7- oxoheptyl)carbamate (AB) (0.037 g, 0.086 mmol) in dry DCM (1.0 ml) is added 4.0 N HCl in dioxane (0.40 mL) and the reaction is stirred at room temperature for 5 h. The solvent is evaporated to dryness and the solid is triturated with DEE and collected by filtration, yielding the title compound as a white solid (0.030 g, 97% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.81 (s, 1H), 9.93 (s, 1H), 7.78 (bs, 3H), 7.54 (d, J = 8.5 Hz, 2H), 7.13 (d, J = 8.5 Hz, 2H), 3.78 (dd, J = 11.4, 4.9 Hz, 1H), 2.83 – 2.71 (m, 2H), 2.71 – 2.60 (m, 1H), 2.48 – 2.43 (m, 1H), 2.31 (t, J = 7.3 Hz, 2H), 2.23 – 2.10 (m, 1H), 2.07 – 1.95 (m, 1H), 1.65 – 1.48 (m, 4H), 1.40 – 1.24 (m, 4H). [00211] 2-(2-(2-aminoethoxy)ethoxy)-N-(4-(2,6-dioxopiperidin-3-yl)ph enyl)acetamide hydrochloride (AF). [00212] To a solution of tert-butyl (2-(2-(2-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-2- oxoethoxy)ethoxy)ethyl)carbamate (AC) (0.034 g, 0.076 mmol) in dry DCM (1.0 ml) is added 4.0 N HCl in dioxane (0.40 mL) and the reaction is stirred at room temperature for 2 h. The solvent is evaporated to dryness and the solid is triturated with DEE and collected by filtration, yielding the title compound as a white solid (0.029 g, 100% yield). HRMS (ESI) m/z [M+H]+ calcd for C 17 H 23 N 3 O 5 350.17105, found 350.17186. [00213] Scheme 8. Synthesis of Example 12. [00214] 9-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)nonanoic acid (AG). [00215] Under nitrogen atmosphere, to a suspension of NaH 60% (0.024 g, 0.672 mmol) in dry DMF (1.0 mL) at 0° C is added a solution of 2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenol (ARB-1) (0.100 g, 0.336 mmol) in dry DMF (1.0 mL). After 20 min at 0°C, a solution of 9- bromononanoic acid (0.119 g, 0.503 mmol) in dry DMF (1.0 mL) is added and the reaction mixture is stirred at 60° C for 24 h. The reaction mixture is quenched with EA and water and then extracted with EA (x3). The reunited organic layers are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness yielding a crude light-yellow oil. The crude oil is triturated with DEE and collected by filtration to afford the title compound as a white solid (0.084 g, 55% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.96 (bs, 1H), 7.87 – 7.73 (m, 1H), 7.33 (s, 1H), 7.25 – 7.11 (m, 1H), 4.15 – 4.02 (m, 2H), 3.82 – 3.64 (m, 4H), 3.48 – 3.37 (m, 4H), 2.23 – 2.12 (m, 2H), 1.81 – 1.67 (m, 2H), 1.51 – 1.43 (m, 2H), 1.42 – 1.34 (m, 2H), 1.29 – 1.20 (m, 6H). [00216] 9-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)-N-(4-(2 ,6-dioxopiperidin-3- yl)phenyl)nonanamide (Example 12). [00217] Under nitrogen atmosphere, to a stirred solution of 9-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)nonanoic acid (AG) (0.033 g, 0.073 mmol), 3-(4- aminophenyl)piperidine-2,6-dione (E3LB-3) (0.015 g, 0.073 mmol) and HATU (0.035 g, 0.091 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.051 mL, 0.292 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 97:3) to afford the title compound as a colorless oil (19.5 mg, 41% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.84 (s, 1H), 7.79 (t, J = 7.0 Hz, 1H), 7.53 (d, J = 8.3 Hz, 2H), 7.33 (s, 1H), 7.24 – 7.09 (m, 3H), 4.07 (t, J = 6.1 Hz, 2H), 3.82 – 3.68 (m, 5H), 3.48 – 3.39 (m, 4H), 2.72 – 2.59 (m, 1H), 2.47 – 2.42 (m, 1H), 2.33 – 2.23 (m, 2H), 2.22 – 2.08 (m, 1H), 2.05 – 1.94 (m, 1H), 1.78 – 1.69 (m, 2H), 1.61 – 1.53 (m, 2H), 1.43 – 1.34 (m, 2H), 1.32 – 1.25 (m, 6H). 13 C NMR (101 MHz, DMSO-d 6 ) δ 174.80, 173.88, 171.62, 170.01, 150.04 (dd, J = 247.5, 11.5 Hz), 145.77 (dd, J = 9.1, 1.3 Hz), 145.47-145.35 (m), 144.41 (dd, J = 245.0, 13.4 Hz), 138.56, 134.01, 129.18 (2C), 125.63 (d, J = 2.6 Hz), 124.62- 124.39 (m), 119.46 (2C), 111.83 (d, J = 17.0 Hz), 107.31, 74.40 (d, J = 5.2 Hz), 65.87 (2C), 48.60 (2C), 47.21, 36.83, 31.78, 30.01, 29.20, 29.06, 29.03, 26.36, 25.72, 25.60. HRMS (ESI) m/z [M+H]+ calcd for C 33 H 38 F 2 N 4 O 5 S 641.26037, found 641.26016. [00218] Scheme 9. Synthesis of Examples 4, 13, and 14. [00219] 9-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)-N-(5-(( 4-(2,6- dioxopiperidin-3-yl)phenyl)amino)-5-oxopentyl)nonanamide (Example 4). [00220] Under nitrogen atmosphere, to a stirred solution of 9-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)nonanoic acid (AG) (0.040 g, 0.088 mmol), 5-amino-N-(4-(2,6- dioxopiperidin-3-yl)phenyl)pentanamide hydrochloride (AD) (0.030 g, 0.088 mmol) and HATU (0.042 g, 0.110 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.061 mL, 0.352 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water yielding a white precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1) to afford the title compound as a white solid (11 mg, 17% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.85 (s, 1H), 7.85 – 7.71 (m, 2H), 7.52 (d, J = 8.3 Hz, 2H), 7.32 (s, 1H), 7.19 (dd, J = 17.2, 9.3 Hz, 1H), 7.12 (d, J = 8.4 Hz, 2H), 4.06 (t, J = 6.3 Hz, 2H), 3.81 – 3.68 (m, 5H), 3.48 – 3.38 (m, 4H), 3.09 – 2.99 (m, 2H), 2.70 – 2.58 (m, 1H), 2.47 – 2.42 (m, 1H), 2.29 (t, J = 7.2 Hz, 2H), 2.21 – 2.08 (m, 1H), 2.07 – 1.95 (m, 3H), 1.76 – 1.68 (m, 2H), 1.61 – 1.52 (m, 2H), 1.51 – 1.43 (m, 2H), 1.43 – 1.33 (m, 4H), 1.30 – 1.18 (m, 6H). HRMS (ESI) m/z [M+H]+ calcd for C 3 8H47F 2 N 5 O 6 S 740.32879, found 740.32893. [00221] 9-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)-N-(7-(( 4-(2,6- dioxopiperidin-3-yl)phenyl)amino)-7-oxoheptyl)nonanamide (Example 13). [00222] Under nitrogen atmosphere, to a stirred solution of 9-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)nonanoic acid (AG) (0.032 g, 0.071 mmol), 7-amino-N-(4-(2,6- dioxopiperidin-3-yl)phenyl)heptanamide hydrochloride (AE) (0.026 g, 0.071 mmol) and HATU (0.034 g, 0.089 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.050 mL, 0.284 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water yielding a white precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 95:5) to afford the title compound as a white solid (15 mg, 27% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.84 (s, 1H), 7.79 (ddd, J = 8.5, 6.4, 2.0 Hz, 1H), 7.70 (t, J = 5.1 Hz, 1H), 7.52 (d, J = 8.4 Hz, 2H), 7.32 (s, 1H), 7.19 (dd, J = 17.2, 9.3 Hz, 1H), 7.12 (d, J = 8.5 Hz, 2H), 4.06 (t, J = 6.4 Hz, 2H), 3.83 – 3.69 (m, 5H), 3.48 – 3.39 (m, 4H), 3.05 – 2.96 (m, 2H), 2.71 – 2.58 (m, 1H), 2.47 – 2.42 (m, 1H), 2.27 (t, J = 7.3 Hz, 2H), 2.22 – 2.08 (m, 1H), 2.07 – 1.96 (m, 3H), 1.79 – 1.67 (m, 2H), 1.63 – 1.52 (m, 2H), 1.52 – 1.43 (m, 2H), 1.41 – 1.32 (m, 4H), 1.31 – 1.16 (m, 10H). HRMS (ESI) m/z [M+H]+ calcd for C 40 H 51 F 2 N 5 O 6 S 768.36009, found 768.36037. [00223] 9-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)-N-(2-(2 -(2-((4-(2,6- dioxopiperidin-3-yl)phenyl)amino)-2-oxoethoxy)ethoxy)ethyl)n onanamide (Example 14). [00224] Under nitrogen atmosphere, to a stirred solution of 9-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)nonanoic acid (AG) (0.031 g, 0.068 mmol), 2-(2-(2- aminoethoxy)ethoxy)-N-(4-(2,6-dioxopiperidin-3-yl)phenyl)ace tamide hydrochloride (AF) (0.026 g, 0.068 mmol) and HATU (0.032 g, 0.085 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.047 mL, 0.272 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 97:3), followed by preparative TLC eluting with DCM:Acetone:MeOH (88:10:2), to afford the title compound as a white solid (10.6 mg, 20% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.61 (s, 1H), 8.00 (s, 1H), 7.85 – 7.77 (m, 1H), 7.59 (d, J = 8.5 Hz, 2H), 7.24 (s, 1H), 7.18 (d, J = 8.4 Hz, 2H), 6.90 (dd, J = 16.7, 9.1 Hz, 1H), 5.85 – 5.76 (m, 1H), 4.12 (s, 2H), 4.07 (t, J = 6.5 Hz, 2H), 3.88 – 3.80 (m, 4H), 3.81 – 3.67 (m, 5H), 3.61 (t, J = 5.1 Hz, 2H), 3.55 – 3.44 (m, 6H), 2.78 – 2.60 (m, 2H), 2.31 – 2.19 (m, 2H), 2.12 (t, J = 7.6 Hz, 2H), 1.83 – 1.73 (m, 2H), 1.59 – 1.54 (m, 2H), 1.46 – 1.36 (m, 2H), 1.35 – 1.24 (m, 6H). HRMS (ESI) m/z [M+H]+ calcd for C 39 H 49 F 2 N 5 O 8 S 786.33427, found 786.33456. [00225] Scheme 10. Synthesis of E3LB-4 and linker connection. [00226] 3-((4-(tert-butoxycarbonyl)phenyl)amino)propanoic acid (AH). [00227] To a solution of tert-butyl 4-aminobenzoate (0.800 g, 4.139 mmol) in toluene (8.0 mL) is added acrylic acid (1.044 g, 14.489 mmol) and the reaction mixture is stirred at 100°C for 18 h. After cooling, a mixture of PE:EA (20:1) (32.0 mL) is added, yielding a precipitate which is collected by filtration and dried to afford the title compound as a white solid (0.826 g, 75% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.28 (bs, 1H), 7.62 (d, J = 8.6 Hz, 2H), 6.56 (d, J = 8.6 Hz, 2H), 6.45 (bs, 1H), 3.32 – 3.25 (m, 4H), 1.49 (s, 9H). [00228] 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid (E3LB-4). [00229] A mixture of 3-((4-(tert-butoxycarbonyl)phenyl)amino)propanoic acid (AH) (0.826 g, 3.113) and urea (0.467 g, 7.783 mmol) in acetic acid (7.6 mL) is stirred at 120°C for 18 h. After cooling at 0° C, EA (10 mL) is added yielding a precipitate which is collected by filtration and dried to afford the title compound as a white solid (0.230 g, 31% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.87 (bs, 1H), 10.49 (s, 1H), 7.94 (d, J = 8.5 Hz, 2H), 7.46 (d, J = 8.3 Hz, 2H), 3.86 (t, J = 6.3 Hz, 2H), 2.72 (t, J = 6.4 Hz, 2H). 13 C NMR (101 MHz, DMSO-d 6 ) δ 170.95, 167.30, 152.41, 146.32, 130.15 (2C), 127.88, 124.80 (2C), 44.56, 31.41. [00230] tert-butyl (4-(4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzamido)butyl ) carbamate (AI). [00231] Under nitrogen atmosphere, to a stirred solution of 4-(2,4-dioxotetrahydropyrimidin- 1(2H)-yl)benzoic acid (E3LB-4) (0.070 g, 0.299 mmol), tert-butyl (4-aminobutyl)carbamate (0.056 g, 0.299 mmol) and HATU (0.142 g, 0.374 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.208 mL, 1.196 mmol) and the reaction mixture is stirred at room temperature for 4 h. The mixture is poured into ice-water and then extracted with EA (x3). The reunited organic phases are washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 95:5) to afford the title compound as a white solid (79 mg, 65% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.44 (s, 1H), 8.42 (t, J = 5.5 Hz, 1H), 7.84 (d, J = 8.4 Hz, 2H), 7.41 (d, J = 8.1 Hz, 2H), 6.83 – 6.76 (m, 1H), 3.84 (t, J = 6.7 Hz, 2H), 3.28 – 3.19 (m, 2H), 2.98 – 2.87 (m, 2H), 2.71 (t, J = 6.4 Hz, 2H), 1.55 – 1.30 (m, 13H). [00232] N-(4-aminobutyl)-4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)be nzamide hydrochloride (AJ). [00233] To a solution of tert-butyl (4-(4-(2,4-dioxotetrahydropyrimidin-1(2H)- yl)benzamido)butyl)carbamate (AI) (0.070 g, 0.173 mmol) in dry DCM (2.0 ml) is added 4.0 N HCl in dioxane (0.70 mL) and the reaction is stirred at room temperature for 3 h. The solvent is evaporated to dryness and the solid is triturated with DEE and collected by filtration, yielding a the title compound as a white solid (0.058 g, 98% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.45 (s, 1H), 8.56 (bs, 1H), 7.97 – 7.77 (m, 5H), 7.42 (d, J = 8.7 Hz, 2H), 3.84 (t, J = 6.6 Hz, 2H), 3.33 – 3.23 (m, 2H), 2.84 – 2.75 (m, 2H), 2.72 (t, J = 6.7 Hz, 2H), 1.58 (s, 4H). [00234] Scheme 11. Synthesis of Example 15. [00235] N-(4-(9-(2,3-difluoro-6-(2-morpholinothiazol-4-yl)phenoxy)no nanamido)butyl)-4- (2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzamide (Example 15). [00236] Under nitrogen atmosphere, to a stirred solution of 9-(2,3-difluoro-6-(2- morpholinothiazol-4-yl)phenoxy)nonanoic acid (AG) (0.064 g, 0.141 mmol), N-(4-aminobutyl)- 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzamide hydrochloride (AJ) (0.048 g, 0.141 mmol) and HATU (0.067 g, 0.176 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.098 mL, 0.564 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water yielding a precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 99:1 to 92:8) to afford the title compound as a white solid (39 mg, 37% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.44 (s, 1H), 8.43 (t, J = 5.5 Hz, 1H), 7.84 (d, J = 8.6 Hz, 2H), 7.82 – 7.77 (m, 1H), 7.74 (t, J = 5.4 Hz, 1H), 7.40 (d, J = 8.6 Hz, 2H), 7.32 (s, 1H), 7.19 (dd, J = 17.2, 9.3 Hz, 1H), 4.06 (t, J = 6.4 Hz, 2H), 3.83 (t, J = 6.6 Hz, 2H), 3.78 – 3.68 (m, 4H), 3.47 – 3.39 (m, 4H), 3.25 (dd, J = 12.3, 6.3 Hz, 2H), 3.05 (dd, J = 12.4, 6.4 Hz, 2H), 2.71 (t, J = 6.6 Hz, 2H), 2.03 (t, J = 7.4 Hz, 2H), 1.78 – 1.67 (m, 2H), 1.55 – 1.32 (m, 8H), 1.29 – 1.18 (m, 6H). 13 C NMR (101 MHz, DMSO-d 6 ) δ 172.38, 170.97, 170.03, 165.91, 152.46, 150.05 (dd, J = 247.5, 11.4 Hz), 145.83, 145.48 – 145.35 (m), 144.75, 144.43 (dd, J = 245.7, 14.3 Hz), 131.98, 127.94 (2C), 125.64 (d, J = 2.4 Hz), 124.77 (2C), 124.64-124.42 (m), 111.85 (d, J = 17.0 Hz), 107.32, 74.41 (d, J = 5.2 Hz), 65.88 (2C), 48.61 (2C), 44.68, 38.62, 35.91, 31.45, 30.01, 29.19, 29.10, 29.05, 27.25, 27.14, 25.79, 25.72. HRMS (ESI) m/z [M+H]+ calcd for C 3 7H46F 2 N6O 6 S 741.32404, found 741.32545. [00237] Scheme 12. Synthesis of ARB-2 and ARB-3 moieties. [00238] 4-(4-Bromothiazol-2-yl)morpholine (AK). [00239] A solution of 2,4-dibromothiazole (0.250 g, 1.029 mmol) in morpholine (0.990 mL, 11.320 mmol) is warmed at 50°C in a sealed tube. After 16 h, the mixture is cooled to room temperature and the white inorganic precipitate is collected by filtration. The organic phase is diluted with 50 mL of water and extracted with EA (20 mL x 3), the reunited organic phases are dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford the title compound (0.215 g, 87% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ): δ 6.92 (s, 1H), 3.73 – 3.64 (m, 4H), 3.42 – 3.33 (m, 4H). 13 C NMR (101 MHz, DMSO-d 6 ) δ 171.18, 121.01, 105.39, 65.71(2C), 48.07 (2C). HRMS (ESI) m/z [M+H]+ calcd for C 7 H 9 BrN 2 OS 248.96917, found 248.96923. [00240] 4-(2-Morpholinothiazol-4-yl)phenol (ARB-2). [00241] The titled compound can be prepared according to the process described by Bhumireddy A, et al., Bioorg. Med. Chem. Lett.2022, 128448. [00242] Under nitrogen atmosphere, to a solution of compound AK (0.204 g, 0.819 mmol) in dioxane (4.08 mL) and H 2 O (1.428 mL) are added (4-hydroxyphenyl) boronic acid (0.169 g, 1.228 mmol), and sodium carbonate (0.260 g, 2.457 mmol). The mixture is degassed with nitrogen purging for 20 min. Then, tetrakis triphenylphosphine palladium (0) (0.095 g, 0.082 mmol) is added and the reaction is heated at 100°C for 16 h. After cooling, the reaction mixture is concentrated under reduced pressure and the residue is diluted with EA (30 mL), washed with water (40 mL x 2), dried over Na 2 SO 4 , and concentrated under reduced pressure. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 95:5) to give the title compound as a white solid (0.112 g, 52% yield). 1 H NMR (400 MHz, DMSO-d 6 ): δ 9.52 (s, 1H), 7.67 (d, J = 8.7 Hz, 2H), 7.02 (s, 1H), 6.76 (d, J = 8.7 Hz, 2H), 3.77 – 3.67 (m, 4H), 3.46 – 3.38 (m, 4H). [00243] 3-(2-Morpholinothiazol-4-yl)phenol (ARB-3). [00244] Under nitrogen atmosphere, to a solution of compound AK (0.204 g, 0.819 mmol) in 1,4-dioxane (4.08 mL) and H 2 O (1.428 mL) are added (3-hydroxyphenyl) boronic acid (0.169 g, 1.228 mmol) and sodium carbonate (0.260 g, 2.457 mmol). The mixture is degassed with nitrogen purging for 20 min. Then, tetrakis triphenylphosphine palladium (0) (0.095 g, 0.082 mmol) is added and the reaction is heated at 100°C for 16 h. After cooling, the reaction mixture is concentrated under reduced pressure and the residue is diluted with EA (30 mL), washed with water (40 mL x 2), dried over Na 2 SO 4 and concentrated under reduced pressure. The crude product is purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 95:5) to give the title compound as a white solid (0.107 g, 50% yield). 1 H NMR (400 MHz, DMSO-d 6 ): δ 9.39 (s, 1H), 7.31 – 7.23 (m, 2H), 7.21 (s, 1H), 7.16 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 7.3 Hz, 1H), 3.78 – 3.68 (m, 4H), 3.47 – 3.38 (m, 4H). 13 C NMR (101 MHz, DMSO-d 6 ): δ 171.01, 157.93, 151.14, 136.36, 129.90, 116.99, 115.06, 113.17, 103.13, 65.88 (2C), 48.64 (2C). [00245] Scheme 13. Synthesis of Example 7.

[00246] 9-(4-(2-Morpholinothiazol-4-yl)phenoxy)nonanoic acid (AL). [00247] Under nitrogen atmosphere, to a suspension of NaH 60% (0.025 g, 0.627 mmol) in dry DMF (1.0 mL) at 0° C is added a solution of 4-(2-morpholinothiazol-4-yl)phenol (ARB-2) (0.055 g, 0.209 mmol) in dry DMF (1.0 mL). After 20 min at 0°C, a solution of 9-bromononanoic acid (0.075 g, 0.314 mmol) in dry DMF (1.0 mL) is added and the reaction mixture is stirred at room temperature for 18 h. The reaction mixture is quenched with EA and water and then extracted with EA (x3). The reunited organic layers are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness yielding a light-yellow oil which is triturated with DEE and collected by filtration to afford the title compound as a white solid (0.052 g, 60% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.95 (s, 1H), 7.76 (d, J = 8.7 Hz, 2H), 7.12 (s, 1H), 6.92 (d, J = 8.6 Hz, 2H), 3.97 (t, J = 6.3 Hz, 2H), 3.78 – 3.66 (m, 4H), 3.47 – 3.37 (m, 4H), 2.19 (t, J = 7.2 Hz, 2H), 1.75 – 1.66 (m, 2H), 1.55 – 1.46 (m, 2H), 1.45 – 1.36 (m, 2H), 1.34 – 1.24 (m, 6H). [00248] N-(5-((4-(2,6-Dioxopiperidin-3-yl)phenyl)amino)-5-oxopentyl) -9-(4- (2morpholinothiazol-4-yl)phenoxy)nonanamide (Example 7). [00249] Under nitrogen atmosphere, to a stirred solution of 9-(4-(2-morpholinothiazol-4- yl)phenoxy)nonanoic acid (AL) (0.047 g, 0.112 mmol), 5-amino-N-(4-(2,6-dioxopiperidin-3- yl)phenyl)pentanamide hydrochloride (AD) (0.038 g, 0.112 mmol), and HATU (0.053 g, 0.140 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.078 mL, 0.448 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water yielding a white precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 95:5) to afford the title compound as a white solid (37 mg, 47% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.85 (s, 1H), 7.76 (d, J = 8.7 Hz, 3H), 7.53 (d, J = 8.5 Hz, 2H), 7.15 – 7.09 (m, 3H), 6.92 (d, J = 8.7 Hz, 2H), 3.95 (t, J = 6.5 Hz, 2H), 3.81 – 3.67 (m, 5H), 3.48 – 3.37 (m, 4H), 3.10 – 3.00 (m, 2H), 2.70 – 2.57 (m, 1H), 2.49 – 2.41 (m, 1H), 2.29 (t, J = 7.3 Hz, 2H), 2.21 – 2.09 (m, 1H), 2.07 – 1.95 (m, 3H), 1.75 – 1.64 (m, 2H), 1.62 – 1.35 (m, 8H), 1.33 – 1.20 (m, 6H). 13 C NMR (101 MHz, DMSO-d 6 ): δ 174.82, 173.89, 172.40, 171.50, 171.11, 158.72, 150.92, 138.55, 134.04, 129.20 (2C), 127.82, 127.49 (2C), 119.45 (2C), 114.81(2C), 101.04, 67.90, 65.90 (2C), 48.65 (2C), 47.22, 38.57, 36.47, 35.89, 31.79, 29.29, 29.22, 29.18, 29.17, 29.09, 26.36, 25.98, 25.79, 23.10. HRMS (ESI) m/z [M+H] + calcd for C 38 H 49 N 5 O 6 S 704.34763, found 704.3507. [00250] Scheme 14. Synthesis of Example 6. [00251] 9-(3-(2-Morpholinothiazol-4-yl)phenoxy)nonanoic acid (AM). [00252] Under nitrogen atmosphere, to a suspension of NaH 60% (0.045 g, 1.143 mmol) in dry DMF (1.0 mL) at 0° C is added a solution of 3-(2-morpholinothiazol-4-yl)phenol (ARB-3) (0.100 g, 0.381 mmol) in dry DMF (1.0 mL). After 20 min at 0°C, a solution of 9-bromononanoic acid (0.136 g, 0.572 mmol) in dry DMF (1.0 mL) is added and the reaction mixture is stirred at room temperature for 18 h. The reaction mixture is quenched with EA and water and then extracted with EA (x3). The reunited organic layers are washed with water (x3), brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness yielding a light-yellow oil which is triturated with DEE and collected by filtration to afford the title compound as a white solid (0.098 g, 62% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.96 (s, 1H), 7.47 – 7.37 (m, 2H), 7.33 (s, 1H), 7.27 (t, J = 7.9 Hz, 1H), 6.88 – 6.80 (m, 1H), 3.98 (t, J = 6.3 Hz, 2H), 3.78 – 3.68 (m, 4H), 3.49 – 3.38 (m, 4H), 2.19 (t, J = 7.4 Hz, 2H), 1.76 – 1.67 (m, 2H), 1.54 – 1.46 (m, 2H), 1.45 – 1.36 (m, 2H), 1.35 – 1.24 (m, 6H). [00253] N-(5-((4-(2,6-dioxopiperidin-3-yl)phenyl)amino)-5-oxopentyl) -9-(3- (2morpholinothiazol-4-yl)phenoxy)nonanamide (Example 6). [00254] Under nitrogen atmosphere, to a stirred solution of 9-(3-(2-morpholinothiazol-4- yl)phenoxy)nonanoic acid (AM) (0.050 g, 0.119 mmol), 5-amino-N-(4-(2,6-dioxopiperidin-3- yl)phenyl)pentanamide hydrochloride (AD) (0.041 g, 0.119 mmol), and HATU (0.057 g, 0.149 mmol) in dry DMF (2.0 mL) at 0° C is added DIPEA (0.083 mL, 0.476 mmol) and the reaction mixture is stirred at room temperature for 2 h. The mixture is poured into ice-water yielding a white precipitate which is collected by filtration and then purified by automated flash chromatography on SiO 2 cartridge (DCM/MeOH, 95:5) to afford the title compound as a white solid (52 mg, 62% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.80 (s, 1H), 9.85 (s, 1H), 7.75 (t, J = 6.2 Hz, 1H), 7.53 (d, J = 8.5 Hz, 2H), 7.45 – 7.37 (m, 2H), 7.33 (s, 1H), 7.27 (t, J = 7.9 Hz, 1H), 7.12 (d, J = 8.5 Hz, 2H), 6.87 – 6.81 (m, 1H), 3.97 (t, J = 6.3 Hz, 2H), 3.81 – 3.70 (m, 5H), 3.49 – 3.38 (m, 4H), 3.10 – 3.00 (m, 2H), 2.71 – 2.58 (m, 1H), 2.49 – 2.42 (m, 1H), 2.29 (t, J = 7.2 Hz, 2H), 2.22 – 2.09 (m, 1H), 2.07 – 1.95 (m, 3H), 1.74 – 1.65 (m, 2H), 1.61 – 1.37 (m, 8H), 1.35 – 1.20 (m, 6H). 13 C NMR (101 MHz, DMSO-d 6 ): δ 174.80, 173.87, 172.37, 171.48, 171.03, 159.36, 150.86, 138.54, 136.40, 134.02, 130.01, 129.19 (2C), 119.44 (2C), 118.47, 113.89, 112.32, 103.56, 67.82, 65.89 (2C), 48.63 (2C), 47.21, 38.57, 36.46, 35.89, 31.78, 29.28, 29.23, 29.20 (2C), 29.10, 26.36, 26.02, 25.79, 23.10. HRMS (ESI) m/z [M+Na] + calcd for C 38 H 49 N 5 O 6 S 726.32958, found 726.33266. [00255] Scheme 16. Synthesis of ARB-4A, ARB-4B, and ARB-4C [00256] Using the procedures described in the previous examples, ARB-4A, ARB-4B, and ARB-4C may be coupled with linkers and E3LB moieties as described herein to provide compounds within the scope of the present disclosure. [00257] Scheme 17. Synthesis of ARB-5

[00258] Using the procedures described in the previous examples, ARB-5A, ARB-5B, and ARB-5C may be coupled with linkers and E3LB moieties as described herein to provide compounds within the scope of the present disclosure. [00259] Experimental Example 1: 22Rv1 Cell Proliferation Assays [00260] The human prostate cancer cell line, 22Rv1 has been reported to express a high level of AR-V7. According to one procedure, 22Rv1 is seeded at 50,000 cells/well on a 24-well plate in quadruplicate and treated with test compounds in concentrations ranging up to 20 µM for four days. Standard culture media is RPMI-1640 supplemented with 10% fetal bovine serum. The test compound initially is dissolved in DMSO at 50 mM. This stock solution is then diluted as needed for the indicated concentrations. At the end of the four-day period, cells are harvested using 1% trypsin and counted using an automated cell counter. [00261] Experimental Example 2: Immunoblot [00262] Immunoblot may be carried out to determine the effect of the test compound on AR- V7.22Rv1 is plated at 200,000 cell/well on a 6-well plate and cultured as described with 10 µM test compound. After four days of treatment, cells are harvested using a cell scraper and lysed in a standard fashion using SDS. After removing debris via centrifuge, 30 µg of protein is loaded onto SDS-PAGE gel. After electrophoresis, protein is transferred to a nylon membrane and ECL is carried out using primary antibody against AR-V7 (Thermofisher Scientific, cat# NC0752138). Protein bands are visualized using the commercially available Enhanced Chemiluminescence (ECL) kit (Thermofisher). [00263] Experimental Example 3 [00264] Compounds according to the present disclosure (“Test Compound”) may be tested for in vitro and/or in vivo efficacy according to one or more of the procedures outlined below. [00265] Cell Culture. Human CaP cell lines, LNCaP, 22Rv1, VCaP, PC 3 , and DU145 are obtained from the American Type Culture Collection (ATCC) and maintained in the standard culture media: RPMI-1640 supplemented with 10% fetal bovine serum (FBS). LNCaP, 22Rv1, and VCaP are androgen-responsive cell lines, while PC 3 and DU145 are not. To establish SAT resistant CaP cell lines, LNCaP, 22Rv1, and VCaP are treated continuously with 10-50 μM abiraterone, apalutamide, darolutamide, or enzalutamide. After 3-6 months, stable cell lines are established and designated as LNCaP-Abi R , LNCaP-Apa R , LNCaP-Daro R , LNCaP-Enz R , VCaP- Abi R , VCaP-Apa R , VCaP-Daro R , VCaP-Enz R , 22Rv1-Abi R , 22Rv1-Apa R , 22Rv1-Daro R , and 22Rv1-Enz R . Unless otherwise specified, the standard culture media for these SAT-resistant cell lines included 10 μM of their respective SAT. For the proteasome inhibitor study, the inhibitors MG132 and Epoxomicin are used. The E3 ligase inhibitors Heclin, Nutlin 3a, Thalidomide, and VH298 are used. Cell lines obtained from ATCC are confirmed by checking their morphology using optical microscopy, establishing baselines for cell proliferation, verifying species of origin using isoenzymology, and characterizing the cell’s DNA fingerprint using short tandem repeat (STR) profiling. Mycoplasma contamination is also assessed using a PCR based detection system. [00266] Apoptosis Assay. An apoptosis assay is carried out using the Thermo Fisher ApoDETECT Annexin V-FITC kit following the protocol recommended by the vendor. Briefly, after treatment with 1 μM of Test Compound for 3 to 24 hours, cells are fixed with 80% ethanol and washed with PBS three times. Then, fixed cells are incubated with Annexin V-FITC in PBS solution for 30 minutes at room temperature. After washing three times with PBS, cells are treated with 300 nM DAPI in PBS for 5 minutes at room temperature. Finally, after washing three times with PBS, mounting solution is added and the cells are visualized using immunofluorescence microscopy. Next, a TUNEL assay is performed using Promega DeadEnd Fluorometric TUNEL system. After treatment with Test Compound and fixation as described above in the Annexin-V experiment, 100 μl of equilibration buffer is incubated for 10 min. Then, 50 μl of TdT reaction mix is added and incubated for 60 min at 37 ^C in a humidified chamber. Finally, stop solution is added and samples are mounted on slides using mounting medium. To assess non-specific cytotoxicity, an LDH assay kit is used. [00267] Transient Transfections. One ^g of a plasmid containing cDNA of AR-V7 or AR-FL is transfected into indicated the CaP cell lines on 6-well plates. Three ^l of lipofectamine 3000 is used for each transfection. [00268] Immunoblot Analysis. CaP cells are collected and lysed with the lysis buffer (20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM Na 2 EDTA, 1 mM EGTA, 1% Triton, 2.5 mM sodium pyrophosphate, 1 mM beta-glycerophosphate, 1 mM Na 3 VO 4 , and 1 µg/ml leupeptin) containing 1 mM phenylmethylsulfonyl fluoride (PMSF). Cell lysates are then centrifuged and protein in the supernatant is quantified. After separating 25-50μg of protein using SDS-PAGE, samples are incubated with AR-V7, GR, PR, ER α, AR-FL, ubiquitin, or β-actin antibodies. For AR-V7, AR- FL, PR, GR, and ER α immunoblots, primary antibody is diluted 1:1000 in 5% skim milk. For the β-actin immunoblot, 1:10000 diluted primary antibody is used. All membranes are incubated overnight at 4 C. Following the incubation with appropriate secondary antibody, immunoblots are analyzed using SuperSignal West Femto Maximum Sensitivity Substrate (ThermoFisher). [00269] In Vivo Study. To explore the therapeutic potential of test compounds, 22Rv1, 22Rv1- Enz R , VCaP, and VCaP-Enz R are injected into nu/nu immunodeficient mice. When the resulting tumors reached an average size of 3 mm in diameter, all animals are surgically castrated via bilateral orchiectomy and divided into four groups of five mice each. For anesthesia, 3% isoflurane gas inhalation is used. Tumor size was measured using calipers and tumor volume was calculated using the formula: tumor volume= length x width 2 /0.361. [00270] Mice are then treated daily with test compound with or without enzalutamide via the indicated route (intratumoral, intraperitoneal, or oral) for five to six weeks. At the end of the study, all animals are sacrificed and tumors are harvested and analyzed. Statistical significance is calculated using the Student’s t-test for paired comparisons of experimental groups and, where appropriate, by Wilcoxon rank sum test, and by 2-way ANOVA. In vitro experiments are repeated a minimum of three times. [00271] Experimental Example 4: 22Rv1-Enz R Cell Proliferation Assays [00272] Selected compounds may be tested in the human prostate cancer cell line, 22Rv1-Enz R , as described in Geun Taek Lee et al., Molecular Cancer Therapeutics, 20:490-9 (2021). These procedures are similar to that described in Experimental Example 1, above, except that the assay conditions are a 6-day culture with cell media changed every 24 hours. [00273] The results demonstrate that the test compounds inhibited proliferation in a concentration dependent manner. In the following Table (half maximal growth inhibition constant, GIC 50 ): (*) – GIC 50 2 – 10 μM; (++) – GIC 50 1.0 – 2.0 μM; (+++) – GIC 50 < 1.0 μM. [00274] The Examples provided herein are exemplary only and are not intended to be limiting in any way to the various aspects and embodiments of the invention described herein.