ANDROGEN RECEPTOR MODULATORS AND METHODS FOR THEIR USE

Abstract
The present invention relates to compounds of formula (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or any subgenera thereof, or a pharmaceutically acceptable salt, tautomer or stereoisomer. The compounds of the present disclosure are useful in modulating androgen receptor activity and for treating cancer, including prostate cancer.
Description
FIELD OF THE INVENTION

The present disclosure generally relates to tricyclic compounds and their use for treatment of various indications. In particular the disclosure relates to tricyclic compounds and their use for treatment of various cancers, for example prostate cancer, including but not limited to, primary/localized prostate cancer (newly diagnosed), locally advanced prostate cancer, recurrent prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic prostate cancer, metastatic castration-resistant prostate cancer (CRPC), and hormone-sensitive prostate cancer. This invention also relates to tricyclic compounds and their use for modulating androgen receptor (AR) activity, including truncated AR.


BACKGROUND OF THE INVENTION

Androgens mediate their effects through the androgen receptor (AR). Androgens play a role in a wide range of developmental and physiological responses and are involved in male sexual differentiation, maintenance of spermatogenesis, and male gonadotropin regulation (R. K. Ross, G. A. Coetzee, C. L. Pearce, J. K. Reichardt, P. Bretsky, L. N. Kolonel, B. E. Henderson, E. Lander, D. Altshuler & G. Daley, Eur Urol 35, 355-361 (1999); A. A. Thomson, Reproduction 121, 187-195 (2001); N. Tanji, K. Aoki & M. Yokoyama, Arch Androl 47, 1-7 (2001)). Several lines of evidence show that androgens are associated with the development of prostate carcinogenesis. Firstly, androgens induce prostatic carcinogenesis in rodent models (R. L. Noble, Cancer Res 37, 1929-1933 (1977); R. L. Noble, Oncology 34, 138-141 (1977)) and men receiving androgens in the form of anabolic steroids have a higher incidence of prostate cancer (J. T. Roberts & D. M. Essenhigh, Lancet 2, 742 (1986); J. A. Jackson, J. Waxman & A. M. Spiekerman, Arch Intern Med 149, 2365-2366 (1989); P. D. Guinan, W. Sadoughi, H. Alsheik, R. J. Ablin, D. Alrenga & I. M. Bush, Am J Surg 131, 599-600 (1976)). Secondly, prostate cancer does not develop if humans or dogs are castrated before puberty (J. D. Wilson & C. Roehrborn, J Clin Endocrinol Metab 84, 4324-4331 (1999); G. Wilding, Cancer Surv 14, 113-130 (1992)). Castration of adult males causes involution of the prostate and apoptosis of prostatic epithelium while eliciting no effect on other male external genitalia (E. M. Bruckheimer & N. Kyprianou, Cell Tissue Res 301, 153-162 (2000); J. T. Isaacs, Prostate 5, 545-557 (1984)). This dependency on androgens provides the underlying rationale for treating prostate cancer with chemical or surgical castration (androgen ablation), also known as androgen ablation therapy (ABT) or androgen depravation therapy (ADT).


Androgens also play a role in female diseases such as polycystic ovary syndrome as well as cancers. One example is ovarian cancer where elevated levels of androgens are associated with an increased risk of developing ovarian cancer (K. J. Helzlsouer, A. J. Alberg, G. B. Gordon, C. Longcope, T. L. Bush, S. C. Hoffman & G. W. Comstock, JAMA 274, 1926-1930 (1995); R. J. Edmondson, J. M. Monaghan & B. R. Davies, Br J Cancer 86, 879-885 (2002)). The AR has been detected in a majority of ovarian cancers (H. A. Risch, J Natl Cancer Inst 90, 1774-1786 (1998); B. R. Rao & B. J. Slotman, Endocr Rev 12, 14-26 (1991); G. M. Clinton & W. Hua, Crit Rev Oncol Hematol 25, 1-9 (1997)), whereas estrogen receptor-alpha (ERa) and the progesterone receptor are detected in less than 50% of ovarian tumors.


The only effective treatment available for advanced prostate cancer is the withdrawal of androgens which are essential for the survival of prostate luminal cells. Androgen ablation therapy causes a temporary reduction in tumor burden concomitant with a decrease in serum prostate-specific antigen (PSA). Unfortunately, prostate cancer can eventually grow again in the absence of testicular androgens (castration-resistant disease) (Huber et al 1987 Scand J. Urol Nephrol. 104, 33-39). Castration-resistant prostate cancer that is still driven by AR is biochemically characterized before the onset of symptoms by a rising titre of serum PSA (Miller et al 1992 J. Urol. 147, 956-961). Once the disease becomes castration-resistant most patients succumb to their disease within two years.


The AR has distinct functional domains that include the carboxy-terminal ligand-binding domain (LBD), a DNA-binding domain (DBD) comprising two zinc finger motifs, and an N-terminus domain (NTD) that contains two transcriptional activation units (tau1 and tau5) within activation function-1 (AF-1). Binding of androgen (ligand) to the LBD of the AR results in its activation such that the receptor can effectively bind to its specific DNA consensus site, termed the androgen response element (ARE), on the promoter and enhancer regions of “normally” androgen regulated genes, such as PSA, to initiate transcription. The AR can be activated in the absence of androgen by stimulation of the cAMP-dependent protein kinase (PKA) pathway, with interleukin-6 (IL-6) and by various growth factors (Culig et al 1994 Cancer Res. 54, 5474-5478; Nazareth et al 1996 J. Biol. Chem. 271, 19900-19907; Sadar 1999 J. Biol. Chem. 274, 7777-7783; Ueda et al 2002 A J. Biol. Chem. 277, 7076-7085; and Ueda et al 2002 B J. Biol. Chem. 277, 38087-38094). The mechanism of ligand-independent transformation of the AR has been shown to involve: 1) increased nuclear AR protein suggesting nuclear translocation; 2) increased AR/ARE complex formation; and 3) the AR-NTD (Sadar 1999 J. Biol. Chem. 274, 7777-7783; Ueda et al 2002 A J Biol. Chem. 277, 7076-7085; and Ueda et al 2002 B J. Biol. Chem. 277, 38087-38094). The AR can be activated in the absence of testicular androgens by alternative signal transduction pathways in castration-resistant disease, which is consistent with the finding that nuclear AR protein is present in secondary prostate cancer tumors (Kim et al 2002 Am. J. Pathol. 160, 219-226; and van der Kwast et al 1991 Inter. J. Cancer 48, 189-193).


The AR-NTD is also a target for drug development (e.g. WO 2000/001813; Myung et al. J. Clin. Invest 2013, 123, 2948), since the NTD contains Activation-Function-1 (AF-1) which is the essential region required for AR transcriptional activity (Jenster et al 1991. Mol Endocrinol. 5, 1396-404). The AR-NTD importantly plays a role in activation of the AR in the absence of androgens (Sadar, M. D. 1999 J. Biol. Chem. 274, 7777-7783; Sadar M D et al 1999 Endocr Relat Cancer. 6, 487-502; Ueda et al 2002 J. Biol. Chem. 277, 7076-7085; Ueda 2002 J. Biol. Chem. 277, 38087-38094; Blaszczyk et al 2004 Clin Cancer Res. 10, 1860-9; Dehm et al 2006 J Biol Chem. 28, 27882-93; Gregory et al 2004 J Biol Chem. 279, 7119-30). The AR-NTD is important in hormonal progression of prostate cancer as shown by application of decoy molecules (Quayle et al 2007, Proc Natl Acad Sci USA. 104, 1331-1336).


While the crystal structure has been resolved for the AR C-terminus LBD, this has not been the case for the NTD due to its high flexibility and intrinsic disorder in solution (Reid et al 2002 J. Biol. Chem. 277, 20079-20086) thereby hampering virtual docking drug discovery approaches. Compounds that modulate AR, potentially through interaction with NTD domain, include the bisphenol compounds disclosed in published PCT Nos: WO 2010/000066, WO 2011/082487; WO 2011/082488; WO 2012/145330; WO 2012/139039; WO 2012/145328; WO 2013/028572; WO 2013/028791; WO 2014/179867; WO 2015/031984; WO 2016/058080; WO 2016/058082; WO 2016/112455; WO 2016/141458; WO 2017/177307; WO 2017/210771; WO 2018/045450, and WO 2019/226991, WO 2020/081999, and WO 2022/226349, and which are hereby incorporated by reference in their entireties.


While significant advances have been made in this field, there remains a need for improved treatment for AR-mediated disorders including prostate cancer, especially metastatic castration-resistant prostate cancer. Development of compounds, via unique interactions with AR NTD, would provide patients alternative options and new hope.


SUMMARY OF THE INVENTION

The compounds of the present disclosure are androgen receptor modulators which may be useful in treating various diseases and conditions as disclosed herein.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (I-A):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 3- to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —(CR5R6)t—, —O—, or —NR7;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, —NR7—, or —N(COCH3)—;
      • W is a bond, —(CR8aR9a)m—, —C(═O)—, —NR7—, —N(R7)CO—, —CONR7—, —NSO2R7—, heteroarylene, or heterocyclene;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is a bond, —(CR8aR9a)m—, —CR8a═CR9a—, or phenylene;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —SO2R17, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), —N═S(═O)(C1-C3 alkyl)2,







embedded image




    •  optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 deuterated, C2-C6 alkenyl, or C2-C6 alkynyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3 to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R11 and R12 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • R17 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, or optionally substituted C2-C3 alkynyl;
      • each m is independently 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, 2, 3, 4 or 5; and
      • t is 0, 1 or 2.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (A):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 4 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is —O—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, —CONR7—, or heteroarylene;
      • Z is a bond or —O—;
      • V is a bond, —(CR8aR9a)m(CR8aR9a)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, oxo, —CN, or —CF3;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14C00R16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), —N═S(═O)(C1-C3 alkyl)2,







embedded image




    •  or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen or C1-C4 alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (B-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a phenyl or a pyridyl ring;
      • C is a monocyclic carbocycle, a spiral bicyclic carbocycle, a bridged bicyclic carbocycle, a monocyclic heterocycle, or a spiral bicyclic heterocycle;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1, 2, 3, 4 or 5; and

    • wherein when C is a bridged bicyclic carbocycle, —Z—V-L is not —OCH2CH2Cl or —OCH3.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (C-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond;
      • W is a bond, heteroaryl, heterocyclyl, -heteroaryl-NH—*, or -heterocyclyl-NH—*, wherein heteroaryl or heterocyclyl of W is optionally substituted and wherein * indicates connection to ring C;
      • Z is a bond or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, oxo, —CN, or —CF3;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14C00R16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), optionally substituted 3 to 7-membered carbocycle, or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5;
      • wherein when A is a fused or a bridged bicyclic ring; C is a 4- to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl; and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond; and
      • wherein when A is phenyl, C is a fused or a bridged bicyclic ring selected from







embedded image


and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (D-J):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —(CR8aR9a)m-(optionally substituted carbocycle), —(CR8aR9a)m1-(optionally substituted aryl), —(CR8aR9a)m-(optionally substituted heterocycle), —(CR8aR9a)m-(optionally substituted heteroaryl), —CHCH2, —CHF2, —CH3, —CH2CH2CH3, —CH2CH2NHSO2CH3, or —CH2CH2CH2OC(O)NH-(optionally substituted aryl);
      • Z is a bond and —V-L is —(CR8aR9a)m—NR11R12; or
      • Z is a bond and —V-L is —CH2CH2OH, —CH2CH2Cl, or —CHCH2;
      • R2 is Cl or —CN;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • n3 is 1, 2, or 3;
      • each m is independently 0, 1, 2, or 3;
      • each m1 is independently 1, 2, or 3; and

    • wherein when R2 is —CN, then —Z—V-L is not







embedded image


In one embodiment, the present disclosure provides compounds comprising the structure of formula (F):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently phenyl or pyridyl;
      • C is -pyrimidyl-NHSO2-(optionally substituted heteroaryl) or -pyrimidyl-(optionally substituted heteroaryl)-R3;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), or —NHCOCF3;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O; and
      • m is 0, 1, 2, or 3.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (E-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is a bond —NH—, or —O—; and
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4-6 membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4-6 membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl; and
      • n3 is 1, 2, or 3.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (G):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1;
      • R1 is each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n3 is 1, 2, or 3;
      • wherein:
      • a) n1 is 1; and/or
      • b) n3 is 2, wherein the second R3 is —NH2, optionally substituted piperidine, optionally substituted piperazine.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (H):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is phenyl or pyridyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1;
      • R1 and R2 are each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • at least one R3 is halogen and at least one other R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and if a third R3 is present, then it is halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n2 is 0, 1, or 2;
      • n3 is 2 or 3; and

    • wherein







embedded image


is not




embedded image


In one embodiment, the present disclosure provides compounds comprising the structure of formula (J):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • B is 6-membered heteroaryl or 6-membered heterocyclyl;
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1; or
      • Z is a bond and —V-L is —CH2CH2CH2C1;
      • R1 and R2 are each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14 optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • when B is a 6-membered heterocyclyl, R2 can also be an oxo;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3, if present, is halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n2 is 0, 1, or 2;
      • n3 is 1, 2 or 3; and

    • wherein B is







embedded image




    •  R2 is not Cl.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (K):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a —CH2—, —C(O)—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1;
      • R1 and R2 are each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14 optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n2 is 0, 1, or 2; and
      • n3 is 1, 2, or 3





In one embodiment of the compound of the present disclosure is selected from Compounds A1-A185, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of the present disclosure is selected from Compounds A1-A245, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compounds of the present disclosure, compounds disclosed in WO 2020/081999 is excluded. In one embodiment of the compounds of the present disclosure, compounds disclosed in WO 2022/226349 is excluded.


The present disclosure further provides a pharmaceutical composition comprising a compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), and a pharmaceutically acceptable carrier.


The present disclosure further provides a method for treating cancer, comprising administering the compound, pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), to a subject in need thereof.


In one embodiment of the methods disclosed herein, cancer is prostate cancer. In one embodiment, the prostate cancer is metastatic castration-resistant prostate cancer. In one embodiment, the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.


DETAILED DESCRIPTION

All publications, patents and patent applications, including any drawings and appendices therein are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent or patent application, drawing, or appendix was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.


Definitions

While the following terms are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter.


Throughout the present specification, the terms “about” and/or “approximately” may be used in conjunction with numerical values and/or ranges. The term “about” is understood to mean those values near to a recited value. Furthermore, the phrases “less than about [a value]” or “greater than about [a value]” should be understood in view of the definition of the term “about” provided herein. The terms “about” and “approximately” may be used interchangeably.


Throughout the present specification, numerical ranges are provided for certain quantities. It is to be understood that these ranges comprise all subranges therein. Thus, the range “from 50 to 80” includes all possible ranges therein (e.g., 51-79, 52-78, 53-77, 54-76, 55-75, 60-70, etc.). Furthermore, all values within a given range may be an endpoint for the range encompassed thereby (e.g., the range 50-80 includes the ranges with endpoints such as 55-80, 50-75, etc.).


The term “a” or “an” refers to one or more of that entity; for example, “a androgen receptor modulator” refers to one or more androgen receptor modulators or at least one androgen receptor modulator. As such, the terms “a” (or “an”), “one or more” and “at least one” are used interchangeably herein. In addition, reference to “an inhibitor” by the indefinite article “a” or “an” does not exclude the possibility that more than one of the inhibitors is present, unless the context clearly requires that there is one and only one of the inhibitors.


As used herein, the verb “comprise” as is used in this description and in the claims and its conjugations are used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. The present invention may suitably “comprise”, “consist of”, or “consist essentially of”, the steps, elements, and/or reagents described in the claims.


It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely”, “only” and the like in connection with the recitation of claim elements, or the use of a “negative” limitation.


The term “pharmaceutically acceptable salts” includes both acid and base addition salts. Pharmaceutically acceptable salts include those obtained by reacting the active compound functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, camphorsulfonic acid, oxalic acid, maleic acid, succinic acid, citric acid, formic acid, hydrobromic acid, benzoic acid, tartaric acid, fumaric acid, salicylic acid, mandelic acid, carbonic acid, etc. Those skilled in the art will further recognize that acid addition salts may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods.


The term “treating” means one or more of relieving, alleviating, delaying, reducing, improving, or managing at least one symptom of a condition in a subject. The term “treating” may also mean one or more of arresting, delaying the onset (i.e., the period prior to clinical manifestation of the condition) or reducing the risk of developing or worsening a condition.


The compounds of the invention, or their pharmaceutically acceptable salts can contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that can be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms whether or not they are specifically depicted herein. Optically active (+) and (−), (R)- and (S)-, or (D)- and (L)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.


A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposable mirror images of one another.


A “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any said compounds.


A “prodrug” refers to a derivative of a compound of the present disclosure that will be converted to the compound in vivo. In one embodiment of the present disclosure, a prodrug includes a compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), having a free hydroxyl group (—OH) that is acetylated (—OCOMe) at one or more positions.


An “effective amount” means the amount of a formulation according to the invention that, when administered to a patient for treating a state, disorder or condition is sufficient to effect such treatment. The “effective amount” will vary depending on the active ingredient, the state, disorder, or condition to be treated and its severity, and the age, weight, physical condition and responsiveness of the mammal to be treated.


The term “therapeutically effective” applied to dose or amount refers to that quantity of a compound or pharmaceutical formulation that is sufficient to result in a desired clinical benefit after administration to a patient in need thereof.


As used herein, the term “pharmaceutical composition” refers to a formulation comprising at least one therapeutically active agent and a pharmaceutically acceptable excipient or carrier. A non-limiting example of pharmaceutical compositions includes tablets, capsules, gel capsules, syrup, liquid, gel, suspension, solid dispersion, or combinations thereof.


As used herein, the term “dosage form” refers to one or more pharmaceutical compositions which provides a specific amount of a therapeutically active agent, such as a unit dose.


As used herein, a “subject” can be a human, non-human primate, mammal, rat, mouse, cow, horse, pig, sheep, goat, dog, cat and the like. The subject can be suspected of having or at risk for having a cancer, such as prostate cancer, breast cancer, ovarian cancer, salivary gland carcinoma, or endometrial cancer, or suspected of having or at risk for having acne, hirsutism, alopecia, benign prostatic hyperplasia, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, or age-related macular degeneration. Diagnostic methods for various cancers, such as prostate cancer, breast cancer, ovarian cancer, bladder cancer, pancreatic cancer, hepatocellular cancer, salivary gland carcinoma, or endometrial cancer, and diagnostic methods for acne, hirsutism, alopecia, benign prostatic hyperplasia, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, or age-related macular degeneration and the clinical delineation of cancer, such as prostate cancer, breast cancer, ovarian cancer, bladder cancer, pancreatic cancer, hepatocellular cancer, salivary gland carcinoma, or endometrial cancer, diagnoses and the clinical delineation of acne, hirsutism, alopecia, benign prostatic hyperplasia, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, or age-related macular degeneration are known to those of ordinary skill in the art.


“Mammal” includes humans and both domestic animals such as laboratory animals (e.g., mice, rats, monkeys, dogs, etc.) and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.


All weight percentages (i.e., “% by weight” and “wt. %” and w/w) referenced herein, unless otherwise indicated, are measured relative to the total weight of the pharmaceutical composition.


As used herein, “substantially” or “substantial” refers to the complete or nearly complete extent or degree of an action, characteristic, property, state, structure, item, or result. For example, an object that is “substantially” enclosed would mean that the object is either completely enclosed or nearly completely enclosed. The exact allowable degree of deviation from absolute completeness may in some cases depend on the specific context. However, generally speaking, the nearness of completion will be so as to have the same overall result as if absolute and total completion were obtained. The use of “substantially” is equally applicable when used in a negative connotation to refer to the complete or near complete lack of action, characteristic, property, state, structure, item, or result. For example, a composition that is “substantially free of” other active agents would either completely lack other active agents, or so nearly completely lack other active agents that the effect would be the same as if it completely lacked other active agents. In other words, a composition that is “substantially free of” an ingredient or element or another active agent may still contain such an item as long as there is no measurable effect thereof.


The terms below, as used herein, have the following meanings, unless indicated otherwise:


“Amino” refers to the —NH2 radical.


“Cyano” refers to the —CN radical.


“Halo” or “halogen” refers to bromo, chloro, fluoro or iodo radical, including their radioisotopes. “123I” refers to the radioactive isotope of iodine having atomic mass 123. The compounds of Formula I can comprise at least one 123I moiety. Throughout the present application, where structures depict a 123I moiety at a certain position it is meant that the I moiety at this position is enriched for 123I. In other words, the compounds contain more than the natural abundance of 123I at the indicated position(s). It is not required that the compounds comprise 100% 123I at the indicated positions, provided 123I is present in more than the natural abundance. Typically, the 123I isotope is enriched to greater than 50%, greater than 60%, greater than 70%, greater than, 80% or greater than 90%, relative to 127I. “18F” refers to the radioactive isotope of fluorine having atomic mass 18. “F” or “19F” refers to the abundant, non-radioactive fluorine isotope having atomic mass 19. The compounds of Formula I can comprise at least one 18F moiety. Throughout the present application, where structures depict a 18F moiety at a certain position it is meant that the F moiety at this position is enriched for 18F. In other words, the compounds contain more than the natural abundance of 18F at the indicated position(s). It is not required that the compounds comprise 100% 18F at the indicated positions, provided 18F is present in more than the natural abundance. Typically, the 18F isotope is enriched to greater than 50%, greater than 60%, greater than 70%, greater than 80% or greater than 90%, relative to 19F.


“Hydroxy” or “hydroxyl” refers to the —OH radical.


“Imino” refers to the ═NH substituent.


“Nitro” refers to the —NO2 radical.


“Oxo” refers to the ═O substituent.


“Thioxo” refers to the ═S substituent.


“Alkyl” or “alkyl group” refers to a fully saturated, straight or branched hydrocarbon chain radical having from one to twelve carbon atoms, and which is attached to the rest of the molecule by a single bond. Alkyls comprising any number of carbon atoms from 1 to 12 are included. An alkyl comprising up to 12 carbon atoms is a C1-C12 alkyl, an alkyl comprising up to 10 carbon atoms is a C1-C10 alkyl, an alkyl comprising up to 6 carbon atoms is a C1-C6 alkyl and an alkyl comprising up to 5 carbon atoms is a C1-C5 alkyl. A C1-C5 alkyl includes C5 alkyls, C4 alkyls, C3 alkyls, C2 alkyls and C1 alkyl (i.e., methyl). A C1-C6 alkyl includes all moieties described above for C1-C5 alkyls but also includes C6 alkyls. A C1-C10 alkyl includes all moieties described above for C1-C5 alkyls and C1-C6 alkyls, but also includes C7, C8, C9 and C10 alkyls. Similarly, a C1-C12 alkyl includes all the foregoing moieties, but also includes C11 and C12 alkyls. Non-limiting examples of C1-C12 alkyl include methyl, ethyl, n-propyl, i-propyl, sec-propyl, n-butyl, i-butyl, sec-butyl, t-butyl, n-pentyl, t-amyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, n-undecyl, and n-dodecyl. Unless stated otherwise specifically in the specification, an alkyl group can be optionally substituted.


“Alkylene” or “alkylene chain” refers to a fully saturated, straight or branched divalent hydrocarbon chain radical, and having from one to twelve carbon atoms. Non-limiting examples of C1-C12 alkylene include methylene, ethylene, propylene, n-butylene, ethenylene, propenylene, n-butenylene, propynylene, n-butynylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain can be optionally substituted.


“Alkenyl” or “alkenyl group” refers to a straight or branched hydrocarbon chain radical having from two to twelve carbon atoms and having one or more carbon-carbon double bonds. Each alkenyl group is attached to the rest of the molecule by a single bond. Alkenyl group comprising any number of carbon atoms from 2 to 12 are included. An alkenyl group comprising up to 12 carbon atoms is a C2-C12 alkenyl, an alkenyl comprising up to 10 carbon atoms is a C2-C10 alkenyl, an alkenyl group comprising up to 6 carbon atoms is a C2-C6 alkenyl and an alkenyl comprising up to 5 carbon atoms is a C2-C5 alkenyl. A C2-C5 alkenyl includes C5 alkenyls, C4 alkenyls, C3 alkenyls, and C2 alkenyls. A C2-C6 alkenyl includes all moieties described above for C2-C5 alkenyls but also includes C6 alkenyls. A C2-C10 alkenyl includes all moieties described above for C2-C5 alkenyls and C2-C6 alkenyls, but also includes C7, C8, C9 and C10 alkenyls. Similarly, a C2-C12 alkenyl includes all the foregoing moieties, but also includes C11 and C12 alkenyls. Non-limiting examples of C2-C12 alkenyl include ethenyl (vinyl), 1-propenyl, 2-propenyl (allyl), iso-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-heptenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 5-heptenyl, 6-heptenyl, 1-octenyl, 2-octenyl, 3-octenyl, 4-octenyl, 5-octenyl, 6-octenyl, 7-octenyl, 1-nonenyl, 2-nonenyl, 3-nonenyl, 4-nonenyl, 5-nonenyl, 6-nonenyl, 7-nonenyl, 8-nonenyl, 1-decenyl, 2-decenyl, 3-decenyl, 4-decenyl, 5-decenyl, 6-decenyl, 7-decenyl, 8-decenyl, 9-decenyl, 1-undecenyl, 2-undecenyl, 3-undecenyl, 4-undecenyl, 5-undecenyl, 6-undecenyl, 7-undecenyl, 8-undecenyl, 9-undecenyl, 10-undecenyl, 1-dodecenyl, 2-dodecenyl, 3-dodecenyl, 4-dodecenyl, 5-dodecenyl, 6-dodecenyl, 7-dodecenyl, 8-dodecenyl, 9-dodecenyl, 10-dodecenyl, and 11-dodecenyl. Unless stated otherwise specifically in the specification, an alkyl group can be optionally substituted.


“Alkenylene” or “alkenylene chain” refers to a straight or branched divalent hydrocarbon chain radical, having from two to twelve carbon atoms, and having one or more carbon-carbon double bonds. Non-limiting examples of C2-C12 alkenylene include ethene, propene, butene, and the like. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkenylene chain can be optionally substituted.


“Alkynyl” or “alkynyl group” refers to a straight or branched hydrocarbon chain radical having from two to twelve carbon atoms and having one or more carbon-carbon triple bonds. Each alkynyl group is attached to the rest of the molecule by a single bond. Alkynyl group comprising any number of carbon atoms from 2 to 12 are included. An alkynyl group comprising up to 12 carbon atoms is a C2-C12 alkynyl, an alkynyl comprising up to 10 carbon atoms is a C2-C10 alkynyl, an alkynyl group comprising up to 6 carbon atoms is a C2-C6 alkynyl and an alkynyl comprising up to 5 carbon atoms is a C2-C5 alkynyl. A C2-C5 alkynyl includes C5 alkynyls, C4 alkynyls, C3 alkynyls, and C2 alkynyls. A C2-C6 alkynyl includes all moieties described above for C2-C5 alkynyls but also includes C6 alkynyls. A C2-C10 alkynyl includes all moieties described above for C2-C5 alkynyls and C2-C6 alkynyls, but also includes C7, C8, C9 and C10 alkynyls. Similarly, a C2-C12 alkynyl includes all the foregoing moieties, but also includes C11 and C12 alkynyls. Non-limiting examples of C2-C12 alkynyl include ethynyl, propynyl, butynyl, pentynyl and the like. Unless stated otherwise specifically in the specification, an alkynyl group can be optionally substituted.


“Alkynylene” or “alkynylene chain” refers to a straight or branched divalent hydrocarbon chain radical, having from two to twelve carbon atoms, and having one or more carbon-carbon triple bonds. Non-limiting examples of C2-C12 alkynylene include ethynylene, propargylene and the like. The alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkynylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkynylene chain can be optionally substituted.


“Alkoxy” refers to a radical of the formula —ORa where Ra is an alkyl, alkenyl or alknyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group can be optionally substituted.


“Alkylamino” refers to a radical of the formula —NHRa or —NRaRa where each Ra is, independently, an alkyl, alkenyl or alkynyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylamino group can be optionally substituted.


“Alkylcarbonyl” refers to the —C(═O)Ra moiety, wherein Ra is an alkyl, alkenyl or alkynyl radical as defined above. A non-limiting example of an alkyl carbonyl is the methyl carbonyl (“acetal”) moiety. Alkylcarbonyl groups can also be referred to as “Cw-Cz acyl” where w and z depicts the range of the number of carbon atoms in Ra, as defined above. For example, “C1-C10 acyl” refers to alkylcarbonyl group as defined above, where Ra is C1-C10 alkyl, C1-C10 alkenyl, or C1-C10 alkynyl radical as defined above. Unless stated otherwise specifically in the specification, an alkylcarbonyl group can be optionally substituted.


“Aryl” refers to a hydrocarbon ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl radical can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term “aryl” is meant to include aryl radicals that are optionally substituted.


“Aralkyl” or “arylalkyl” refers to a radical of the formula —Rb-Rc where Rb is an alkylene group as defined above and Rc is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated otherwise specifically in the specification, an aralkyl group can be optionally substituted.


“Aralkenyl” or “arylalkenyl” refers to a radical of the formula —Rb-Rc where Rb is an alkenylene o group as defined above and Rc is one or more aryl radicals as defined above. Unless stated otherwise specifically in the specification, an aralkenyl group can be optionally substituted.


“Aralkynyl” or “arylalkynyl” refers to a radical of the formula —Rb-Rc where Rb is an alkynylene group as defined above and Rc is one or more aryl radicals as defined above. Unless stated otherwise specifically in the specification, an aralkynyl group can be optionally substituted.


“Carbocyclyl,” “carbocyclic ring” or “carbocycle” refers to a rings structure, wherein the atoms which form the ring are each carbon. Carbocyclic rings can comprise from 3 to 20 carbon atoms in the ring. Carbocyclic rings include aryls and cycloalkyl. cycloalkenyl and cycloalkynyl as defined herein. Unless stated otherwise specifically in the specification, a carbocyclyl group can be optionally substituted.


“Cycloalkyl” refers to a stable non-aromatic monocyclic or polycyclic fully saturated hydrocarbon radical consisting solely of carbon and hydrogen atoms, which can include fused or bridged ring systems, having from three to twenty carbon atoms, preferably having from three to ten carbon atoms, and which is attached to the rest of the molecule by a single bond. Monocyclic cycloalkyl radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyl radicals include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyl group can be optionally substituted.


“Cycloalkenyl” refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having one or more carbon-carbon double bonds, which can include fused or bridged ring systems, having from three to twenty carbon atoms, preferably having from three to ten carbon atoms, and which is attached to the rest of the molecule by a single bond. Monocyclic cycloalkenyl radicals include, for example, cyclopentenyl, cyclohexenyl, cycloheptenyl, cycloctenyl, and the like. Polycyclic cycloalkenyl radicals include, for example, bicyclo[2.2.1]hept-2-enyl and the like. Unless otherwise stated specifically in the specification, a cycloalkenyl group can be optionally substituted.


“Cycloalkynyl” refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having one or more carbon-carbon triple bonds, which can include fused or bridged ring systems, having from three to twenty carbon atoms, preferably having from three to ten carbon atoms, and which is attached to the rest of the molecule by a single bond. Monocyclic cycloalkynyl radicals include, for example, cycloheptynyl, cyclooctynyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkynyl group can be optionally substituted.


“Cycloalkylalkyl” refers to a radical of the formula —Rb-Rd where Rb is an alkylene, alkenylene, or alkynylene group as defined above and Rd is a cycloalkyl, cycloalkenyl, cycloalkynyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group can be optionally substituted.


“Haloalkyl” refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group can be optionally substituted.


“Haloalkenyl” refers to an alkenyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., 1-fluoropropenyl, 1,1-difluorobutenyl, and the like. Unless stated otherwise specifically in the specification, a haloalkenyl group can be optionally substituted.


“Haloalkynyl” refers to an alkynyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., 1-fluoropropynyl, 1-fluorobutynyl, and the like. Unless stated otherwise specifically in the specification, a haloalkynyl group can be optionally substituted.


“Heterocyclyl,” “heterocyclic ring” or “heterocycle” refers to a stable 3- to 20-membered non-aromatic, partially aromatic, or aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Heterocyclycl or heterocyclic rings include heteroaryls as defined below. Unless stated otherwise specifically in the specification, the heterocyclyl radical can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical can be optionally oxidized; the nitrogen atom can be optionally quaternized; and the heterocyclyl radical can be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocyclyl group can be optionally substituted.


“Heterocyclylalkyl” refers to a radical of the formula —Rb-Re where Rb is an alkylene group as defined above and Re is a heterocyclyl radical as defined above. Unless stated otherwise specifically in the specification, a heterocycloalkylalkyl group can be optionally substituted.


“Heterocyclylalkenyl” refers to a radical of the formula —Rb-Re where Rb is an alkenylene group as defined above and Re is a heterocyclyl radical as defined above. Unless stated otherwise specifically in the specification, a heterocycloalkylalkenyl group can be optionally substituted.


“Heterocyclylalkynyl” refers to a radical of the formula —Rb-Re where Rb is an alkynylene group as defined above and Re is a heterocyclyl radical as defined above. Unless stated otherwise specifically in the specification, a heterocycloalkylalkynyl group can be optionally substituted.


“N-heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a N-heterocyclyl group can be optionally substituted.


“Heteroaryl” refers to a 5- to 20-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical can be optionally oxidized; the nitrogen atom can be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophene), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophene, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophene (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group can be optionally substituted.


“N-heteroaryl” refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an N-heteroaryl group can be optionally substituted.


“Heteroarylalkyl” refers to a radical of the formula —Rb-Rf where Rb is an alkylene chain as defined above and Rf is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group can be optionally substituted.


“Heteroarylalkenyl” refers to a radical of the formula —Rb-Rf where Rb is an alkenylene, chain as defined above and Rf is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkenyl group can be optionally substituted.


“Heteroarylalkynyl” refers to a radical of the formula —Rb-Rf where Rb is an alkynylene chain as defined above and Rf is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkynyl group can be optionally substituted.


“Ring” refers to a cyclic group which can be fully saturated, partially saturated, or fully unsaturated. A ring can be monocyclic, bicyclic, tricyclic, or tetracyclic. Unless stated otherwise specifically in the specification, a ring can be optionally substituted.


“Thioalkyl” refers to a radical of the formula —SRa where Ra is an alkyl, alkenyl, or alkynyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group can be optionally substituted.


The term “substituted” used herein means any of the above groups (i.e., alkyl, alkylene, alkenyl, alkenylene, alkynyl, alkynylene, alkoxy, alkylamino, alkylcarbonyl, thioalkyl, aryl, aralkyl, carbocyclyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups.


“Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, “substituted” includes any of the above groups in which one or more hydrogen atoms are replaced with —NRgRh, —NRgC(═O)Rh, —NRgC(═O)NRgRh, —NRgC(═O)ORh, —NRgSO2Rh, —OC(═O)NRgRh, —ORg, —SRg, —SORg, —SO2Rg, —OSO2Rg, —SO2ORg, ═NSO2Rg, and —SO2NRgRh. “Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with —C(═O)Rg, —C(═O)ORg, —C(═O)NRgRh, —CH2SO2Rg, —CH2SO2NRgRh. In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkenyl, alkynyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, haloalkenyl, haloalkynyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl. “Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkenyl, alkynyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, haloalkenyl, haloalkynyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing substituents can also be optionally substituted with one or more of the above substituents.


As used herein, the symbol




embedded image


(hereinafter can be referred to as “a point of attachment bond”) denotes a bond that is a point of attachment between two chemical entities, one of which is depicted as being attached to the point of attachment bond and the other of which is not depicted as being attached to the point of attachment bond. For example,




embedded image


indicates that the chemical entity “XY” is bonded to another chemical entity via the point of attachment bond. Furthermore, the specific point of attachment to the non-depicted chemical entity can be specified by inference. For example, the compound CH3—R3, wherein R3 is H or




embedded image


infers that when R3 is “XY”, the point of attachment bond is the same bond as the bond by which R3 is depicted as being bonded to CH3. When a group is divalent which can be denoted as having two points of attachment such as




embedded image


or —X—Y—, unless it is expressly stated, the divalent group can attach in either direction. That is, if the above divalent group (—X—Y—) represents U in T-U—V, then it can be T-X—Y—V or T-Y—X-V.


“Fused” refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention. When the fused ring is a heterocyclyl ring or a heteroaryl ring, any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring can be replaced with a nitrogen atom.


The following description includes information that may be useful in understanding the present invention. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed inventions, or that any publication specifically or implicitly referenced is prior art.


Compounds of the Present Disclosure

The compound of the present disclosure can be useful for modulating androgen receptor (AR). Further, the compound of the present disclosure can be useful for treating various diseases and conditions including, but not limited to, cancer. In some embodiments, the cancer is prostate cancer or breast cancer.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 3 to 15-membered ring;
      • X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, —NR7—, —N(R7)CO—, —CON(R7)—, or —NSO2R7—;
      • Y and Z are each independently a bond, —(CR8R9)m—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W and V are each independently a bond, —(CR8aR9a)m—, a ring, -(a ring)(NR7)—, C(═O)—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10; —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16, optionally substituted —(C1-C6 alkyl)-SO2R16, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R3 is hydrogen, halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —SR16, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16, optionally substituted —(C1-C6 alkyl)-SO2R16, —N═S(═O)(C1-C3 alkyl)2,







embedded image




    •  optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted C1-C6 alkoxy, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C6 alkyl)-CONR14R15, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R5 and R6 taken together form an optionally substituted carbocyclyl or optionally substituted heterocyclyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted C1-C6 alkoxy, optionally substituted —OCO(C1-C6 alkyl), —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C6 alkyl)-CONR14R15, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R8a and R9a taken together form an optionally substituted carbocyclyl or optionally substituted heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R7 and R8a taken together form an optionally substituted heterocyclyl;
      • R11, R12, R13, R14 and R15 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or (R11 and R12) or (R14 and R15) taken together form an optionally substituted heterocyclyl;
      • each m is independently 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, 2, 3, or 4;
      • n3 is 0, 1, 2, 3, 4 or 5; and
      • each t is independently 0, 1 or 2.





In one embodiment of the compounds of formula (I), W is a ring. In some embodiments, W is carbocycle, aryl, heterocycle, or heteroaryl. In some embodiments, W is 3-10 membered carbocycle, aryl, heterocycle, or heteroaryl.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (I-A):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 3- to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —(CR5R6)t—, —O—, or —NR7;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, —NR7—, or —N(COCH3)—;
      • W is a bond, a ring, —(CR8aR9a)m—, —C(═O)—, —NR7—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is a bond, —(CR8aR9a)m—, —CR8a═CR9a—, or phenylene;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —SO2R17, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), optionally substituted carbocyclyl, —N═S(═O)(C1-C3 alkyl)2,







embedded image




    •  optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 deuterated, C2-C6 alkenyl, or C2-C6 alkynyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R10 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R11 and R12 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • R17 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, or optionally substituted C2-C3 alkynyl;
      • each m is independently 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, 2, 3, 4 or 5; and
      • t is 0, 1 or 2.





In one embodiment of the compounds of formula (I-A), W is heteroarylene or heterocyclene.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (ABC):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is bond, —CH2—, —C(CH3)2—, —O— or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, ring, -ring-NR7—, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, —CONR7—, or heteroarylene;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (ABC), when A is a fused or a bridged bicyclic ring; C is a 4- to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl; X is a bond, —CH2—, or —C(CH3)2—; and Y is a bond; then W is not a bond.


In one embodiment of the compounds of formula (ABC), when A is phenyl, C is a fused or a bridged bicyclic ring; X is a bond, —CH2—, or —C(CH3)2—; and Y is a bond; then W is not a bond.


In one embodiment of the compounds of formula (ABC), A is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl.


In one embodiment of the compounds of formula (ABC), B is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl.


In one embodiment of the compounds of formula (ABC), C is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl.


In one embodiment of the compounds of formula (ABC), C is a monocyclic carbocycle, a spiral bicyclic carbocycle, a monocyclic heterocycle, or a spiral bicyclic heterocycle.


In one embodiment of the compounds of formula (ABC), X is O—.


In one embodiment of the compounds of formula (ABC), W is heteroaryl, heterocyclyl, -heteroaryl-NR7—, or -heterocyclyl-NR7—.


In one embodiment of the compounds of formula (ABC), Y is a bond and W is a bond, heteroaryl, heterocyclyl, -heteroaryl-NH—*, or -heterocyclyl-NH—*, wherein heteroaryl or heterocyclyl of W is optionally substituted and wherein * indicates connection to ring C.


In one embodiment of the compounds of formula (ABC), R1 and R2 are each independently halogen, oxo, —CN, or —CF3.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (A):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 4 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is —O—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, —CONR7—, or heteroarylene;
      • Z is a bond or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, oxo, —CN, or —CF3;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen or C1-C4 alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (ABC) or formula (A), C is not quinazoline ring.


In one embodiment of the compounds of formula (ABC) or formula (A), the compound is not




embedded image


In one embodiment of the compounds of formula (ABC) or formula (A), A is a 6-membered monocyclic ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl. In some embodiments, A is phenyl, pyridyl, pyrimidinyl, cyclohexyl, or piperidinyl. In some embodiments, A is phenyl, pyridyl, cyclohexyl, or piperidinyl.


In some embodiments of the compounds of formula (ABC) or formula (A), B is phenyl.


In some embodiments of the compounds of formula (ABC) or formula (A), Y is a bond. In some embodiments, Y is a bond or —O—.


In some embodiments of the compounds of formula (ABC) or formula (A), W is a bond, —CH2—, —CH2CH2—, or heteroarylene. In some embodiments, W is pyrazole, pyridine, pyrimidine, or pyrazine.


In some embodiments of the compounds of formula (ABC) or formula (A), C is a 4-10 membered monocyclic heterocyclyl, a 5-10 membered monocyclic heteroaryl, or a fused 8-10 membered bicyclic heteroaryl. In some embodiments, C is a 4-10 membered monocyclic carbocyclyl or a bridged 5-10 membered bicyclic carbocyclyl. In some embodiments, C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle. In some embodiments, C is




embedded image


embedded image


embedded image


In some embodiments, C is




embedded image


In some embodiments C is




embedded image


embedded image


In some embodiments, C is




embedded image


In some embodiments, C is




embedded image


In some embodiments of the compounds of formula (A), Y is a bond; W is a bond; and C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle.


In some embodiments of the compounds of formula (ABC) or formula (A), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —NH2, —NHCH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In some embodiments of the compounds of formula (ABC) or formula (A), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In some embodiments of the compounds of formula (ABC) or formula (A), at least one R3 is an optionally substituted 4- to 6-membered heterocyclyl or heteroaryl. In some embodiments, at least one R3 is an optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl). In some embodiments, at least one R3 is an optionally substituted 4- to 6-membered heterocyclyl containing one or two heteroatoms selected from N, O, or S. In some embodiments, at least one R3 is an optionally substituted




embedded image


In some embodiments, at least one R3 is




embedded image


In some embodiments of the compounds of formula (ABC) or formula (A), Z is —O—; V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; and L is hydrogen or halogen.


In some embodiments of the compounds of formula (ABC) or formula (A), Z—V-L is —O—CH3, —O—CH2CH2Cl, —O-cyclopropyl, or —O-cyclobutyl.


In some embodiments of the compounds of formula (ABC) or formula (A), Z is a bond; V is —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O; and L is hydrogen.


In some embodiments of the compounds of formula (A), B is phenyl and X and Z are connected at para positions of B.


In some embodiments of the compounds of formula (A), A is a 6-membered monocyclic ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl; and X and Y are connected at 1 and 4 positions of A.


In some embodiments of the compounds of formula (A), Z is —O—; V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; L is hydrogen or halogen; and C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle; wherein the compound is not




embedded image


In some embodiments of the compounds of formula (A), Z is —O—; V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; L is hydrogen or halogen; and C is a 6,6-fused heteroaryl or heterocycle; wherein the compound is not




embedded image


In some embodiments of the compounds of formula (A), Z—V-L is —O—CH3, —O—CH2CH2Cl, —O-cyclopropyl, or —O-cyclobutyl and C is a 6,6-fused heteroaryl or heterocycle; wherein the compound is not




embedded image


In some embodiments of the compounds of formula (A), Y is a bond; W is a bond; Z—V-L is —O—CH3, —O—CH2CH2Cl, —O-cyclopropyl, or —O-cyclobutyl; and C is a 6,6-fused heteroaryl or heterocycle; wherein the compound is not




embedded image


In some embodiments of the compounds of formula (ABC) or formula (A), Z—V-L is —O-cyclopropyl or —O-cyclobutyl; and C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle. In some embodiments of the compounds of formula (ABC) or formula (A), Z—V-L is —O-cyclopropyl or —O-cyclobutyl; and C is a 6,6-fused heteroaryl or heterocycle.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (BC):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, —CONR7—, heteroaryl, heterocyclyl, -heteroaryl-NR7—, or -heterocyclyl-NR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (BC), when A is a fused or a bridged bicyclic ring; C is a 4- to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl; X is a bond, —CH2—, or —C(CH3)2—; and Y is a bond; then W is not a bond.


In one embodiment of the compounds of formula (BC), when A is phenyl, C is a fused or a bridged bicyclic ring; X is a bond, —CH2—, or —C(CH3)2—; and Y is a bond; then W is not a bond.


In one embodiment of the compounds of formula (BC), A is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl. In some embodiments, A is phenyl or pyridyl.


In one embodiment of the compounds of formula (BC), B is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl. In some embodiments, B is phenyl or pyridyl.


In one embodiment of the compounds of formula (ABC), C is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl.


In one embodiment of the compounds of formula (ABC), C is a monocyclic carbocycle, a spiral bicyclic carbocycle, a monocyclic heterocycle, or a spiral bicyclic heterocycle.


In one embodiment of the compounds of formula (BC), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 5- or 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 5- or 6-membered heterocyclyl;


In one embodiment of the compounds of formula (BC), W is heteroaryl, heterocyclyl, -heteroaryl-NR7—, or -heterocyclyl-NR7—.


In one embodiment of the compounds of formula (BC), Y is a bond and W is a bond, heteroaryl, heterocyclyl, -heteroaryl-NH—*, or -heterocyclyl-NH—*, wherein heteroaryl or heterocyclyl of W is optionally substituted and wherein * indicates connection to ring C.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (B):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a phenyl or a pyridyl ring;
      • C is a monocyclic carbocycle, a spiral bicyclic carbocycle, a monocyclic heterocycle, or a spiral bicyclic heterocycle;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (B-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a phenyl or a pyridyl ring;
      • C is a monocyclic carbocycle, a spiral bicyclic carbocycle, a bridged bicyclic carbocycle, a monocyclic heterocycle, or a spiral bicyclic heterocycle;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In some embodiments of the compounds of formula (B-I), when C is a bridged bicyclic carbocycle, then —Z—V-L is not —OCH2CH2Cl or —OCH3.


In some embodiments of the compounds of formula (B-I), when —Z—V-L is —OCH2CH2Cl or —OCH3, C is a monocyclic carbocycle, a spiral bicyclic carbocycle, a monocyclic heterocycle, or a spiral bicyclic heterocycle.


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), A and B are phenyl.


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), Y is a bond or —O—.


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), W is a bond, —CH2—, or —CH2CH2—.


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), C is a 4-6 membered monocyclic carbocycle, a 4-6 membered monocyclic heterocycle, or a 7-10 membered spiral bicyclic heterocycle. In some embodiments, C is a bridged bicyclic carbocycle. In some embodiments, C is




embedded image


In some embodiments, C is




embedded image


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl.


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), Z is —O—; V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; and L is hydrogen or halogen. In some embodiments, —Z—V-L is




embedded image


In some embodiments, —Z—V-L is




embedded image


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), Z—V-L is —O—CH2CH2Cl.


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), Z is a bond; V is —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O; and L is hydrogen. In some embodiments, R8a′ and R9a′ taken together form an optionally substituted morpholine. In some embodiments, —Z—V-L is




embedded image


In some embodiments of the compounds of formula (BC), formula (B), or formula (B-I), X and Y are connected at para positions of A and X and Z are connected at para positions of B.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (C):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond;
      • W is a bond, heteroaryl, heterocyclyl, -heteroaryl-NH—*, or -heterocyclyl-NH—*, wherein heteroaryl or heterocyclyl of W is optionally substituted and wherein * indicates connection to ring C;
      • Z is a bond or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, oxo, —CN, or —CF3;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5;
      • wherein when A is a fused or a bridged bicyclic ring; C is a 4- to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl; and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond; and
      • wherein when A is phenyl, C is a fused or a bridged bicyclic ring; and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (C-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 4 to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, —O—, or —NR7—;
      • Y is a bond;
      • W is a bond, heteroaryl, heterocyclyl, -heteroaryl-NH—*, or -heterocyclyl-NH—*, wherein heteroaryl or heterocyclyl of W is optionally substituted and wherein * indicates connection to ring C;
      • Z is a bond or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, oxo, —CN, or —CF3;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), optionally substituted 3- to 7-membered carbocycle, or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 haloalkyl, C3-C6 cycloalkyl, or phenyl;
      • m is 0, 1, 2, or 3;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In some embodiments of the compounds of formula (C-I), when A is a fused or a bridged bicyclic ring; C is a 4- to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl; and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond.


In some embodiments of the compounds of formula (C-I), when A is phenyl, and C is a fused bicyclic ring selected from




embedded image


and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond.


In some embodiments of the compounds of formula (C-I), when A is phenyl, C is a fused bicyclic ring; and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond. In some embodiments of the compounds of formula (C-I), when A is phenyl, C is a fused or a bridged bicyclic ring; and X is a bond, —CH2—, or —C(CH3)2—, then W is not a bond.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), A and B are each independently a 6-membered monocyclic ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl. In some embodiments, A and B are phenyl. In some embodiments, A is phenyl and B is pyridyl. In some embodiments, B is phenyl and A is pyridyl.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), W is a bond, 9-10 membered bicyclic heteroaryl, 5-6 membered monocyclic heteroaryl, 5-6 membered monocyclic heterocyclyl, -(5-6 membered monocyclic heteroaryl)-NH—*, or -(5-6 membered monocyclic heterocyclyl)-NH—*, wherein * indicates connection to ring C. In some embodiments, W is a bond, pyrazole, pyridine, pyrimidine, pyrazine, pyridazine, 1,2,3-triazole, piperazine, piperidine, -quinoxaline-NH—*, -pyrazole-NH—*, -pyridine-NH—*, or -pyrimidine-NH—*.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), W is a bond, 5-6 membered monocyclic heteroaryl, 5-6 membered monocyclic heterocyclyl, -(5-6 membered monocyclic heteroaryl)-NH—*, or -(5-6 membered monocyclic heterocyclyl)-NH—*, wherein * indicates connection to ring C. In some embodiments, W is a bond, pyrazole, pyridine, pyrimidine, pyrazine, pyridazine, 1,2,3-triazole, piperazine, piperidine, -pyrazole-NH—*, -pyridine-NH—*, or -pyrimidine-NH—*. In some embodiments, W is a bond, pyrazole, pyridine, pyrimidine, pyrazine, pyridazine, 1,2,3-triazole, piperazine, piperidine, -pyridine-NH—*, or -pyrimidine-NH—*. In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), C is a 4-10 membered monocyclic carbocyclyl, a bridged 5-10 membered bicyclic carbocyclyl. a 4-6 membered monocyclic heterocycle, a 5-10 membered bicyclic heterocycle, a 10-15 membered tricyclic ring, a 5-10 membered monocyclic heteroaryl, or a fused 8-10 membered bicyclic heteroaryl. In some embodiments, C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, 5,5-fused heteroaryl or heterocycle, or a 7-10 membered spiral bicyclic heterocycle. In some embodiments, C is




embedded image


embedded image


In some embodiments, C is




embedded image


embedded image


In some embodiments C is




embedded image


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 5- or 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl). In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 5- or 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl). In some embodiments, at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl. In some embodiments, at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), at least one R3 is oxo; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl). In some embodiments, at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), at least one R3 is an optionally substituted 3- to 7-membered carbocycle, or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl. In some embodiments, at least one R3 is an optionally substituted 4- to 6-membered heterocyclyl containing one or two heteroatoms selected from N, O, or S. In some embodiments, at least one R3 is an optionally substituted




embedded image


In some embodiments, at least one R3 is




embedded image


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), Z is —O—; V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; and L is hydrogen or halogen. In some embodiments, —Z—V-L is




embedded image


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), —Z—V-L is —O—CH2CH2Cl. In some embodiments, —Z—V-L is —O—CH2CH2Cl or —OCH3.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), Z is a bond; V is —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O; and L is hydrogen. In some embodiments, R8a′ and R9a′ taken together form an optionally substituted morpholine. In some embodiments, Z is a bond; V is a bond, and L is —NR11R12. In some embodiments, R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O. In some embodiments, —Z—V-L is




embedded image


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), R7 is hydrogen, —CH3, —CH2CH3, —CH2CF3, t-butyl, or cyclopropyl. In some embodiments, R7 is hydrogen, —CH3, -CD3, —CH2CH3, —CH2CF3, t-butyl, or cyclopropyl.


In some embodiments of the compounds of formula (BC), formula (C), or formula (C-I), A and B are each independently a 6-membered monocyclic ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl; X and Y are connected at 1 and 4 positions of A; and X and Z are connected at 1 and 4 positions of B.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (D):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is a carbocycle, heterocycle, —CHCH2, —CHF2, or —CH3;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl; and
      • n3 is 1, 2, or 3.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (D-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —(CR8aR9a)m-(optionally substituted carbocycle), —(CR8aR9a)m1-(optionally substituted aryl), —(CR8aR9a)m-(optionally substituted heterocycle), —(CR8aR9a)m-(optionally substituted heteroaryl), —CHCH2, —CH1F2, —CH3, —CH2CH2CH3, —CH2CH2NHSO2CH3, or —CH2CH2CH2OC(O)NH-(optionally substituted aryl);
      • Z is a bond and —V-L is —(CR8aR9a)m—NR11R12; or
      • Z is a bond and —V-L is —CH2CH2OH, —CH2CH2Cl, or —CHCH2;
      • R2 is Cl or —CN;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • n3 is 1, 2, or 3;
      • each m is independently 0, 1, 2, or 3; and
      • each ml is independently 1, 2, or 3.





In some embodiments of the compounds of formula (D-I), when R2 is —CN, then —Z—V-L is not




embedded image


In some embodiments of the compounds of formula (D) or formula (D-I), —V-L is 3-6 membered monocyclic carbocycle or 3-6 membered monocyclic heterocycle.


In some embodiments of the compounds of formula (D-I), Z is —O— and —V-L is —(CR8aR9a)m-(optionally substituted carbocycle), —(CR8aR9a)m1-(optionally substituted aryl), —(CR8aR9a)m-(optionally substituted heterocycle), —(CR8aR9a)m-(optionally substituted heteroaryl), —CHCH2, —CHF2, —CH3, —CH2CH2CH3, —CH2CH2NHSO2CH3, or -CH2CH2CH2OC(O)NH-(optionally substituted aryl). In some embodiments, Z is —O— and —V-L is —(CR8aR9a)m-(optionally substituted 3-6 membered carbocycle), —(CR8aR9a)m1-(optionally substituted phenyl), —(CR8aR9a)m-(optionally substituted 3-6 membered heterocycle), —(CR8aR9a)m-(optionally substituted 5-6 membered heteroaryl), —CHCH2, —CHF2, —CH3, —CH2CH2CH3, —CH2CH2NHSO2CH3, or —CH2CH2CH2OC(O)NH-(optionally substituted phenyl).


In some embodiments of the compounds of formula (D) or formula (D-I), —Z—V-L is —O—CH3, —O—CHF2, —O—CHCH2, —O-cyclopropyl, —O-cyclobutyl, or —O-oxetanyl.


In some embodiments of the compounds of formula (D-I), —Z—V-L is —O—CH3, —O—CHF2, —OCHCH2, —OCH2CH2CH3, —OCH2CH2NHSO2CH3, —O—(CH2)m-(optionally substituted cyclopropyl), —O—(CH2)m-(optionally substituted cyclobutyl), —O—(CH2)m-(optionally substituted phenyl), —O—(CH2)m-(optionally substituted oxetanyl), —O—(CH2)m-(optionally substituted azetidinyl), —O—(CH2)m-(optionally substituted piperidinyl), —O—(CH2)m-(optionally substituted piperazinyl), —O—(CH2)m-(optionally substituted pyridyl), —O—(CH2)m-(optionally substituted pyrazinyl), or —OCH2CH2CH2OC(O)NH-(optionally substituted phenyl). In some embodiments of formula (D-I), —Z—V-L is —O—CH3, —O—CHF2, —CHCH2, —O—(CH2)m-(optionally substituted cyclopropyl), —O—(CH2)m-(optionally substituted cyclobutyl), —O—(CH2)m-(optionally substituted phenyl), —O—(CH2)m-(optionally substituted oxetanyl), —O—(CH2)m-(optionally substituted azetidinyl), —O—(CH2)m-(optionally substituted piperidinyl), —O—(CH2)m-(optionally substituted piperazinyl), —O—(CH2)m-(optionally substituted pyridyl), —O—(CH2)m-(optionally substituted pyrazinyl), or —OCH2CH2CH2OC(O)NH-(optionally substituted phenyl); wherein m is 0 or 1. some embodiments of formula (D-I), —Z—V-L is —O—(CH2)m-(optionally substituted piperidinyl), —O—(CH2)m-(optionally substituted piperidine-2,6,-dione), or —O—(CH2)m-(optionally substituted piperazinyl), wherein m is 2.


In some embodiments of the compounds of formula (D-I), —Z—V-L is —O—CH3, —O—CHF2, —OCHCH2, —OCH2CH2CH3, —OCH2CH2NHSO2CH3, —O-cyclopropyl, —O—(CH2)-cyclopropyl, —O-cyclobutyl, —O—(CH2)-cyclobutyl,




embedded image


or —OCH2CH2CH2OC(O)NH-phenyl. In some embodiments, —Z—V-L is




embedded image


In some embodiments of the compounds of formula (D-I), Z is a bond and —V-L is —(CR8aR9a)m—NR11R12. In some embodiments, Z is a bond; —V-L is —(CR8aR9a)m—NR11R12; and R11 and R12 taken together form an optionally substituted 4-8 membered heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O. In some embodiments, Z is a bond; —V-L is —(CR8aR9a)m-(optionally substituted piperidine), —(CR8aR9a)m-(optionally substituted 6-azaspiro[2.5]octane), —(CR8aR9a)m-(optionally substituted 4-oxa-7-azaspiro[2.5]octane), —(CR8aR9a)m-(optionally substituted piperazine), —(CR8aR9a)m-(optionally substituted morpholine), —(CR8aR9a)m-(optionally substituted 6-oxa-3-azabicyclo[3.1.1]heptane), or —(CR8aR9a)m-(optionally substituted 2-oxa-5-azabicyclo[2.2.1]heptane). In some embodiments, Z is a bond and —V-L is




embedded image


In some embodiments of the compounds of formula (D-I), Z is a bond and —V-L is —CH2CH2OH, —CH2CH2Cl, or —CHCH2.


In some embodiments of the compounds of formula (D-I), substituents on —V-L is halogen, hydroxyl, C1-C4 alkyl, C1-C4 haloalkyl, or C1-C4 alkoxy.


In some embodiments of the compounds of formula (D) or formula (D-I), X is —CH2— or —C(CH3)2. In some embodiments, X is —NR7. In some embodiments, R7 is hydrogen, —CH3, —CD3, —CH2CH3, —CH2CHF2, —CH2CF3, t-butyl, or cyclopropyl.


In some embodiments of the compounds of formula (D) or formula (D-I), C is




embedded image


In some embodiments of the compounds of formula (D) or formula (D-I), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl. In some embodiments of the compounds of formula (D) or formula (D-I), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CHCH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3 or an optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl.


In some embodiments of the compounds of formula (D) or formula (D-I), at least one R3 is an optionally substituted 4- to 6-membered heterocyclyl or heteroaryl. In some embodiments, at least one R3 is an optionally substituted 4- to 6-membered heterocyclyl containing one or two heteroatoms selected from N, O, or S. In some embodiments, at least one R3 is an optionally substituted




embedded image


In some embodiments, at least one R3 is




embedded image


In one embodiment, the present disclosure provides compounds comprising the structure of formula (E):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is a bond —NH—, or —O—; and
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4-6 membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1—C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4-6 membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl; and
      • n3 is 1, 2, or 3.





In one embodiment, the present disclosure provides compounds comprising the structure of formula (E-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is a bond —NH—, or —O—; and
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4-6 membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4-6 membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl; and
      • n3 is 1, 2, or 3.





In some embodiments of the compounds of formula (E) or formula (E-I), X is —CH2— or —C(CH3)2.


In some embodiments of the compounds of formula (E) or formula (E-I), C is




embedded image


In some embodiments of the compounds of formula (E) or formula (E-I), at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, or optionally substituted 4-6 membered heterocyclyl containing at least one N; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl. In some embodiments, at least one R3 is —CH2NHSO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —SO2CH3, or optionally substituted 4-6 membered heterocyclyl containing at least one N; and the other R3 is, if present, halogen, —CN, —CF3, or C1-C3 alkyl. In some embodiments, at least one R3 is —CH2NHSO2CH3 or




embedded image


In some embodiments, at least one R3 is —N(CH3)SO2CH3, —CH2NHSO2CH3 or




embedded image


In one embodiment, the present disclosure provides compounds comprising the structure of formula (F):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently phenyl or pyridyl;
      • C is -pyrimidyl-NHSO2-(optionally substituted heteroaryl) or -pyrimidyl-(optionally substituted heteroaryl)-R3;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —(CR8aR9a)m(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), or —NHCOCF3;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O; and
      • m is 0, 1, 2, or 3.





In some embodiments of the compounds of formula (F), A and B are phenyl.


In some embodiments of the compounds of formula (F), Z is —O—; V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)—; R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; and L is hydrogen or halogen.


In some embodiments of the compounds of formula (F), —Z—V-L is —O—CH2CH2Cl.


In some embodiments of the compounds of formula (F), Y is a bond or —O—.


In some embodiments of the compounds of formula (F), W is a bond, —CH2—, or —CH2CH2—.


In some embodiments of the compounds of formula (F), C is -pyrimidyl-NHSO2-(optionally substituted 5-6 membered heteroaryl). In some embodiments, C is -pyrimidyl-NHSO2-(optionally substituted thiophene), -pyrimidyl-NHSO2-(optionally substituted furan), -pyrimidyl-NHSO2-(optionally substituted pyrazole), or -pyrimidyl-NHSO2-(optionally substituted pyridine). In some embodiments, C is -pyrimidyl-(optionally substituted heteroaryl)-SO2NH2. In some embodiments, C is -pyrimidyl-pyrazole-SO2NH2, -pyrimidyl-imidazole-SO2NH2, or -pyrimidyl-triazole-SO2NH2.


In some embodiments of the compounds of formula (F), X and Y are connected at para positions of A; and X and Z are connected at para positions of B


In one embodiment, the present disclosure provides compounds comprising the structure of formula (G):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1;
      • R1 is each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1, 2, or 3;
      • wherein: a) n1 is 1; and/or b) n3 is 2, wherein the second R3 is —NH2, optionally substituted piperidine, optionally substituted piperazine.





In some embodiments of the compounds of formula (G), X is —CH2— or —C(CH3)2.


In some embodiments of the compounds of formula (G), n1 is 1 and R1 is optionally substituted —(C1-C6 alkyl)-NR14R15.


In some embodiments of the compounds of formula (G), n1 is 1 and R1 is —CH2OH, —CH2NH2,




embedded image


In some embodiments of the compounds of formula (G), n3 is 2 and the second R3 is —NH2,




embedded image


In one embodiment, the present disclosure provides compounds comprising the structure of formula (H):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is phenyl or pyridyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1;
      • R1 and R2 are each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • at least one R3 is halogen and at least one other R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and if a third R3 is present, then it is halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n2 is 0, 1, or 2; and
      • n3 is 2 or 3.





In some embodiments of the compounds of formula (H),




embedded image


is not




embedded image


In some embodiments of the compounds of formula (H), at least one R3 is fluorine and at least one other R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (J):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • B is 6-membered heteroaryl or 6-membered heterocyclyl;
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2, —O—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1; or
      • Z is a bond and —V-L is —CH2CH2CH2C1;
      • R1 and R2 are each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14 optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • when B is a 6-membered heterocyclyl, R2 can also be an oxo;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3, if present, is halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, or C1-C4 haloalkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n2 is 0, 1, or 2; and
      • n3 is 1, 2 or 3.







embedded image


In some embodiments of the compounds of formula (J), B is N Z L, R2 is not Cl.


In some embodiments of the compounds of formula (J), B is 6-membered N-heteroaryl or 6-membered N-heterocyclyl. In some embodiments, B is a ring selected from pyridine, pyrimidine, pyridazine, pyrazine, 1,6-dihydropyrimidine, or pyrimidin-4(3H)-one.


In some embodiments of the compounds of formula (H) or formula (J), X is —C(CH3)2, or —N(CH3)—. In some embodiments, X is —C(CH3)2.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (K):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a —CH2—, —C(O)—, or —NR7—;
      • Z is —O— and —V-L is —CH2CH2C1;
      • R1 and R2 are each independently halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14 optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, halogen, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen, C1-C4 alkyl, C1-C4 haloalkyl, or C1-C4 deuterated alkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • n1 is 0, 1, or 2;
      • n2 is 0, 1, or 2; and
      • n3 is 1, 2, or 3.





In some embodiments of the compounds of formula (K), X is a —CH2—, —C(O)—, or —N(CH3)—. In some embodiments of the compounds of formula (K), X is a —CH2—, —C(O)—, —N(CH3)—, or —NCH2CF3. In some embodiments, X is —NR7. In some embodiments, R7 is hydrogen, —CH3, -CD3, —CH2CH3, —CH2CHF2, —CH2CF3, t-butyl, or cyclopropyl.


In some embodiments of the compounds of formula (H), formula (J), or formula (K), at least one other R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —CH(CH3)NHSO2CH3, —CH2CH2NHSO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3.


In some embodiments of the compounds of formula (H), formula (J), or formula (K), R1 and R2 are each independently halogen, —CN, or —CF3.


In one embodiment, the present disclosure provides compounds comprising the structure of formula (I-B):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a 3- to 15-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —(CR5R6)t—, —O—, or —NR7;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, —NR7—, or —N(COCH3)—;
      • W is a bond, heteroaryl, heterocyclyl, heteroaryl-NR7—, heterocyclyl-NR7—, —(CR8aR9a)m—, —C(═O)—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is a bond, —(CR8aR9a)m—, or phenylene;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, -OR10; —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14C00R16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R10 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
      • R11 and R12 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • each m is independently 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, 2, 3, 4 or 5; and

    • t is 0, 1 or 2.
      • In a specific embodiment of formula (I), (I-A), (I-B), (ABC), (BC), (A), (B), (B-I), (C), or (C-I), C is a heteroaryl ring.





In one embodiment, the present disclosure provides compounds of formula (II):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a 4- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a heteroaryl ring;
      • X is a bond, —(CR5R6)t—, —O—, or —NR7;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, —NR7—, or —N(COCH3)—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —(CR8aR9a)m—, —C(═O)—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, -5-, —NH—, —NCH3—, or —N(COCH3)—;
      • V is —CH2— or phenylene and L is halogen, —NH2, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen or —NH2;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • each m is independently 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1, 2, 3, 4 or 5; and
      • t is 0, 1 or 2.





In one embodiment of the compounds of formula (I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), or (C-I), C is




embedded image


In one embodiment, the present disclosure provides compounds of formula (A′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a phenyl;
      • C is a heteroaryl ring;
      • X is a bond, —(CR5R6)t—, or —O—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, —NR7—, or —N(COCH3)—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —(CR8aR9a)m—, —C(═O)—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is —CH2— and L is halogen, —NH2, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen or —NH2;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14C00R16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • each m is independently 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1, 2, 3, 4 or 5; and
      • t is 0, 1 or 2.





In one embodiment, the present disclosure provides compounds of formula (A′-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a fused or a bridged bicyclic 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a phenyl;
      • C is a heteroaryl ring;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond or —O—;
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and
      • the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (A′), (A′-I), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • X is bond or —C(CH3)2—;
      • Y and Z are each —O—;
      • V is —CH2— or —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment of the compounds of formula (A′), (A′-I), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • W is —CH2— or —C(CH3)H—;
    • V is —CH2CH2—; and
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3.


In one embodiment, the present disclosure provides compounds of formula (A′-II):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a fused or a bridged bicyclic 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a phenyl or a 6-membered heteroaryl or heterocyclyl;
      • C is a 3- to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond or —O—;
      • V is a bond, —CH2(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, oxo, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment, the present disclosure provides compounds of formula (A′-III):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a fused or a bridged bicyclic 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • B is a phenyl or a 6-membered heteroaryl or heterocyclyl;
      • C is a 3 to 10-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond, —CH2—, —C(CH3)H—, —NH—, or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2—, —CH2CH2—, —C(CH3)H—, —NR7—, —N(R7)CO—, or —CONR7—;
      • Z is a bond or —O—;
      • V is a bond, —CH2(CR8a′R9a′)—, —CR8a═CR9a—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, —SO2R17, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, halogen, oxo, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • R17 is hydrogen or C1-C3 alkyl;
      • m is 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (I), (I-A), (I-B), (II), (A′), (I-A), (ABC), (BC), (A), (C), or (C-I), A is a fused or a bridged bicyclic ring.


In one embodiment of the compounds of formula (I), (II), (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), A is a 6,6-fused ring, 6,5-fused ring, or 5,6-fused ring, each optionally substituted with one or two R1.


In one embodiment of the compounds of formula (I), (II), (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), A is




embedded image


optionally substituted with one or two R1, wherein A2 is a 5- or a 6-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl ring. In one embodiment, A2 is a 6-membered aryl, carbocyclyl, heteroaryl or heterocyclyl ring. In one embodiment, A2 is a 6-membered aryl or heteroaryl ring. In one embodiment, A2 is a 5-membered carbocyclyl, heteroaryl or heterocyclyl ring. In one embodiment, A2 is a 5-membered heterocyclyl or heteroaryl ring. In one embodiment, heterocyclyl ring contains one, two, or three heteroatoms selected from N, S, or O. In one embodiment, heteroaryl contains one, two, or three heteroatoms selected from N, S, or O. In one embodiment, heteroaryl contains one or two nitrogen atoms as ring atoms.


In one embodiment of the compounds of formula (I), (II), (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), A is




embedded image


wherein:

    • ring A3 is aromatic;
    • E1, E2, E3, E4, and E5 are each independently, C, CR1 or N;
    • G1, G2, and G3, are each independently, C, CRI, C(R1)2, O, S, N, or NRI;
    • wherein at least two of E1, E2, E3, E4, and E5 is C or CR1 (i.e., maximum 3 of E1, E2, E3, E4, and E5 can be N); and
    • wherein at least two of E1, E2, G1, G2, and G3, is C, CR1 or C(R1)2 (i.e., maximum 3 of E1, E2, G1, G2, and G3 can be heteroatoms).


In one embodiment, ring A3 is phenyl. In one embodiment, ring A3 is a heteroaryl ring. In one embodiment, ring A3 is a pyridine, pyridazine, pyrimidine, pyrazine, 1,2,4-trizaine, or 1,3,5-triazine ring.


In one embodiment, ring A4 is aromatic. In one embodiment, ring A4 is a heteroaryl ring.


In one embodiment, ring A4 is partially aromatic. In one embodiment, ring A4 is a heterocyclyl or a carbocyclyl ring. In one embodiment, the bond between E1-G1, G1-G2, G2-G3, and G3-E2 are each a single bond.


In one embodiment of the compounds of formula (I), (II), (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), A is




embedded image


wherein:

    • ring A3 is aromatic;
    • E1, E2, E3, E4, and E5 are each independently, C, CR1 or N;
    • G1, G2, G3, and G4, are each independently, C, CR1, C(R1)2, O, S, N, or NR1;
    • wherein at least two of E1, E2, E3, E4, and E5 is C or CR1 (i.e., maximum 3 of E1, E2, E3, E4, and E5 can be N); and
    • wherein at least three of E1, E2, G1, G2, G3, and G4, is C, CR1 or C(R1)2 (i.e., maximum 3 of E1, E2, G1, G2, G3, and G4 can be heteroatoms).


In one embodiment, ring A3 is phenyl. In one embodiment, ring A3 is a heteroaryl ring. In one embodiment, ring A3 is a pyridine, pyridazine, pyrimidine, pyrazine, 1,2,4-trizaine, or 1,3,5-triazine ring.


In one embodiment, E1, E2, E3, E4, and E5 is N, C, CH or CR1. In one embodiment, E1 and E2 are C, and E3, E4, and E5 are each independently N, CH or CR1.


In one embodiment, ring A5 is aromatic. In one embodiment, ring A5 is a heteroaryl ring.


In one embodiment, ring A5 is partially aromatic. In one embodiment, ring A5 is a heterocyclyl or a carbocyclyl ring. In one embodiment, the bond between E1-G1, G1-G2, G2-G3, G3-G4 and G3-E2 are each a single bond. In one embodiment, one of the bond between E1-G1, G1-G2, G2-G3, G3-G4 and G3-E2 is a double bond.


In one embodiment, E1 and E2 are each independently N or C, and G1, G2, G3, and G4, are each independently N, NR1, C, CH, CR1, O, or S.


In one embodiment of the compounds of formula (I), (II), (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), A is a ring selected from bicyclo[1.1.1]pentane, 4,5,6,7-tetrahydroindole, indoline, indole, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyridine, indazole, benzo[d]imidazole, benzo[d]isoxazole, benzo[b]thiophene, 1,3-dihydroisobenzofuran, quinazoline, 3,4-dihydrobenzo[b][1,4]oxazine, benzo[d][1,2,3]triazole, naphthalene, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthalene, 2,3-dihydroindene, 2,3-dihydrobenzo[b][1,4]dioxine, isoindoline, or isoindolin-1-one, each ring is optionally substituted with one or two R1. In one embodiment, A is a 5- to 10-membered fused or a bridged bicyclic ring. In one embodiment, A is ring selected from bicyclo[1.1.1]pentane, indoline, indole, indazole, quinazoline, 3,4-dihydro-2H-benzo[b][1,4]oxazine, 1H-benzo[d][1,2,3]triazole, naphthalene, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthalene, 2,3-dihydro-1H-indene, 2,3-dihydrobenzo[b][1,4]dioxine, isoindoline, or isoindolin-1-one, each ring is optionally substituted with one or two R1.


In one embodiment of the compounds of formula (I), (II), (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), A is




embedded image


wherein each ring is optionally substituted with one or two R1. In one embodiment, A is




embedded image


wherein each ring is optionally substituted with one or two R1.


In one embodiment of the compounds of formula (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


embedded image


In one embodiment of the compounds of formula (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • X is bond or —C(CH3)2—;
    • Y and Z are each —O—;
    • V is —CH2— or —CH2CH2—;
    • L is halogen;
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and
    • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment of the compounds of formula (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • W is —CH2— or —C(CH3)H—;
    • V is —CH2CH2—; and
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3.


In one embodiment of the compounds of formula (A′-III), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • Y is —CH2— or —C(CH3)H—; and
    • W is —NH—.


In one embodiment of the compounds of formula (A′-III), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), W is —NH—.


In one embodiment of the compounds of formula (A′-III), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • Y is —CH2— or —C(CH3)H—;
    • W is —NH—;
    • X is bond or —C(CH3)2—;
    • Z is —O—;
    • V is —CH2— or —CH2CH2—;
    • L is halogen; and
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3.


In one embodiment of the compounds of formula (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I):

    • A is a 6,6-fused ring, 6,5-fused ring, or 5,6-fused ring, each optionally substituted with one or two R1;
    • X is bond;
    • Y and Z are each —O—;
    • V is —CH2— or —CH2CH2—;
    • L is halogen;
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and
    • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment of the compounds of formula (A′), (A′-I), (A′-II), (A′-III), (I-A), (ABC), (BC), (A), (C), or (C-I):

    • A is a 6,6-fused ring, 6,5-fused ring, or 5,6-fused ring, each optionally substituted with one or two R1;
    • X is bond;
    • Y and Z are each —O—;
    • W is —CH2— or —C(CH3)H—;
    • V is —CH2CH2—;
    • L is halogen;
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and
    • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment, the present disclosure provides compounds of formula (B′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a phenyl;
      • B is a 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a heteroaryl ring;
      • X is a bond, —(CR5R6)t—, or —O—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, —NR7—, or —N(COCH3)—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —(CR8aR9a)m—, —C(═O)—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, -5-, —NH—, —NCH3—, or —N(COCH3)—;
      • V is absent or —CH2— and L is halogen, —NH2, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen or —NH2;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted —(C1-C6 alkyl)-(C1-C6 alkoxy), optionally substituted —(C1-C6 alkyl)-OH, oxo, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14R15, optionally substituted —(C1-C6 alkyl)-NR14R15, —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR13R14, optionally substituted —(C1-C6 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C6 alkyl)-SO2NR14R15, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —NR14COOR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), —(C1-C6 alkyl)-SO2(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl;
      • each m is independently 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1, 2, 3, 4 or 5; and
      • t is 0, 1 or 2.





In one embodiment, the present disclosure provides compounds of formula (B′-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a phenyl;
      • B is a fused or a bridged bicyclic 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • C is a heteroaryl ring;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond or —O—;
      • V is absent or —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and
      • the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (B′), (B′-I), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • X is bond or —C(CH3)2—;
    • Y is —O—;
    • Z is a bond or —O—;
    • V is bond, —CH2— or —CH2CH2—;
    • L is halogen;
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and
    • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment of the compounds of formula (B), (B′-I), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K).

    • Y and Z are each —O—;
    • W is —CH2— or —C(CH3)H—;
    • V is —CH2CH2—; and
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), B is a fused or a bridged bicyclic ring, optionally substituted with one or two R2.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), B is a 6,6-fused ring, 6,5-fused ring, or 5,6-fused ring, each optionally substituted with one or two R2.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), B is




embedded image


optionally substituted with one or two R2, wherein B2 is a 5- or a 6-membered aryl, carbocyclyl, heteroaryl, or heterocyclyl ring. In one embodiment, B2 is a 6-membered aryl, carbocyclyl, heteroaryl or heterocyclyl ring. In one embodiment, B2 is a 6-membered aryl or heteroaryl ring. In one embodiment, B2 is a 5-membered carbocyclyl, heteroaryl or heterocyclyl ring. In one embodiment, B2 is a 5-membered heterocyclyl or heteroaryl ring. In one embodiment, heterocyclyl ring contains one, two, or three heteroatoms selected from N, S, or O. In one embodiment, heteroaryl contains one, two, or three heteroatoms selected from N, S, or O. In one embodiment, heteroaryl contains one or two nitrogen atoms as ring atoms.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), B is




embedded image


wherein:

    • ring B3 is aromatic;
    • E1, E2, E3, E4, and E5 are each independently, C, CR2 or N;
    • G1, G2, and G3, are each independently, C, CR2, C(R2)2, O, S, N, or NR2;
    • wherein at least two of E1, E2, E3, E4, and E5 is C or CR2 (i.e., maximum 3 of E1, E2, E3, E4, and E5 can be N); and
    • wherein at least two of E1, E2, G1, G2, and G3, is C, CR2 or C(R2)2 (i.e., maximum 3 of E1, E2, G1, G2, and G3 can be heteroatoms).


In one embodiment, ring B3 is phenyl. In one embodiment, ring B3 is a heteroaryl ring. In one embodiment, ring B3 is a pyridine, pyridazine, pyrimidine, pyrazine, 1,2,4-trizaine, or 1,3,5-triazine ring.


In one embodiment, ring B4 is aromatic. In one embodiment, ring B4 is a heteroaryl ring.


In one embodiment, ring B4 is partially aromatic. In one embodiment, ring B4 is a heterocyclyl or a carbocyclyl ring. In one embodiment, the bond between E1-G1, G1-G2, G2-G3, and G3-E2 are each a single bond.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), B is




embedded image


wherein:

    • ring B3 is aromatic;
    • E1, E2, E3, E4, and E5 are each independently, C, CR2 or N;
    • G1, G2, G3, and G4, are each independently, C, CR2, C(R2)2, O, S, N, or NR2;
    • wherein at least two of E1, E2, E3, E4, and E5 is C or CR2 (i.e., maximum 3 of E1, E2, E3, E4, and E5 can be N); and
    • wherein at least three of E1, E2, G1, G2, G3, and G4, is C, CR2 or C(R2)2 (i.e., maximum 3 of E1, E2, G1, G2, G3, and G4 can be heteroatoms).


In one embodiment, ring B3 is phenyl. In one embodiment, ring B3 is a heteroaryl ring. In one embodiment, ring B3 is a pyridine, pyridazine, pyrimidine, pyrazine, 1,2,4-trizaine, or 1,3,5-triazine ring.


In one embodiment, ring B5 is aromatic. In one embodiment, ring B5 is a heteroaryl ring.


In one embodiment, E1, E2, E3, E4, and E5 are each independently N, C, CH or CR2. In one embodiment, E1 and E2 are C, and E3, E4, and E5 are each independently N, CH or CR2.


In one embodiment, ring B5 is partially aromatic. In one embodiment, ring B5 is a heterocyclyl or a carbocyclyl ring. In one embodiment, the bond between E1-G1, G1-G2, G2-G3, G3-G4 and G3-E2 are each a single bond. In one embodiment, one of the bonds between E1-G1, G1-G2, G2-G3, G3-G4 and G3-E2 is a double bond.


In one embodiment, E1 and E2 are each independently N or C, and G1, G2, G3, and G4, are each independently N, NR1, C, CH, CR1, O, or S.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), B is a 5- to 10-membered fused or a bridged bicyclic ring, optionally substituted with one or two R2. In one embodiment, B is an 8- to 10-membered fused or a bridged bicyclic ring, optionally substituted with one or two R2. In one embodiment, B is ring selected from bicyclo[1.1.1]pentane, indoline, indole, indazole, quinazoline, 3,4-dihydrobenzo[b][1,4]oxazine, benzo[d][1,2,3]triazole, naphthalene, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthalene, 2,3-dihydroindene, 2,3-dihydrobenzo[b][1,4]dioxine, isoindoline, or isoindolin-1-one, each ring is optionally substituted with one or two R2. In one embodiment, B is ring selected from bicyclo[1.1.1]pentane, indoline, indole, indazole, quinazoline, 3,4-dihydro-2H-benzo[b][1,4]oxazine, 1H-benzo[d][1,2,3]triazole, naphthalene, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthalene, 2,3-dihydro-1H-indene, 2,3-dihydrobenzo[b][1,4]dioxine, isoindoline, or isoindolin-1-one, each ring is optionally substituted with one or two R2. In one embodiment, B is:




embedded image


wherein each ring is optionally substituted with one or two R2.


In one embodiment of the compounds of formula (B) or (B-I):

    • B is a 6,6-fused ring, 6,5-fused ring, or 5,6-fused ring, each optionally substituted with one or two R2;
    • X is bond;
    • Y is —O—;
    • Z is a bond or —O—;
    • V is bond, —CH2— or —CH2CH2—;
    • L is halogen;
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and
    • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment of the compounds of formula (B′), (B′-I), (I-A), (ABC), (BC), (A), (C), or (C-I):

    • B is a 6,6-fused ring, 6,5-fused ring, or 5,6-fused ring, each optionally substituted with one or two R2;
    • X is bond;
    • Y and Z are each —O—;
    • W is —CH2— or —C(CH3)H—;
    • V is —CH2CH2—;
    • L is halogen;
    • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3; and


R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), C is a pyrimidine, triazine, or thiophene ring.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


In one embodiment, C is




embedded image


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (C), or (C-I), C is a pyrimidine ring. In one embodiment, C is




embedded image


In one embodiment, C is




embedded image


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R1 and R2 are each independently Cl, —CN, or —CF3. In one embodiment, R1 and R2 are each independently Cl or —CN.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), at least one R2 is —CN. In one embodiment, at least one R2 is —Cl. In one embodiment, n2 is at least 2; at least one R2 is —CN; and at least one R2 is —Cl.


In one embodiment of the compounds of formula (I) or (I-A), R3 is not hydrogen.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —NR14SO2R16, wherein R14 and R16 together form a 5 or 6 membered ring including the nitrogen and sulfur atoms.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (ABC), (BC), (A), (C), or (C-I), R3 is —NR14SO2R16, wherein R16 is optionally substituted C1-C6 alkyl. In one embodiment, R3 is —NR14SO2R16, wherein R16 is C1-C6 alkyl optionally substituted with one or more groups selected from halogen, —CN, —CF3, —OH, C1-C3 alkyl, C1-C3 alkoxy, —NH2, —NH(C1-C3 alkyl), —N(C1-C3 alkyl)2, —SCH3. In one embodiment, R3 is —NR14SO2R16, wherein R16 is C1-C3 alkyl substituted with —NH2.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is F, Cl, Br, I, —CN, —CF3, —OH, methyl, methoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —NHCO(C1-C3 alkyl).


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3. In one embodiment, if more than one R3 is present, the other R3 is —CN, C1-C3 alkyl, C1-C3 alkoxy, —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3. In one embodiment, if more than one R3 is present, the other R3 is —CN, C1-C3 alkyl, C1-C3 alkoxy, —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3. In one embodiment, R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3 and the other R3 is, if present (e.g., when n3 is 2, 3, or 5), —CN, C1-C3 alkyl, C1-C3 alkoxy, —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, or —NH2. In one embodiment, at least one R3 is —NHSO2CH3.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), X is a bond, —CH2—, —C(CH3)2—, or —O—. In one embodiment, X is a bond. In some embodiments, X is —O—.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), X is —NR7. In one embodiment of the compounds of formula (B-I), (C-I), (D-I), or (K), X is —NR7. In some embodiments, R7 is hydrogen, —CH3, -CD3, —CH2CH3, —CH2CHF2, —CH2CF3, t-butyl, or cyclopropyl.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), Y and Z are each —O—.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), or (A), (B), (B-I), (C), or (C-I):

    • C is a 5- or 6-membered heteroaryl ring;
    • at least one R2 is CN; and
    • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3.


In embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K):

    • C is a pyrimidine ring;
    • at least one R2 is CN; and
    • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K),

    • C is a pyrimidine ring;
    • at least one R2 is CN; and
    • at least one R3 is —NHSO2CH3.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —V-L is —CH2CH2Cl, —CH2CH2CH2Cl, —CH2CH2NH2, or —CH2CH2CH2NH2. In one embodiment, —V-L is —CH2CH2Cl or —CH2CH2CH2Cl.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —V-L is —CH2CH2Cl.


In one embodiment of the compounds of formula (I), (A′-II), (A′-III), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), or (F), —Z—V-L comprises a carbocycle. In some embodiments, —Z—V-L comprises a cyclopropyl or a cyclobutyl. In some embodiments, —Z—V-L is —O-cyclopropyl or —O-cyclobutyl.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —Y—W— is a bond, —OCH2—, —OCH2CH2—, —OCH(CH3)—, —NH—, —NHCH2—, —NHC(═O)—, or —C(═O)NH—.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —Y—W— is —OCH2— or —OCH(CH3)—.


In one embodiment of the compounds of formula (I), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R5 and R6 are each independently hydrogen, halogen, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, or C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl. In some embodiments, R5 and R6 are each independently hydrogen, halogen, —OH, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl. In one embodiment, R5 and R6 are hydrogen, halogen, —OH, or C1-C3 alkyl. In one embodiment, R5 and R6 are each independently hydrogen, F, —OH, or C1-C3 alkyl. In one embodiment, R5 and R6 are each independently, hydrogen, F, —OH, or methyl. In one embodiment, R5 and R6 are each H. In one embodiment, R5 and R6 are each methyl. In one embodiment of the compounds of formula (I), (II), (A), (B), or (B-I), R5 and R6 are each H or methyl.


In one embodiment of the compounds of formula (I), (B′), (I-A), (I-B), (II), (ABC), (BC), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), X is —NR7.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R7 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments, R7 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments, R7 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl. In some embodiments, R7 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl. In some embodiments, R7 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl. In some embodiments, R7 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl. In some embodiments, R7 is hydrogen or C1-C6 alkyl. In some embodiments, R7 is hydrogen or C1-C4 alkyl. In some embodiments of the compounds of formula (I), (II), (A), (B), or (B-I), R7 is hydrogen or C1-C3 alkyl.


In some embodiment of the compounds of formula (B′), (I-A), (B), (C-I), (D-I), (H), or (K), R7 is C1-C4 haloalkyl. In some embodiments, R7 is hydrogen, —CH3, -CD3, —CH2CH3, —CH2CHF2, —CH2CF3, t-butyl, or cyclopropyl.


In some embodiment of the compounds of formula (B′), (I-A), (ABC), (BC), (B), (B-I), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R7 is C1-C4 haloalkyl. In some embodiments, R7 is hydrogen, —CH3, —CH2CH3, —CH2CHF2, —CH2CF3, t-butyl, or cyclopropyl.


In one embodiment of the compounds of formula (I), (A′), (A′-II), (A′-III), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R8a and R9a are each independently hydrogen, halogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted C1-C6 alkoxy, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —NR14COR16, optionally substituted —(C1-C6 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C6 alkyl)-CONR14R15, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or R8a and R9a taken together form an optionally substituted carbocyclyl or optionally substituted heterocyclyl. In some embodiments, R8a and R9a are each independently hydrogen, —OH, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15; or R8a and R9a taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl. In one embodiment, R8a and R9a on the same carbon atom is taken together to form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl. In one embodiment, R8a and R9a are each independently hydrogen, halogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or —(C1-C3 alkyl)-CONR14R15. In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R8a and R9a are not —OH. In one embodiment, R8a and R9a are not —OH.


In one embodiment of the compounds of formula (I) or (I-A), R7 and R8a taken together form an optionally substituted heterocyclyl. In one embodiment, R7 and R8a taken together form an optionally substituted 3- to 7-membered heterocycle.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R13 and R14 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In one embodiment, R13 and R14 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, or optionally substituted C2-C6 alkynyl. In some embodiments R13 and R14 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl. In some embodiments R13 and R14 are each independently hydrogen or C1-C3 alkyl.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R15 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In one embodiment, R15 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, or optionally substituted C2-C6 alkynyl. In some embodiments, R15 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl. In some embodiments, R15 is hydrogen or C1-C3 alkyl.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R14 and R15 taken together form an optionally substituted heterocyclyl. In one embodiment, R14 and R15 taken together form an optionally substituted 3- to 7-membered heterocyclyl. In one embodiment, R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl. In other embodiments, R14 and R15 taken together form 3- to 7-membered heterocyclyl.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R16 is hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments, R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments, R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl. In some embodiments, R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl. In some embodiments, R16 is hydrogen or C1-C3 alkyl.


In one embodiment of the compounds of formula (I), (A′), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), m is 1 or 2.


In one embodiment of the compounds of formula (I), (A′), (A′-II), (A′-III), (B′), (I-A), (I-B), (II), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), t is 1 or 2. In one embodiment, t is 1.


In one embodiment, the present disclosure provides compounds of formula (C′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a phenyl ring;
      • C is a heteroaryl ring;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond or —O—;
      • V is a phenylene;
      • L is halogen, —CHCl2, —CCl3, or —CF3; or
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment, the present disclosure provides compounds of formula (E′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 5- or 6-membered heterocyclyl or heteroaryl;
      • B is phenyl;
      • C is a heteroaryl ring;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond or —O—;
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (E′), C is a pyrimidine ring.


In one embodiment, the present disclosure provides compounds of formula (E′-I):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 5- or 6-membered heterocyclyl or heteroaryl;
      • B is phenyl;
      • C is a 6- to 12-membered heteroaryl ring;
      • X is a bond, —CH2—, —CH(CH3)—, —C(CH3)2—, or —O—;
      • Y is a bond, —O—, or —NH—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond —NH—, or —O—;
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, or 4;
      • wherein A is not thiophene.





In one embodiment, the present disclosure provides compounds of formula (E′-I), (I-A), (ABC), (BC), (A), (C), or (C-I), C is a fused bicyclic heteroaryl ring. In one embodiment, C is any fused bicyclic heteroaryl as described for formula (G′).


In one embodiment, the present disclosure provides compounds of formula (E′-I), (I-A), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


In one embodiment of the compounds of formula (E′), (E′-I), (I-A), (ABC), (BC), (A), (C), or (C-I), A is piperidine, pyrazole, or oxadiazole ring, wherein each ring is optionally substituted with one or two R1. In one embodiment, A is piperidine or oxadiazole ring, wherein each ring is optionally substituted with one or two R1.


In one embodiment, the present disclosure provides compounds of formula (F′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is a 5- or 6-membered heterocyclyl or heteroaryl;
      • B is phenyl;
      • C is a heteroaryl ring;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond or —O—;
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and
      • the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —NHCOO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment, the present disclosure provides compounds of formula (G′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a phenyl ring;
      • C is a fused or a bridged bicyclic 5- to 15-membered ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond —NH—, or —O—;
      • V is a bond, —CH2(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, or —NH(C1-C3 alkyl); and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • R7 is hydrogen or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5;







embedded image


In one embodiment of the compounds of formula (G′), (I-A), (ABC), (BC), (A), (C), or (C-I), C is a bridged bicyclic carbocycle, 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle. In one embodiment, C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle.


In one embodiment of the compounds of formula (G′), (I-A), (ABC) BC), (A), (C), or (C-I), C is




embedded image


As used herein




embedded image


depicts where R3 substituent(s) can be on the phenyl portion of the fused ring, on the pyrimidine portion of the fused ring, and/or on both the phenyl portion and the pyrimidine portion of the fused ring. This type of depiction of substituents on a multi-cyclic ring applies throughout this disclosure.


In one embodiment of the compounds of formula (G′), (I-A), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


wherein:

    • ring C3 and C5 is aromatic;
    • E1, E2, E3, E4, E5, and E6 are each independently, C, CH, CR3 or N;
    • G1, G2, and G3, are each independently, CH, CH2, CR3, C(R3)2, O, S, N, NH, or NR3;
    • wherein at least two of E1, E2, E3, E4, and E5 in ring C3 is C, CH, or CR3 (i.e., maximum 3 of E1, E2, E3, E4, and E5 is N);
    • wherein at least three of E1, E2, E3, E4, E5, and E6 in ring C5 is C, CH, or CR3 (i.e., maximum 3 of E1, E2, E3, E4, E5, and E6 is N);
    • wherein at least two of E1, E2, G1, G2, and G3, is CH, CH2, CR3, C(R3)2 (i.e., maximum 3 of E1, E2, G1, G2, and G3 can be heteroatoms).


In one embodiment of the compounds of formula (G′), (I-A), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


wherein:

    • ring C3 and C5 is aromatic;
    • E1, E2, E3, E4, E5 and E6 are each independently, C, CH, CR3 or N;
    • G1, G2, G3, and G4, are each independently, CH, CH2, CR3, C(R3)2, O, S, N, NH, or NR3;
    • wherein at least two of E1, E2, E3, E4, and E5 in ring C3 is C, CH, or CR3;
    • wherein at least three of E1, E2, E3, E4, E5 and E6 in ring C7 is C, CH, or CR3; and
    • wherein at least three of E1, E2, G1, G2, G3, and G4, is CH, CH2, CR3, or C(R3)2.


In one embodiment, ring C3 or C5 is phenyl. In one embodiment, ring C3 or C5 is a heteroaryl ring. In one embodiment, ring C3 or C5 is a pyridine, pyridazine, pyrimidine, pyrazine, 1,2,4-trizaine, or 1,3,5-triazine ring.


In one embodiment, ring C4 is aromatic. In one embodiment, ring C4 is a heteroaryl ring.


In one embodiment, ring C4 is partially aromatic. In one embodiment, ring C4 is a heterocyclyl or a carbocyclyl ring. In one embodiment, the bond between E1-G1, G1-G2, G2-G3, and G3-E2 are each a single bond.


In one embodiment, E1, E2, E3, E4, E5 and E6 are each independently N, C, CH or CR2. In one embodiment, E1 and E2 are C, and E3, E4, and E5 are each independently N, CH or CR3.


In one embodiment, ring C6 is partially aromatic. In one embodiment, ring C6 is a heterocyclyl or a carbocyclyl ring. In one embodiment, the bond between E1-GI, GI-G2, G2-G3, G3-G4 and G3-E2 are each a single bond. In one embodiment, one of the bonds between E1-GI, GI-G2, G2-G3, G3-G4 and G3-E2 is a double bond.


In one embodiment, E1 and E2 are each independently N or C and GI, G2, G3, and G4, are each independently N, NH, NR3, C, CH, CR3, O, or S.


In one embodiment, the present disclosure provides compounds of formula (H′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently a phenyl or a pyridyl ring;
      • C is 5- or 6-membered heteroaryl, 4- to 6-membered heterocyclyl, phenyl, or bicyclic 5- or 6-membered carbocycle;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond, —CH2—, —NH—, or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2—, —CH2CH2—, —C(CH3)H—, —N(R7)CO—, or —CONR7—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —CH2(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently halogen, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 4- to 6-membered heterocyclyl;
      • R7 is hydrogen or C1-C3 alkyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 0, 1, or 2;
      • n1 and n2 are each independently 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5;
      • wherein:
      • a) at least one R3 is optionally substituted 4- to 6-membered heterocyclyl or heteroaryl, provided that 5- or 6-membered heterocyclyl is not







embedded image




    •  when R3 is pyrimidyl; or
      • b) Z is a bond; V is a bond; L is —NR11R12; and R11 and R12, taken together with the nitrogen atom to which it is attached, form an optionally substituted heterocyclic ring.





In one embodiment of the compounds of formula (H′), (I-A), (ABC), (BC), (A), (C), or (C-I), C is a pyrimidine, pyrazine, pyridine, pyrazole, azetidine, phenyl, or bicyclo[1.1.1]pentane.


In one embodiment of the compounds of formula (H′), (I-A), (ABC), (BC), (A), (C), or (C-I), R11 and R12, taken together with the nitrogen atom to which it is attached, form an optionally substituted piperidine or 2-azaspiro[3.3]heptane.


In one embodiment of the compounds of formula (H′), (I-A), (ABC), (BC), (A), (C), or (C-I), at least one R3 is optionally substituted 4- to 6-membered heterocyclyl or heteroaryl containing one or two heteroatoms selected from N and O. In one embodiment, at least one R3 is an optionally substituted group selected from: pyrazolyl, morpholinyl, piperazinyl, piperidinyl, pyrrolidinyl, or azetidinyl. In one embodiment, at least one R3 is substituted with —OH, oxo, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), or —NHCOCF3.


In one embodiment of the compounds of formula (H′), (I-A), (ABC), (BC), (A), (C), or (C-I), Z is —NH— or —O—; V is —(CR8aR9a)m—; and L is halogen or H.


In one embodiment, the present disclosure provides compounds of formula (J′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is pyrimidyl;
      • X is a bond, —O—, —CH2— or —C(CH3)2—;
      • Z is —O— and —V-L is —CH2CH(OH)CH3, —CH2CH(OH)CH2Cl, or —CH2CH2CH2C1; or

    • Z is —NH—, V is —(CR8aR9a)m—, and L is hydrogen or halogen;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • m is 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment, the present disclosure provides compounds of formula (K′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • B is pyridyl;
      • C is pyrimidyl;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Z is a bond, —NH—, or —O—;
      • V is a bond, —CH2(CR8a′R9a′)—, or —(CR8aR9a)m—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10, —NR11R12, or —CONR11R12;
      • R2 is each independently halogen, —CN, or —CF3;
      • at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), —NHCOCF3 or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl;
      • R8a and R9a are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O;
      • R10 is hydrogen, or C1-C3 alkyl;
      • R11 and R12 are each independently hydrogen or C1-C3 alkyl; or R11 and R12 taken together form an optionally substituted heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O;
      • m is 1, 2, or 3;
      • n2 is 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), n2 is 1 and R2 is halogen.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), X is a bond, —CH2— or —C(CH3)2—. In one embodiment, X is —C(CH3)2—. In some embodiments, X is —O—.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), Y and Z are each —O—.


In one embodiment of the compounds of formula (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —V-L is —CH2CH2Cl.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —Y—W— is —OCH2— or —OCH(CH3)—.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), L is a halogen.


In one embodiment of the compounds of formula (G′), (H′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K).

    • Z is a bond —NH—, or —O—; and
    • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
    • V is —CH2CH2— and L is halogen.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (H′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), C is a pyrimidine, triazine, or thiophene ring.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (H′), (I-A), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


In one embodiment, C is




embedded image


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), C is a pyrimidine ring. In one embodiment, C is




embedded image


In one embodiment, C is




embedded image


In one embodiment of the compounds of formula (E′-I), (F′), (G′), (H′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R1 and R2 are each independently Cl, —CN, or —CF3. In one embodiment, R1 and R2 are each independently Cl or —CN.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is each independently, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 5- or 6-membered heterocyclyl. In one embodiment, at least one R3 is —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, —NH2, —NH(C1-C3 alkyl), or —NHCOCF3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)CO(C1-C3 alkyl), —NHCOCF3, —N(CH3)COCF3, —NHCOO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl); or optionally substituted 5- or 6-membered heterocyclyl.


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3. In one embodiment, at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3 and the other R3 is, if present, —CN, C1-C3 alkyl, C1-C3 alkoxy, —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In one embodiment of the compounds of formula (C′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, or —NH2.


In one embodiment, the present disclosure provides compounds of formula (D′):




embedded image




    • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A is absent;
      • B is 6- to 15-membered aryl ring;
      • C is a pyrimidine, triazine, or thiophene ring;
      • X is a bond, —CH2—, —C(CH3)2—, or —O—;
      • Y is a bond or —O—;
      • W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—, —CH2— or —C(CH3)H—;
      • Z is a bond or —O—;
      • V is —CH2— and L is halogen, —CHCl2, —CCl3, or —CF3; or
      • V is —CH2CH2— and L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, or —CF3;
      • at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3; and the other R3 is, if present, —CN, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), or optionally substituted 4- to 6-membered heterocyclyl or heteroaryl;
      • n1 is 0;
      • n2 is 0, 1, or 2; and
      • n3 is 1, 2, 3, 4 or 5.





In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), or (B-I), B is phenyl or fluorene ring, wherein each ring is optionally substituted with one or two R2.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), X is a bond, —CH2— or —C(CH3)2. In one embodiment, X is —C(CH3)2—. In one embodiment, X is —O—.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), Y and Z are each —O—.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —V-L is —CH2CH2Cl.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —V-L is —O-cyclopropyl.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), —Y—W— is —OCH2— or —OCH(CH3)—.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (C), or (C-I), C is




embedded image


In one embodiment, C is




embedded image


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), C is a pyrimidine ring. In one embodiment, C is




embedded image


In one embodiment, C is




embedded image


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R1 and R2 are each independently Cl, —CN, or —CF3. In one embodiment, R1 and R2 are each independently Cl or —CN.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, —SO2CH3, or —NH2.


In one embodiment of the compounds of formula (D′), (I-A), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3. In one embodiment, at least one R3 is —NHSO2CH3, —NHSO2CH2CH3, —SO2NH2, or —SO2CH3 and the other R3 is, if present, —CN, C1-C3 alkyl, C1-C3 alkoxy, —SO2(C1-C3 alkyl), —NH2, —(C1-C3 alkyl)NH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), —N(CH3)COO(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)COO(C1-C3 alkyl).


In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —N═S(═O)R14R15. In some embodiments, R3 is —N═S(═O)R14R15, wherein R14 and R15 are each independently C1-C3 alkyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O. In some embodiments, R3 is —N═S(═O)R14R15, wherein R14 and R15 taken together form an optionally substituted 5- to 6-membered heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O. In some embodiments, R3 is —N═S(═O)R14R15, wherein R14 and R15 taken together form an optionally substituted 5- to 6-membered heterocyclyl which optionally contains one additional N as a ring atom. In some embodiments, R3 is




embedded image


In some embodiments, R3 is —N═S(═O)(C1-C3 alkyl)2.


In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), R3 is —N═S(═O)R14R15, wherein R14 and R15 are each independently C1-C3 alkyl; or R14 and R15 taken together form an optionally substituted 3- to 6-membered heterocyclyl which optionally contains one or two additional heteroatoms selected from N, S, or O.


In one embodiment of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—. In one embodiment, X is —CH2—, —C(CH3)H—, or —C(CH3)2—. In some embodiments, X is —C(CH3)2—. In one embodiment, X is —O—.


In some embodiments of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), W is a bond, heteroaryl, heterocycle, heteroaryl-NR7—, heterocyclyl-NR7—.


In one embodiment of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), n1 is 0, 1, or 2. In some embodiments, n1 is 0 or 1. In other embodiments, n1 is 0. In some embodiments, n1 is 1. In one embodiment, the sum of n1 and n2 is 0, 1, 2, 3, or 4. In some embodiments, the sum of n1 and n2 is 1, 2, 3, or 4. In one embodiment, the sum of n1 and n2 is 2.


In one embodiment of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), n2 is 0, 1, or 2. In some embodiments, n2 is 1 or 2. In other embodiments, n2 is 0. In some embodiments, n2 is 1. In some embodiments, n2 is 2.


In some embodiments of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), n3 is 1, 2, 3, 4, or 5. In some embodiments, n3 is 1, 2, 3, or 4. In one embodiment, n3 is 1, 2, or 3. In one embodiment, n3 is 1 or 2.


In one embodiment of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), optional substituent is selected from halogen, —CN, —CF3, —OH, —S(C1-C3 alkyl), C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —(C1-C3 alkyl)-(C1-C3 alkoxy), —(C1-C3 alkyl)-OH, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO(C1-C3 alkyl), —SO2(C1-C3 alkyl), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl). In another embodiment, the optional substituent is halogen, —CN, —CF3, —OH, C1-C3 alkyl, C1-C3 alkoxy, —NH2, —SCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3. In another embodiment, the optional substituent is halogen, —CN, —CF3, —OH, C1-C3 alkyl, C1-C3 alkyl-OH, C1-C3 alkoxy, —NH2, —SCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3.


In some embodiments of the compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), a hydrogen atom can be replaced with a deuterium atom.


In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), the compound excludes Compounds A1-A234 disclosed in WO 2020/081999. In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), the compound excludes Compounds presented in Table A of WO 2020/081999. In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), the compound excludes Compounds A1-A285 disclosed in WO 2022/226349. In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), the compound excludes Compounds presented in Table A of WO 2022/226349. In one embodiment of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), the compound is selected from Table A below, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the present disclosure, the compound is selected from Compounds A1-A245, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the present disclosure, the compound is selected from Compounds A1-A185, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the present disclosure, the compound is selected from Compounds A1-A119, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the present disclosure, the compound is selected from Compounds A120-A185, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the present disclosure, the compound is selected from Compounds A186-A245, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (A), the compound is Compound A7, A9, A16, A21, A23, A24, A30, A35, A38, A43, A49, A61, A65, A66, A70, A78, A79, A83, A88, A89, A90, A91, A94, A96, A97, A98, A99, A188, A189, A190, A191, A192, A193, A194, A195, A196, A198, A200, A204, A205, A206, A207, A209, A210, A211, A212, A213, A214, A217, A218, A219, A225, A227, A229, A230, A231, A240, A241, A242, or A243, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (B), the compound is Compound A8, A15, A20, A25, A26, A29, A33, A34, A39, A46, A51, A63, A64, A71, A72, A73, or A74, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (B-I), the compound is Compound A8, A15, A20, A25, A26, A29, A33, A34, A39, A46, A51, A63, A64, A71, A72, A73, A74, A137, A140, A141, A142, A143, A150, A228, A234, A235, or A236, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (C), the compound is Compound A1, A2, A3, A4, A5, A6, A10, All, A12, A13, A14, A18, A19, A22, A27, A28, A31, A32, A36, A37, A40, A41, A44, A45, A47, A48, A50, A53, A54, A55, A57, A58, A59, A60, A67, A68, A69, A75, A76, A77, A80, A81, A82, A84, A92, A93, or A95, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (C-I), the compound is Compound A1, A2, A3, A4, A5, A6, A10, All, A12, A13, A14, A18, A19, A22, A27, A28, A31, A32, A36, A37, A40, A41, A44, A45, A47, A48, A50, A53, A54, A55, A57, A58, A59, A60, A67, A68, A69, A75, A76, A77, A80, A81, A82, A84, A92, A93, A95, A120, A123, A124, A127, A138, A144, A145, A151, A154, A155, A156, A157, A158, A163, A164, A177, A186, A187, A188, A189, A190, A191, A192, A193, A194, A195, A196, A197, A198, A199, A200, A203, A204, A205, A206, A207, A208, A209, A210, A211, A212, A213, A214, A215, A216, A217, A218, A219, A220, A222, A224, A226, A229, A230, A231, A232, A233, A239, A240, A241, A242, or A243, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (C-I), the compound is Compound A1, A2, A3, A4, A5, A6, A10, All, A12, A13, A14, A18, A19, A22, A27, A28, A31, A32, A36, A37, A40, A41, A44, A45, A47, A48, A50, A53, A54, A55, A57, A58, A59, A60, A67, A68, A69, A75, A76, A77, A80, A81, A82, A84, A92, A93, A95, A124, A138, A144, A145, A151, A163, A164, or A177 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (D), the compound is Compound A17, A42, A52, A56, A62, or A102, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (D-I), the compound is Compound A17, A42, A52, A56, A62, A102, A121, A126, A129, A132, A133, A134, A136, A139, A146, A147, A148, A149, A152, A153, A159, A160, A161, A165, A166, A167, A168, A169, A178, A179, A180, A181, A182, A183, A184, A201, A202, A221, A223, A237, A244, or A245, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (E), the compound is Compound A85, A86, or A87, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (E-I), the compound is Compound A85, A86, A87, or A135, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (F), the compound is Compound A100, A101, A103, A104, A105, A106, A107, A108, A109, or A110 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (G), the compound is Compound A111, A112, A113, A114, A115, A116, A117, A118, or A119, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (H), the compound is Compound A125, A130, or A131, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (J), the compound is Compound A170, A171, A172, A173, A174, A175, or A176, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment of the compound of formula (K), the compound is Compound A128, A162, A185, or A238, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.









TABLE A







Compounds








Compd.



ID
Structure





A1


embedded image







A2


embedded image







A3


embedded image







A4


embedded image







A5


embedded image







A6


embedded image







A7


embedded image







A8


embedded image







A9


embedded image







A10


embedded image







A11


embedded image







A12


embedded image







A13


embedded image







A14


embedded image







A15


embedded image







A16


embedded image







A17


embedded image







A18


embedded image







A19


embedded image







A20


embedded image







A21


embedded image







A22


embedded image







A23


embedded image







A24


embedded image







A25


embedded image







A26


embedded image







A27


embedded image







A28


embedded image







A29


embedded image







A30


embedded image







A31


embedded image







A32


embedded image







A33


embedded image







A34


embedded image







A35


embedded image







A36


embedded image







A37


embedded image







A38


embedded image







A39


embedded image







A40


embedded image







A41


embedded image







A42


embedded image







A43


embedded image







A44


embedded image







A45


embedded image







A46


embedded image







A47


embedded image







A48


embedded image







A49


embedded image







A50


embedded image







A51


embedded image







A52


embedded image







A53


embedded image







A54


embedded image







A55


embedded image







A56


embedded image







A57


embedded image







A58


embedded image







A59


embedded image







A60


embedded image







A61


embedded image







A62


embedded image







A63


embedded image







A64


embedded image







A65


embedded image







A66


embedded image







A67


embedded image







A68


embedded image







A69


embedded image







A70


embedded image







A71


embedded image







A72


embedded image







A73


embedded image







A74


embedded image







A75


embedded image







A76


embedded image







A77


embedded image







A78


embedded image







A79


embedded image







A80


embedded image







A81


embedded image







A82


embedded image







A83


embedded image







A84


embedded image







A85


embedded image







A86


embedded image







A87


embedded image







A88


embedded image







A89


embedded image







A90


embedded image







A91


embedded image







A92


embedded image







A93


embedded image







A94


embedded image







A95


embedded image







A96


embedded image







A97


embedded image







A98


embedded image







A99


embedded image







A100


embedded image







A101


embedded image







A102


embedded image







A103


embedded image







A104


embedded image







A105


embedded image







A106


embedded image







A107


embedded image







A108


embedded image







A109


embedded image







A110


embedded image







A111


embedded image







A112


embedded image







A113


embedded image







A114


embedded image







A115


embedded image







A116


embedded image







A117


embedded image







A118


embedded image







A119


embedded image







A120


embedded image







A121


embedded image







A122


embedded image







A123


embedded image







A124


embedded image







A125


embedded image







A126


embedded image







A127


embedded image







A128


embedded image







A129


embedded image







A130


embedded image







A131


embedded image







A132


embedded image







A133


embedded image







A134


embedded image







A135


embedded image







A136


embedded image







A137


embedded image







A138


embedded image







A139


embedded image







A140


embedded image







A141


embedded image







A142


embedded image







A143


embedded image







A144


embedded image







A145


embedded image







A146


embedded image







A147


embedded image







A148


embedded image







A149


embedded image







A150


embedded image







A151


embedded image







A152


embedded image







A153


embedded image







A154


embedded image







A155


embedded image







A156


embedded image







A157


embedded image







A158


embedded image







A159


embedded image







A160


embedded image







A161


embedded image







A162


embedded image







A163


embedded image







A164


embedded image







A165


embedded image







A166


embedded image







A167


embedded image







A168


embedded image







A169


embedded image







A170


embedded image







A171


embedded image







A172


embedded image







A173


embedded image







A174


embedded image







A175


embedded image







A176


embedded image







A177


embedded image







A178


embedded image







A179


embedded image







A180


embedded image







A181


embedded image







A182


embedded image







A183


embedded image







A184


embedded image







A185


embedded image







A186


embedded image







A187


embedded image







A188


embedded image







A189


embedded image







A190


embedded image







A191


embedded image







A192


embedded image







A193


embedded image







A194


embedded image







A195


embedded image







A196


embedded image







A197


embedded image







A198


embedded image







A199


embedded image







A200


embedded image







A201


embedded image







A202


embedded image







A203


embedded image







A204


embedded image







A205


embedded image







A206


embedded image







A207


embedded image







A208


embedded image







A209


embedded image







A210


embedded image







A211


embedded image







A212


embedded image







A213


embedded image







A214


embedded image







A215


embedded image







A216


embedded image







A217


embedded image







A218


embedded image







A219


embedded image







A220


embedded image







A221


embedded image







A222


embedded image







A223


embedded image







A224


embedded image







A225


embedded image







A226


embedded image







A227


embedded image







A228


embedded image







A229


embedded image







A230


embedded image







A231


embedded image







A232


embedded image







A233


embedded image







A234


embedded image







A235


embedded image







A236


embedded image







A237


embedded image







A238


embedded image







A239


embedded image







A240


embedded image







A241


embedded image







A242


embedded image







A243


embedded image







A244


embedded image







A245


embedded image











In one embodiment, the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound of any one of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or compounds of Table A, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


a, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


In one embodiment, the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1-A119 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment, the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1-A185 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment, the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1-A245 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.


Therapeutic Use

The present compounds find use in any number of methods. For example, in some embodiments the compounds are useful in methods for modulating androgen receptor (AR). Accordingly, in one embodiment, the present disclosure provides the use of any one of the foregoing compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, for modulating androgen receptor (AR) activity. For example, in some embodiments, modulating androgen receptor (AR) activity is in a mammalian cell. Modulating androgen receptor (AR) can be in a subject in need thereof (e.g., a mammalian subject) and for treatment of any of the described conditions or diseases.


In one embodiment, the modulating AR is binding to AR. In other embodiments, the modulating AR is inhibiting AR.


In one embodiment, the modulating AR is modulating AR N-terminal domain (NTD). In one embodiment, the modulating AR is binding to AR NTD. In other embodiments, the modulating AR is inhibiting AR NTD. In one embodiment, the modulating AR is modulating AR N-terminal domain (NTD). In some embodiments, modulating the AR is inhibiting transactivation of androgen receptor N-terminal domain (NTD).


In other embodiments, modulating androgen receptor (AR) activity is for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, bladder cancer, pancreatic cancer, hepatocellular cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, age related macular degeneration, and combinations thereof. For example, in some embodiments, the indication is prostate cancer. In other embodiments, the prostate cancer is primary/localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic prostate cancer, advanced prostate cancer, or metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. While in other embodiments, the prostate cancer is androgen dependent prostate cancer. In other embodiments, the spinal and bulbar muscular atrophy is Kennedy's disease. In other embodiments, the prostate cancer is primary/localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, metastatic prostate cancer, advanced prostate cancer, or metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. While in other embodiments, the prostate cancer is androgen dependent prostate cancer. In other embodiments, the spinal and bulbar muscular atrophy is Kennedy's disease.


In one embodiment of the present disclosure, a method of treating a condition associated with cell proliferation in a patient in need thereof is provided, comprising administering a compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, to a subject in need thereof. In one embodiment, the present invention provides a method of treating cancer or tumors. In another embodiment, the present invention provides a method of treating prostate cancer or breast cancer.


In one embodiment of the present disclosure, a method of reducing, inhibiting, or ameliorating proliferation, comprising administering a therapeutically effective amount of a compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof is provided. In one embodiment, the reducing, inhibiting, or ameliorating in the method disclosed herein, is in vivo. In another embodiment, the reducing, inhibiting, or ameliorating is in vitro.


In one embodiment, the cells in the method disclosed herein, are a cancer cells. In one embodiment, the cancer cells are a prostate cancer cells. In one embodiment, the prostate cancer cells are cells of primary/localized prostate cancer (newly diagnosed or early stage), locally advanced prostate cancer, recurrent prostate cancer (e.g., prostate cancer which was not responsive to primary therapy), metastatic prostate cancer, non-metastatic castration-resistant prostate cancer, advanced prostate cancer (e.g., after castration for recurrent prostate cancer), metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. In one embodiment, the prostate cancer cells are cells of primary/localized prostate cancer (newly diagnosed or early stage), locally advanced prostate cancer, recurrent prostate cancer (e.g., prostate cancer which was not responsive to primary therapy), metastatic prostate cancer, advanced prostate cancer (e.g., after castration for recurrent prostate cancer), metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. In another embodiment, the prostate cancer cells are cells of a metastatic castration-resistant prostate cancer. In other embodiments, the prostate cancer cells are an androgen-dependent prostate cancer cells or an androgen-independent prostate cancer cells. In one embodiment, the cancer cells are breast cancer cells.


In one embodiment, the condition or disease associated with cell proliferation is cancer. In one embodiment of any one of the methods disclosed herein, the cancer is selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. In one embodiment, the condition or disease is prostate cancer. In one embodiment, prostate cancer is selected from primary/localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, metastatic prostate cancer, non-metastatic castration-resistant prostate cancer, advanced prostate cancer, metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. In one embodiment, prostate cancer is selected from primary/localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, metastatic prostate cancer, advanced prostate cancer, metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. In another embodiment, the prostate cancer is a metastatic castration-resistant prostate cancer. In some embodiments, the prostate cancer is an androgen-dependent prostate cancer cells or an androgen-independent prostate cancer. In one embodiment, the condition or disease is breast cancer.


In another embodiment of the present disclosure, a method for reducing or preventing tumor growth, comprising contacting tumor cells with a therapeutically effective amount of a compound of (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof is provided.


In one embodiment, reducing or preventing tumor growth includes reduction in tumor volume. In one embodiment, reducing or preventing tumor growth includes complete elimination of tumors. In one embodiment, reducing or preventing tumor growth includes stopping or halting the existing tumor to grow. In one embodiment, reducing or preventing tumor growth includes reduction in the rate of tumor growth. In one embodiment, reducing or preventing tumor growth includes reduction in the rate of tumor growth such that the rate of tumor growth before treating a patient with the methods disclosed herein (r1) is faster than the rate of tumor growth after said treatment (r2) such that r1>r2.


In one embodiment, the reducing or preventing in the method disclosed herein is in vivo. In another embodiment, the treating is in vitro.


In one embodiment, the tumor cell in the method disclosed herein is selected from prostate cancer, breast cancer, ovarian cancer, endometrial cancer, or salivary gland carcinoma. In one embodiment, the tumor cells are prostate cancer tumor cells. In one embodiment, the prostate cancer tumor cells are tumor cells of primary/localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, metastatic prostate cancer, non-metastatic castration-resistant prostate cancer, advanced prostate cancer, metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. In one embodiment, the prostate cancer tumor cells are tumor cells of primary/localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, metastatic prostate cancer, advanced prostate cancer, metastatic castration-resistant prostate cancer (CRPC), or hormone-sensitive prostate cancer. In other embodiments, the prostate cancer is a metastatic castration-resistant prostate cancer. In some embodiments, the prostate cancer is androgen-dependent prostate cancer or androgen-independent prostate cancer. In another embodiment, the tumor cells are is breast cancer tumor cells.


In one embodiment, the present disclosure provides compounds which demonstrate blocking androgen-induced PSA-luciferase activity (PSA-luciferase assay). In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 4500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 4000 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 3500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 3000 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 2500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 2000 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 1500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 1000 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 950 nm. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 900 nM.


In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 890 nM, less than about 880 nM, less than about 870 nM, less than about 860 nM, less than about 850 nM, less than about 840 nM, less than about 830 nM, less than about 820 nM, less than about 810 nM, or less than about 800 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro 1C50 in a PSA-luciferase assay of less than about 790 nM, less than about 780 nM, less than about 770 nM, less than about 760 nM, less than about 750 nM, less than about 740 nM, less than about 730 nM, less than about 720 nM, less than about 710 nM, or less than about 700 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 690 nM, less than about 680 nM, less than about 670 nM, less than about 660 nM, less than about 650 nM, less than about 640 nM, less than about 630 nM, less than about 620 nM, less than about 610 nM, or less than about 600 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 590 nM, less than about 580 nM, less than about 570 nM, less than about 560 nM, less than about 550 nM, less than about 540 nM, less than about 530 nM, less than about 520 nM, less than about 510 nM, or less than about 500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds having in vitro IC50 in a PSA-luciferase assay of less than about 490 nM, less than about 480 nM, less than about 470 nM, less than about 460 nM, less than about 450 nM, less than about 440 nM, less than about 430 nM, less than about 420 nM, less than about 410 nM, or less than about 400 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 390 nM, less than about 380 nM, less than about 370 nM, less than about 360 nM, less than about 350 nM, less than about 340 nM, less than about 330 nM, less than about 320 nM, less than about 310 nM, or less than about 300 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 290 nM, less than about 280 nM, less than about 270 nM, less than about 260 nM, less than about 250 nM, less than about 240 nM, less than about 230 nM, less than about 220 nM, less than about 210 nM, or less than about 200 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 200 nM. In one embodiment, the compound is selected from any of the compound of Table A or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.


In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 80 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 2000 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), and (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 90 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 1500 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 100 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 1000 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 105 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 900 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 110 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 850 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 115 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 800 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 115 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 750 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 120 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 120 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 650 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 120 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 600 nM. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Table A, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mammal microsome of greater than about 120 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 550 nM.


In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than 650 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 640 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 635 nM.


In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro IC50 in a PSA-luciferase assay in the range of 200 nM to about 700 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay in the range of about 250 nM to about 700 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay in the range of about 300 nM to about 700 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay in the range of about 350 nM to about 675 nM.


In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro IC50 in a PSA-luciferase assay of less than 525 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 500 nM. In one embodiment, the compounds have an in vitro IC50 in a PSA-luciferase assay of less than about 450 nM.


Pharmaceutical Compositions and Formulations

The present disclosure also includes pharmaceutical compositions for modulating androgen receptor (AR) in a subject. In one embodiment, a pharmaceutical composition comprises one or more compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof.


In one embodiment of the present disclosure, a pharmaceutical composition comprises a therapeutically effective amounts of one or more compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof.


In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Table A, or a pharmaceutically acceptable salt or solvate thereof.


In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A1-A119 or a pharmaceutically acceptable salt or solvate thereof.


In one embodiment, a pharmaceutical composition, as described herein, comprising one or more compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof, further comprises one or more additional therapeutically active agents. In one embodiment, one or more additional therapeutically active agents are selected from therapeutics useful for treating cancer, neurological disease, a disorder characterized by abnormal accumulation of α-synuclein, a disorder of an aging process, cardiovascular disease, bacterial infection, viral infection, mitochondrial related disease, mental retardation, deafness, blindness, diabetes, obesity, autoimmune disease, glaucoma, Leber's Hereditary Optic Neuropathy, and rheumatoid arthritis.


In some embodiments, the one or more additional therapeutic agents is a poly (ADP-ribose) polymerase (PARP) inhibitor including but not limited to olaparib, niraparib, rucaparib, talazoparib; an androgen receptor ligand binding domain inhibitor including but not limited to enzalutamide, apalutamide, darolutamide, bicalutamide, nilutamide, flutamide, ODM-204, TAS3681; an inhibitor of CYP17 including but not limited to galeterone, abiraterone, abiraterone acetate; a microtubule inhibitor including but not limited to docetaxel, paclitaxel, cabazitaxel (XRP-6258); a modulator of PD-1 or PD-L1 including but not limited to pembrolizumab, durvalumab, nivolumab, atezolizumab; a gonadotropin releasing hormone agonist including but not limited to cyproterone acetate, leuprolide, a 5-alpha reductase inhibitor including but not limited to finasteride, dutasteride, turosteride, bexlosteride, izonsteride, FCE 28260, SKF 105,111; a vascular endothelial growth factor inhibitor including but not limited to bevacizumab (Avastin); a histone deacetylase inhibitor including but not limited to OSU-HDAC42; an integrin alpha-v-beta-3 inhibitor including but not limited to VITAXIN; a receptor tyrosine kinase inhibitor including but not limited to sunitumib; a phosphoinositide 3-kinase inhibitor including but not limited to alpelisib, buparlisib, idealisib; an anaplastic lymphoma kinase (ALK) inhibitor including but not limited to crizotinib, alectinib; an endothelin receptor A antagonist including but not limited to ZD-4054; an anti-CTLA4 inhibitor including but not limited to MDX-010 (ipilimumab); an heat shock protein 27 (HSP27) inhibitor including but not limited to OGX 427; an androgen receptor degrader including but not limited to ARV-330, ARV-110; a androgen receptor DNA-binding domain inhibitor including but not limited to VPC-14449; a bromodomain and extra-terminal motif (BET) inhibitor including but not limited to BI-894999, GSK25762, GS-5829; an androgen receptor N-terminal domain inhibitor including but not limited to a sintokamide; an alpha-particle emitting radioactive therapeutic agent including but not limited to radium 233 or a salt thereof; niclosamide; or related compounds thereof; a selective estrogen receptor modulator (SERM) including but not limited to tamoxifen, raloxifene, toremifene, arzoxifene, bazedoxifene, pipindoxifene, lasofoxifene, enclomiphene; a selective estrogen receptor degrader (SERD) including but not limited to fulvestrant, ZB716, OP-1074, elacestrant, AZD9496, GDC0810, GDC0927, GW5638, GW7604; an aromitase inhibitor including but not limited to anastrazole, exemestane, letrozole; selective progesterone receptor modulators (SPRM) including but not limited to mifepristone, lonaprison, onapristone, asoprisnil, lonaprisnil, ulipristal, telapristone; a glucocorticoid receptor inhibitor including but not limited to mifepristone, COR108297, COR125281, ORIC-101, PT150; CDK4/6 inhibitors including palbociclib, abemaciclib, ribociclib; HER2 receptor antagonist including but not limited to trastuzumab, neratinib; a mammalian target of rapamycin (mTOR) inhibitor including but not limited to everolimus, temsirolimus.


In a further embodiment of the present disclosure, a pharmaceutical composition comprising one or more compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient or adjuvant is provided. The pharmaceutically acceptable excipients and adjuvants are added to the composition or formulation for a variety of purposes. In another embodiment, a pharmaceutical composition comprising one or more compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof, further comprises a pharmaceutically acceptable carrier. In one embodiment, a pharmaceutically acceptable carrier includes a pharmaceutically acceptable excipient, binder, and/or diluent. In one embodiment, suitable pharmaceutically acceptable excipients include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylase, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose and polyvinylpyrrolidone.


In certain embodiments, the pharmaceutical compositions of the present disclosure may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels. Thus, for example, the pharmaceutical compositions may contain additional, compatible, pharmaceutically active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the present invention. The formulations can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the oligonucleotide(s) of the formulation.


For the purposes of this disclosure, the compounds of the present disclosure can be formulated for administration by a variety of means including orally, parenterally, by inhalation spray, topically, or rectally in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used here includes subcutaneous, intravenous, intramuscular, and intraarterial injections with a variety of infusion techniques. Intraarterial and intravenous injection as used herein includes administration through catheters.


The compounds disclosed herein can be formulated in accordance with the routine procedures adapted for desired administration route. Accordingly, the compounds disclosed herein can take such forms as suspensions, dispersions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The compounds disclosed herein can also be formulated as a preparation for implantation or injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt). Alternatively, the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. Suitable formulations for each of these methods of administration can be found, for example, in Remington: The Science and Practice of Pharmacy, A. Gennaro, ed., 20th edition, Lippincott, Williams & Wilkins, Philadelphia, PA.


In certain embodiments, a pharmaceutical composition of the present disclosure is prepared using known techniques, including, but not limited to mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.


In one embodiment, the present disclosure provides a pharmaceutical composition comprising a compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof, as disclosed herein, combined with a pharmaceutically acceptable carrier. In one embodiment, suitable pharmaceutically acceptable carriers include, but are not limited to, inert solid fillers or diluents and sterile aqueous or organic solutions. Pharmaceutically acceptable carriers are well known to those skilled in the art and include, but are not limited to, from about 0.01 to about 0.1 M and preferably 0.05M phosphate buffer or 0.8% saline. Such pharmaceutically acceptable carriers can be aqueous or non-aqueous solutions, suspensions and emulsions. Examples of non-aqueous solvents suitable for use in the present application include, but are not limited to, propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.


Aqueous carriers suitable for use in the present application include, but are not limited to, water, ethanol, alcoholic/aqueous solutions, glycerol, emulsions or suspensions, including saline and buffered media. Oral carriers can be elixirs, syrups, capsules, tablets and the like.


Liquid carriers suitable for use in the present application can be used in preparing solutions, suspensions, emulsions, syrups, elixirs and pressurized compounds. The active ingredient can be dissolved or suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, a mixture of both or pharmaceutically acceptable oils or fats. The liquid carrier can contain other suitable pharmaceutical additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers or osmo-regulators.


Liquid carriers suitable for use in the present application include, but are not limited to, water (partially containing additives as above, e.g. cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g. glycols) and their derivatives, and oils (e.g. fractionated coconut oil and arachis oil). For parenteral administration, the carrier can also include an oily ester such as ethyl oleate and isopropyl myristate. Sterile liquid carriers are useful in sterile liquid form comprising compounds for parenteral administration. The liquid carrier for pressurized compounds disclosed herein can be halogenated hydrocarbon or other pharmaceutically acceptable prop ell ent.


Solid carriers suitable for use in the present application include, but are not limited to, inert substances such as lactose, starch, glucose, methylcellulose, magnesium stearate, dicalcium phosphate, mannitol and the like. A solid carrier can further include one or more substances acting as flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aids, binders or tablet-disintegrating agents; it can also be an encapsulating material. In powders, the carrier can be a finely divided solid which is in admixture with the finely divided active compound. In tablets, the active compound is mixed with a carrier having the necessary compression properties in suitable proportions and compacted in the shape and size desired. The powders and tablets preferably contain up to 99% of the active compound. Suitable solid carriers include, for example, calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, polyvinylpyrrolidine, low melting waxes and ion exchange resins. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methylcellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.


Parenteral carriers suitable for use in the present application include, but are not limited to, sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's and fixed oils. Intravenous carriers include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose and the like. Preservatives and other additives can also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases and the like.


Carriers suitable for use in the present application can be mixed as needed with disintegrants, diluents, granulating agents, lubricants, binders and the like using conventional techniques known in the art. The carriers can also be sterilized using methods that do not deleteriously react with the compounds, as is generally known in the art.


Diluents may be added to the formulations of the present invention. Diluents increase the bulk of a solid pharmaceutical composition and may make a pharmaceutical dosage form containing the composition easier for the patient and care giver to handle. Diluents for solid compositions include, for example, microcrystalline cellulose (e.g., AVICEL), microfine cellulose, lactose, starch, pregelatinized starch, calcium carbonate, calcium sulfate, sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, tribasic calcium phosphate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, polymethacrylates (e.g., EUDRAGIT®), potassium chloride, powdered cellulose, sodium chloride, sorbitol, and talc.


Additional embodiments relate to the pharmaceutical formulations wherein the formulation is selected from the group consisting of a solid, powder, liquid and a gel. In certain embodiments, a pharmaceutical composition of the present invention is a solid (e.g., a powder, tablet, a capsule, granulates, and/or aggregates). In certain of such embodiments, a solid pharmaceutical composition comprising one or more ingredients known in the art, including, but not limited to, starches, sugars, diluents, granulating agents, lubricants, binders, and disintegrating agents.


Solid pharmaceutical compositions that are compacted into a dosage form, such as a tablet, may include excipients whose functions include helping to bind the active ingredient and other excipients together after compression. Binders for solid pharmaceutical compositions include acacia, alginic acid, carbomer (e.g., carbopol), carboxymethylcellulose sodium, dextrin, ethyl cellulose, gelatin, guar gum, gum tragacanth, hydrogenated vegetable oil, hydroxyethyl cellulose, hydroxypropyl cellulose (e.g., KLUCEL), hydroxypropyl methyl cellulose (e.g., METHOCEL), liquid glucose, magnesium aluminum silicate, maltodextrin, methylcellulose, polymethacrylates, povidone (e.g., KOLLIDON, PLASDONE), pregelatinized starch, sodium alginate, and starch.


The dissolution rate of a compacted solid pharmaceutical composition in the patient's stomach may be increased by the addition of a disintegrant to the composition. Disintegrants include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g., AC-DI-SOL and PRIMELLOSE), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g., KOLLIDON and POLYPLASDONE), guar gum, magnesium aluminum silicate, methyl cellulose, microcrystalline cellulose, polacrilin potassium, powdered cellulose, pregelatinized starch, sodium alginate, sodium starch glycolate (e.g., EXPLOTAB), potato starch, and starch.


Glidants can be added to improve the flowability of a non-compacted solid composition and to improve the accuracy of dosing. Excipients that may function as glidants include silicon dioxide, magnesium tri silicate, powdered cellulose, starch, talc, and tribasic calcium phosphate.


When a dosage form such as a tablet is made by the compaction of a powdered composition, the composition is subjected to pressure from a punch and dye. Some excipients and active ingredients have a tendency to adhere to the surfaces of the punch and dye, which can cause the product to have pitting and other surface irregularities. A lubricant can be added to the composition to reduce adhesion and ease the release of the product from the dye. Lubricants include magnesium stearate, calcium stearate, glyceryl monostearate, glyceryl palmitostearate, hydrogenated castor oil, hydrogenated vegetable oil, mineral oil, polyethylene glycol, sodium benzoate, sodium lauryl sulfate, sodium stearyl fumarate, stearic acid, talc, and zinc stearate.


Flavoring agents and flavor enhancers make the dosage form more palatable to the patient. Common flavoring agents and flavor enhancers for pharmaceutical products that may be included in the composition of the present invention include maltol, vanillin, ethyl vanillin, menthol, citric acid, fumaric acid, ethyl maltol, and tartaric acid.


Solid and liquid compositions may also be dyed using any pharmaceutically acceptable colorant to improve their appearance and/or facilitate patient identification of the product and unit dosage level.


In certain embodiments, a pharmaceutical composition of the present invention is a liquid (e.g., a suspension, elixir and/or solution). In certain of such embodiments, a liquid pharmaceutical composition is prepared using ingredients known in the art, including, but not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.


Liquid pharmaceutical compositions can be prepared using compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof, and any other solid excipients where the components are dissolved or suspended in a liquid carrier such as water, vegetable oil, alcohol, polyethylene glycol, propylene glycol, or glycerin.


For example, formulations for parenteral administration can contain as common excipients sterile water or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like. In particular, biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers can be useful excipients to control the release of active compounds. Other potentially useful parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation administration contain as excipients, for example, lactose, or can be aqueous solutions containing, for example, polyoxyethylene-9-auryl ether, glycocholate and deoxycholate, or oily solutions for administration in the form of nasal drops, or as a gel to be applied intranasally. Formulations for parenteral administration can also include glycocholate for buccal administration, methoxysalicylate for rectal administration, or citric acid for vaginal administration.


Liquid pharmaceutical compositions can contain emulsifying agents to disperse uniformly throughout the composition an active ingredient or other excipient that is not soluble in the liquid carrier. Emulsifying agents that may be useful in liquid compositions of the present invention include, for example, gelatin, egg yolk, casein, cholesterol, acacia, tragacanth, chondrus, pectin, methyl cellulose, carbomer, cetostearyl alcohol, and cetyl alcohol.


Liquid pharmaceutical compositions can also contain a viscosity enhancing agent to improve the mouth-feel of the product and/or coat the lining of the gastrointestinal tract. Such agents include acacia, alginic acid bentonite, carbomer, carboxymethylcellulose calcium or sodium, cetostearyl alcohol, methyl cellulose, ethylcellulose, gelatin guar gum, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, maltodextrin, polyvinyl alcohol, povidone, propylene carbonate, propylene glycol alginate, sodium alginate, sodium starch glycolate, starch tragacanth, and xanthan gum.


Sweetening agents such as aspartame, lactose, sorbitol, saccharin, sodium saccharin, sucrose, aspartame, fructose, mannitol, and invert sugar may be added to improve the taste.


Preservatives and chelating agents such as alcohol, sodium benzoate, butylated hydroxyl toluene, butylated hydroxyanisole, and ethylenediamine tetraacetic acid may be added at levels safe for ingestion to improve storage stability.


A liquid composition can also contain a buffer such as guconic acid, lactic acid, citric acid or acetic acid, sodium guconate, sodium lactate, sodium citrate, or sodium acetate. Selection of excipients and the amounts used may be readily determined by the formulation scientist based upon experience and consideration of standard procedures and reference works in the field.


In one embodiment, a pharmaceutical composition is prepared for administration by injection (e.g., intravenous, subcutaneous, intramuscular, etc.). In certain of such embodiments, a pharmaceutical composition comprises a carrier and is formulated in aqueous solution, such as water or physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. In certain embodiments, other ingredients are included (e.g., ingredients that aid in solubility or serve as preservatives). In certain embodiments, injectable suspensions are prepared using appropriate liquid carriers, suspending agents and the like. Certain pharmaceutical compositions for injection are presented in unit dosage form, e.g., in ampoules or in multi-dose containers. Certain pharmaceutical compositions for injection are suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Certain solvents suitable for use in pharmaceutical compositions for injection include, but are not limited to, lipophilic solvents and fatty oils, such as sesame oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, and liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, such suspensions may also contain suitable stabilizers or agents that increase the solubility of the pharmaceutical agents to allow for the preparation of highly concentrated solutions.


The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. Formulations for intravenous administration can comprise solutions in sterile isotonic aqueous buffer. Where necessary, the formulations can also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampule or sachet indicating the quantity of active agent. Where the compound is to be administered by infusion, it can be dispensed in a formulation with an infusion bottle containing sterile pharmaceutical grade water, saline or dextrose/water. Where the compound is administered by injection, an ampule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.


Suitable formulations further include aqueous and non-aqueous sterile injection solutions that can contain antioxidants, buffers, bacteriostats, bactericidal antibiotics and solutes that render the formulation isotonic with the bodily fluids of the intended recipient; and aqueous and non-aqueous sterile suspensions, which can include suspending agents and thickening agents.


In certain embodiments, a pharmaceutical composition of the present invention is formulated as a depot preparation. Certain such depot preparations are typically longer acting than non-depot preparations. In certain embodiments, such preparations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. In certain embodiments, depot preparations are prepared using suitable polymeric or hydrophobic materials (for example an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.


In certain embodiments, a pharmaceutical composition of the present invention comprises a delivery system. Examples of delivery systems include, but are not limited to, liposomes and emulsions. Certain delivery systems are useful for preparing certain pharmaceutical compositions including those comprising hydrophobic compounds. In certain embodiments, certain organic solvents such as dimethylsulfoxide are used.


In certain embodiments, a pharmaceutical composition of the present invention comprises a co-solvent system. Certain of such co-solvent systems comprise, for example, benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. In certain embodiments, such co-solvent systems are used for hydrophobic compounds. A non-limiting example of such a co-solvent system is the VPD co-solvent system, which is a solution of absolute ethanol comprising 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80 and 65% w/v polyethylene glycol 300. The proportions of such co-solvent systems may be varied considerably without significantly altering their solubility and toxicity characteristics. Furthermore, the identity of co-solvent components may be varied: for example, other surfactants may be used instead of Polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.


In certain embodiments, a pharmaceutical composition of the present invention comprises a sustained-release system. A non-limiting example of such a sustained-release system is a semi-permeable matrix of solid hydrophobic polymers. In certain embodiments, sustained-release systems may, depending on their chemical nature, release pharmaceutical agents over a period of hours, days, weeks or months.


Appropriate pharmaceutical compositions of the present disclosure can be determined according to any clinically-acceptable route of administration of the composition to the subject. The manner in which the composition is administered is dependent, in part, upon the cause and/or location. One skilled in the art will recognize the advantages of certain routes of administration. The method includes administering an effective amount of the agent or compound (or composition comprising the agent or compound) to achieve a desired biological response, e.g., an amount effective to alleviate, ameliorate, or prevent, in whole or in part, a symptom of a condition to be treated, e.g., oncology and neurology disorders. In various aspects, the route of administration is systemic, e.g., oral or by injection. The agents or compounds, or pharmaceutically acceptable salts or derivatives thereof, are administered orally, nasally, transdermally, pulmonary, inhalationally, buccally, sublingually, intraperintoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally, intraportally, and parenterally. Alternatively, or in addition, the route of administration is local, e.g., topical, intra-tumor and peri-tumor. In some embodiments, the compound is administered orally.


In certain embodiments, a pharmaceutical composition of the present disclosure is prepared for oral administration. In certain of such embodiments, a pharmaceutical composition is formulated by combining one or more agents and pharmaceutically acceptable carriers. Certain of such carriers enable pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject. Suitable excipients include, but are not limited to, fillers, such as sugars, including lactose, lactose monohydrate, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, microcrystalline cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). In certain embodiments, such a mixture is optionally ground and auxiliaries are optionally added. In certain embodiments, pharmaceutical compositions are formed to obtain tablets or dragee cores. In certain embodiments, disintegrating agents (e.g., cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate) are added.


In certain embodiments, dragee cores are provided with coatings. In certain such embodiments, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to tablets or dragee coatings.


In certain embodiments, pharmaceutical compositions for oral administration are push-fit capsules made of gelatin. Certain of such push-fit capsules comprise one or more pharmaceutical agents of the present invention in admixture with one or more filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In certain embodiments, pharmaceutical compositions for oral administration are soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In certain soft capsules, one or more pharmaceutical agents of the present invention are be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added.


In certain embodiments, pharmaceutical compositions are prepared for buccal administration. Certain of such pharmaceutical compositions are tablets or lozenges formulated in conventional manner.


In certain embodiments, a pharmaceutical composition is prepared for transmucosal administration. In one embodiment, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.


In certain embodiments, a pharmaceutical composition is prepared for administration by inhalation. Certain of such pharmaceutical compositions for inhalation are prepared in the form of an aerosol spray in a pressurized pack or a nebulizer. Certain of such pharmaceutical compositions comprise a propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In certain embodiments using a pressurized aerosol, the dosage unit may be determined with a valve that delivers a metered amount. In certain embodiments, capsules and cartridges for use in an inhaler or insufflator may be formulated. Certain of such formulations comprise a powder mixture of a pharmaceutical agent of the invention and a suitable powder base such as lactose or starch.


In other embodiments the compound of the present disclosure are administered by the intravenous route. In further embodiments, the parenteral administration may be provided in a bolus or by infusion.


In certain embodiments, a pharmaceutical composition is prepared for rectal administration, such as a suppository or retention enema. Certain of such pharmaceutical compositions comprise known ingredients, such as cocoa butter and/or other glycerides.


In certain embodiments, a pharmaceutical composition is prepared for topical administration. Certain of such pharmaceutical compositions comprise bland moisturizing bases, such as ointments or creams. Exemplary suitable ointment bases include, but are not limited to, petrolatum, petrolatum plus volatile silicones, and lanolin and water in oil emulsions. Exemplary suitable cream bases include, but are not limited to, cold cream and hydrophilic ointment.


In certain embodiments, the therapeutically effective amount is sufficient to prevent, alleviate or ameliorate symptoms of a disease or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art.


In certain embodiments, one or more compounds of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof are formulated as a prodrug. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically more active form. In certain embodiments, prodrugs are useful because they are easier to administer than the corresponding active form. For example, in certain instances, a prodrug may be more bioavailable (e.g., through oral administration) than is the corresponding active form. In certain instances, a prodrug may have improved solubility compared to the corresponding active form. In certain embodiments, prodrugs are less water soluble than the corresponding active form. In certain instances, such prodrugs possess superior transmittal across cell membranes, where water solubility is detrimental to mobility. In certain embodiments, a prodrug is an ester. In certain such embodiments, the ester is metabolically hydrolyzed to carboxylic acid upon administration. In certain instances, the carboxylic acid containing compound is the corresponding active form. In certain embodiments, a prodrug comprises a short peptide (polyaminoacid) bound to an acid group. In certain of such embodiments, the peptide is cleaved upon administration to form the corresponding active form.


In certain embodiments, a prodrug is produced by modifying a pharmaceutically active compound such that the active compound will be regenerated upon in vivo administration. The prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. By virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, those of skill in this art, once a pharmaceutically active compound is known, can design prodrugs of the compound (see, e.g., Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392).


In various aspects, the amount of the compound of formula (I), (I-A), (I-B), (II), (A′), (A′-I), (A′-II), (A′-III), (B′), (B′-I), (C′), (D′), (E′), (E′-I), (F′), (G′), (H′), (J′), (K′), (ABC), (BC), (A), (B), (B-I), (C), (C-I), (D), (D-I), (E), (E-I), (F), (G), (H), (J), or (K), or a pharmaceutically acceptable salt or solvate thereof, or compounds disclosed in Table A, or a pharmaceutically acceptable salt or solvate thereof, can be administered at about 0.001 mg/kg to about 100 mg/kg body weight (e.g., about 0.01 mg/kg to about 10 mg/kg or about 0.1 mg/kg to about 5 mg/kg).


The concentration of a disclosed compound in a pharmaceutically acceptable mixture will vary depending on several factors, including the dosage of the compound to be administered, the pharmacokinetic characteristics of the compound(s) employed, and the route of administration. The agent may be administered in a single dose or in repeat doses. The dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. Treatments may be administered daily or more frequently depending upon a number of factors, including the overall health of a patient, and the formulation and route of administration of the selected compound(s). An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.


The compounds or pharmaceutical compositions of the present disclosure may be manufactured and/or administered in single or multiple unit dose forms.


Having now generally described the invention, the same will be more readily understood through reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present invention.







EXAMPLES

The disclosure now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention and are not intended to limit the invention.


Synthetic Preparation


The novel compounds of the present invention can be prepared in a variety of ways known to one skilled in the art of organic synthesis. The compounds of the present invention can be synthesized using the methods as hereinafter described below, together with synthetic methods known in the art of synthetic organic chemistry or variations thereon as appreciated by those skilled in the art.


Preparation of compounds can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene and Wuts, Protective Groups in Organic Synthesis, 44th. Ed., Wiley & Sons, 2006, as well as in Jerry March, Advanced Organic Chemistry, 4th edition, John Wiley & Sons, publisher, New York, 1992 which are incorporated herein by reference in their entirety.


Compounds of the present invention can be prepared by the literature methods cited in the following text. The following schemes depict established, known syntheses of these scaffolds.


The groups and/or the substituents of the compounds of the present invention can be synthesized and attached to these scaffolds by the literature methods cited in the following text. The following schemes depict the known techniques for accomplishing this joinder.


General Synthesis


Compounds of the present invention can be synthesized using the following methods. General reaction conditions are given, and reaction products can be purified by general known methods including crystallization, silica gel chromatography using various organic solvents such as hexane, cyclohexane, ethyl acetate, methanol and the like, preparative high pressure liquid chromatography or preparative reverse phase high pressure liquid chromatography.


Representative Synthesis


Example 1: Synthesis of N-((1r,3r)-3-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)cyclobutyl)methanesulfonamide (Compound A15)



embedded image


Tert-butyl ((1r,3r)-3-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)cyclobutyl)carbamate (2): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (inter J) (200 mg, 0.57 mmol) and tert-butyl ((1r,3r)-3-(hydroxymethyl)cyclobutyl)carbamate (1) (230 mg, 1.14 mmol) in THE (5 mL) was added PPh3 (300 mg, 1.14 mol) and DIAD (0.2 mL, 1.14 mmol) at 0° C. under N2 atmosphere. The mixture was stirred at 20° C. for 16 hrs under N2 atmosphere. LCMS showed the reaction was completed. The resulting mixture was quenched with H2O (5 mL). The aqueous layer was extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give tert-butyl ((1r,3r)-3-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)cyclobutyl)carbamate (2) (270 mg, yield: 79.8%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.45 (d, J=2.4 Hz, 1H), 7.33 (d, J=2.4 Hz, 1H), 7.08 (d, J=8.8 Hz, 2H), 6.84 (d, J=8.8 Hz, 2H), 4.75 (br s, 1H), 4.42 (t, J=6.2 Hz, 2H), 4.29 (br d, J=5.2 Hz, 1H), 3.98 (d, J=7.2 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 2.72-2.56 (m, 1H), 2.40-2.26 (m, 2H), 2.13-2.06 (m, 2H), 1.64 (s, 6H), 1.45 (s, 9H).


5-(2-(4-(((1r,3r)-3-aminocyclobutyl)methoxy)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (3): A solution of tert-butyl ((1 r,3r)-3-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)cyclobutyl)carbamate (2) (270 mg, 0.48 mmol) in TFA (0.4 mL) and DCM (2 mL) was stirred at 20° C. for 1 h. LCMS showed the reaction was completed. The reaction mixture was poured into water (5 mL) and then adjusted to pH=7-8 with sat.NaHCO3 and then extracted with DCM (5 mL×3). The combined organic layers were washed with brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 5-(2-(4-(((1r,3r)-3-aminocyclobutyl)methoxy)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (3) (184 mg, yield: 87.5%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.45 (d, J=2.0 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.09 (d, J=8.8 Hz, 2H), 6.85 (d, J=8.8 Hz, 2H), 5.25-4.94 (m, 2H), 4.42 (t, J=6.0 Hz, 2H), 3.96 (d, J=6.0 Hz, 2H), 3.89-3.80 (m, 3H), 2.85-2.80 (m, 1H), 2.40-2.34 (m, 2H), 2.30-2.25 (m, 2H), 1.64 (s, 6H).


N-((1r,3r)-3-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)cyclobutyl)methanesulfonamide (Compound A15): To a solution of 5 (2-(4-(((1r,3 r)-3-aminocyclobutyl)methoxy)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (3) (200 mg, 0.42 mmol) and N,N-diethylethanamine (0.1 mL, 0.84 mmol) in DCM (2 mL) was added methanesulfonyl chloride (95 mg, 0.84 mmol) at 0° C. The mixture was stirred for 2 hrs at 0-25° C. LCMS showed the reaction was completed. The reaction was quenched with water (5 mL) and extracted with DCM (5 mL×3). The combined organic layers were washed with brine (5×2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC(FA) to give N-((1r,3 r)-3-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)cyclobutyl)methanesulfonamide (Compound A15) (70 mg, yield: 32.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.45 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.0 Hz, 1H), 7.10 (d, J=8.8 Hz, 2H), 6.85 (d, J=8.4 Hz, 2H), 4.51 (br d, J=8.8 Hz, 1H), 4.42 (t, J=6.4 Hz, 2H), 4.24-4.10 (m, 1H), 3.99 (d, J=6.4 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 2.95 (s, 3H), 2.71-2.68 (m, 1H), 2.48-2.41 (m, 2H), 2.29-2.17 (m, 2H), 1.65 (s, 6H). LCMS (220 nm): 100.0%. Exact Mass: 510.0; found 533.1/535.1 (M+23).


Example 2: Synthesis of N-(6-(4-(3-chloro-4-(2-chloroethoxy)-5-cyanophenoxy)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A16)



embedded image


5-(4-bromophenoxy)-3-chloro-2-(2-chloroethoxy)benzonitrile (2): To a solution of (3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)boronic acid (inter S1) (3.1 g, 11.8 mmol), 4-bromophenol (1) (1.7 g, 9.83 mmol), Pyridine (1.5 g, 19.7 mmol) and 4 A M.S. (2.5 g) in DCE (60 mL) was added Cu(OAc)2 (1.7 g, 9.83 mmol) at 25° C. The mixture was stirred at 70° C. under 02 for 6 hrs. LCMS showed the reaction was completed. The mixture was quenched with water (100 mL) and extracted with DCM (50 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (FA) to give 5-(4-bromophenoxy)-3-chloro-2-(2-chloroethoxy)benzonitrile (2) (1.1 g, yield: 17.4%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.53 (d, J=8.8 Hz 2H), 7.26 (d, J=3.2 Hz, 1H), 7.08 (d, J=3.2 Hz, 1H), 6.92 (d, J=8.4 Hz 2H), 4.41 (t, J=6.0 Hz, 2H), 3.89 (t, J=6.0 Hz, 2H).


3-chloro-2-(2-chloroethoxy)-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy) benzonitrile (3): To a solution of 5-(4-bromophenoxy)-3-chloro-2-(2-chloroethoxy)benzonitrile (2) (0.9 g, 1.4 mmol) and Pin2B2 (0.53 g, 2.09 mmol) in 1,4-dioxane (10 mL) was added Pd(dppf)Cl2 (0.1 g, 0.14 mmol) and AcOK (0.41 g, 4.19 mmol) at 25° C. and the mixture was stirred at 100° C. for 3 hrs under N2 atmosphere. LCMS showed the reaction was completed. The mixture was quenched with H2O (30 mL). Then the mixture was extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-(2-chloroethoxy)-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)benzonitrile (3) (0.6 g, yield: 89.2%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=7.85 (d, J=8.4 Hz 2H), 7.26 (s, 1H), 7.10 (d, J=2.8 Hz, 1H), 7.01 (d, J=8.8 Hz 2H), 4.41 (t, J=6.0 Hz, 2H), 3.89 (t, J=6.0 Hz, 2H), 1.36 (s, 12H).


3-chloro-2-(2-chloroethoxy)-5-(4-(2-oxo-1,2-dihydroquinoxalin-6-yl)phenoxy)benzonitrile (5): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)benzonitrile (3) (300 mg, 0.62 mmol) and 6-bromobenzo[b][1,4]azaborinin-2(1H)-one (4) (140 mg, 0.62 mmol) in 1,4-dioxane (4 mL) and H2O (1 mL) was added dipotassium;carbonate (215 mg, 1.55 mmol) and Pd(dppf)Cl2 (46 mg, 0.06 mmol) under N2 at 25° C. The mixture was stirred for 4 hrs at 100° C. under N2. LCMS showed the reaction was completed. The mixture was quenched with water (10 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-(2-chloroethoxy)-5-(4-(2-oxo-1,2-dihydroquinoxalin-6-yl)phenoxy)benzonitrile (5) (360 mg, yield: 89.6%) as black solid. 1H-NMR (400 MHz, DMSO-d6) δ=12.54 (s, 1H), 8.21 (s, 1H), 8.05 (d, J=2.0 Hz, 1H), 7.89 (dd, J=2.0, 8.4 Hz, 1H), 7.83-7.77 (m, 2H), 7.64 (d, J=2.8 Hz, 1H), 7.60 (d, J=2.4 Hz, 1H), 7.41 (d, J=8.4 Hz, 1H), 7.19 (d, J=8.8 Hz, 2H), 4.42 (t, J=4.8 Hz, 2H), 3.98 (t, J=4.0 Hz, 2H).


3-chloro-2-(2-chloroethoxy)-5-(5-((3-fluoro-1-(methylsulfonyl)azetidin-3-yl)methoxy)-1H-indol-1-yl)benzonitrile (6): A solution of 3-chloro-2-(2-chloroethoxy)-5-(4-(2-oxo-1,2-dihydroquinoxalin-6-yl)phenoxy)benzonitrile (5) (360 mg, 0.55 mmol) in POCl3 (3 mL) and toluene (6 mL) was stirred at 110° C. for 1 h under N2 atmosphere. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was adjusted to pH=8-9 with sat.NaHCO3. The mixture was extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-(2-chloroethoxy)-5-(5-((3-fluoro-1-(methylsulfonyl) azetidin-3-yl)methoxy)-1H-indol-1-yl)benzonitrile (6) (18 mg, yield: 68.6%) as yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=9.03 (s, 1H), 8.41 (d, J=1.6 Hz, 1H), 8.28 (dd, J=2.0, 8.8 Hz, 1H), 8.13 (d, J=8.8 Hz, 1H), 7.98 (d, J=8.4 Hz, 2H), 7.70 (d, J=2.8 Hz, 1H), 7.66 (d, J=2.8 Hz, 1H), 7.25 (d, J=8.4 Hz, 2H), 4.43 (t, J=5.2 Hz, 2H), 3.99 (t, J=5.2 Hz, 2H).


N-(6-(4-(3-chloro-4-(2-chloroethoxy)-5-cyanophenoxy)phenyl)quinoxalin-2-yl) methanesulfonamide (Compound A16): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(5-((3-fluoro-1-(methyl sulfonyl)azetidin-3-yl)methoxy)-1H-indol-1-yl)benzonitrile (6) (130 mg, 0.27 mmol), methanesulfonamide (131 mg, 1.38 mmol) and Cs2CO3 (180 mg, 0.55 mmol) in 1,4-dioxane (3 mL) was added Pd2(dba)3 (51 mg, 0.05 mmol) and Xantphos (64 mg, 0.11 mmol) at 25° C. and the mixture was stirred at 90° C. for 3 hrs under N2. LCMS showed the reaction was completed. The mixture was poured into water (5 mL) and extracted with EtOAc (3 mL×3), the combined organic layers were washed with brine (3 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (FA) to give N-(6-(4-(3-chloro-4-(2-chloroethoxy)-5-cyanophenoxy)phenyl)quinoxalin-2-yl) methanesulfonamide (Compound A16) (22.3 mg, yield: 15.1%) as yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=11.63 (br s, 1H), 8.61 (s, 1H), 8.25 (s, 1H), 8.12 (dd, J=2.4, 8.8 Hz, 1H), 7.94-7.92 (m, 3H), 7.68 (d, J=3.2 Hz, 1H), 7.63 (d, J=2.8 Hz, 1H), 7.23 (d, J=8.8 Hz, 2H), 4.42 (t, J=4.8 Hz, 2H), 3.99 (t, J=5.2 Hz, 2H), 3.48 (s, 3H), LCMS (220 nm): 99.86%. Exact Mass: 528.04; found 528.9/530.9.


Example 3: Synthesis of N-(4-((4-(2-(3-chloro-5-cyano-4-methoxyphenyl)propan-2-yl) phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (Compound A17)



embedded image


3-chloro-5-(2-(4-hydroxyphenyl)propan-2-yl)-2-methoxybenzonitrile (2): To a solution of 3-chloro-2-hydroxy-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (1) (3.0 g, 10.4 mmol) and methanol (0.33 g, 10.4 mmol) in THF (30 mL) was added PPh3 (4.1 g, 15.6 mmol) and DIAD (3.1 mL, 15.6 mmol) at 20° C. The mixture was stirred at 25° C. for 16 hrs under N2 atmosphere. LCMS showed the reaction was completed. The reaction mixture was poured into H2O (20 mL) and then extracted with EtOAc (15 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel column chromatography to give 3-chloro-5-(2-(4-hydroxyphenyl)propan-2-yl)-2-methoxybenzonitrile (2) (2.7 g, yield: 85.0%) as colorless oil. 1H-NMR (400 MHz, CDCl3) δ=7.43 (d, J=2.4 Hz, 1H), 7.33 (d, J=2.4 Hz, 1H), 7.10-7.02 (m, 2H), 6.85-6.68 (m, 2H), 4.94-4.78 (m, 1H), 4.05 (s, 3H), 1.64 (s, 6H).


3-chloro-2-methoxy-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (3): To a solution of 3-chloro-5-(2-(4-hydroxyphenyl) propan-2-yl)-2-methoxybenzonitrile (2) (500 mg, 1.66 mmol) and 4-(chloromethyl)-2-(methylthio)pyrimidine (10a) (700 mg, 3.31 mmol) in DMF (8 mL) was added Cs2CO3 (2.16 g, 6.63 mmol) at 25° C. The mixture was stirred at 25° C. for 16 hrs. LCMS showed the reaction was completed. The reaction was quenched with water (10 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-methoxy-5-(2-(4-((2-(methylthio) pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (3) (250 mg, yield: 34.0%) as colorless oil. 1H-NMR (400 MHz, CHCl3) δ=8.09 (br s, 1H), 7.82-7.67 (m, 2H), 7.38 (t, J=6.4 Hz, 2H), 6.57-6.43 (m, 2H).


3-chloro-2-methoxy-5-(2-(4-((2-(methylsulfonyl)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (4): To a solution of 3-chloro-2-methoxy-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (3) (250 mg, 0.57 mmol) in THE (1.5 mL) and water (1.5 mL) was added Oxone (1.05 g, 1.70 mmol) at 25° C. and the mixture was stirred at 25° C. for 16 hrs. LCMS showed the reaction was completed. The resulting mixture was quenched with H2O (10 mL). The aqueous layer was extracted with EtOAc (10 mL×3). The combined organic layers was washed with brine (15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-methoxy-5-(2-(4-((2-(methylsulfonyl) pyrimidin-4-yl)methoxy) phenyl) propan-2-yl)benzonitrile (4) (200 mg, yield: 74.6%) as colorless oil. 1H-NMR (400 MHz, CDCl3) δ=8.94 (d, J=5.2 Hz, 1H), 7.85 (d, J=5.2 Hz, 1H), 7.44 (d, J=2.4 Hz, 1H), 7.30 (d, J=2.4 Hz, 1H), 7.18-7.10 (m, 2H), 6.91 (d, J=8.8 Hz, 2H), 5.30 (s, 2H), 4.06 (s, 3H), 3.40 (s, 3H), 1.65 (s, 6H).


N-(4-((4-(2-(3-chloro-5-cyano-4-methoxyphenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (Compound A17): To a solution of 3-chloro-2-methoxy-5-(2-(4-((2-(methyl sulfonyl)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (4) (200 mg, 0.42 mmol) and methanesulfonamide (0.20 g, 2.12 mmol) in DMF (3 mL) was added Cs2CO3 (276 mg, 0.85 mmol) at 20° C. and the mixture was stirred at 20° C. for 16 hrs under N2 atmosphere. LCMS showed the reaction was completed. The mixture was poured into water (20 mL) and extracted with EtOAc (10 mL×3), the combined organic layers were washed with brine (3 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (FA) to give N-(4-((4-(2-(3-chloro-5-cyano-4-methoxyphenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (Compound A17) (39.2 mg, yield: 18.8%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.73 (br s, 1H), 8.64 (d, J=5.2 Hz, 1H), 7.43 (d, J=2.0 Hz, 1H), 7.31 (d, J=2.0 Hz, 1H), 7.29 (d, J=5.2 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 6.90 (d, J=8.8 Hz, 2H), 5.11 (s, 2H), 4.06 (s, 3H), 3.48 (s, 3H), 1.65 (s, 6H) LCMS (220 nm): 94.7%. Exact Mass: 486.11; found 487.0/489.0.


Example 4: Synthesis of N-(6-(4-((3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A22)



embedded image


6-bromo-2-chloroquinoxaline (2): A solution of 6-bromoquinoxalin-2(1H)-one (1) (10.0 g, 44.4 mmol) in POCl3 (102 mL, 1.11 mol) and DMF (5 mL) was stirred at 100° C. for 2 hrs. LCMS showed the reaction was completed and the mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 6-bromo-2-chloroquinoxaline (2) (9.5 g, yield: 79.0%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=8.80 (s, 1H), 8.33 (s, 1H), 7.92 (s, 2H).


6-bromo-2-(methylthio)quinoxaline (3): To a solution of 6-bromo-2-chloroquinoxaline (2) (9.5 g, 35.1 mmol) in DMF (100 mL) was added NaSMe (2.9 g, 42.1 mmol) at 20° C., the mixture was stirred at 20° C. for 2 hrs. LCMS showed the reaction was completed. The mixture was quenched with H2O (100 mL) and extracted with EtOAc (80 mL×3). The combined organic layers were washed with brine (100 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 6-bromo-2-(methylthio)quinoxaline (3) (8.4 g, yield: 84.4%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.59 (s, 1H), 8.18 (d, J=2.0 Hz, 1H), 7.83-7.69 (m, 2H), 2.70 (s, 3H).


4-(2-(methylthio)quinoxalin-6-yl)aniline (5): To a mixture of 6-bromo-2-(methylthio)quinoxaline (3) (3.0 g, 11.8 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (4) (2.8 g, 12.9 mmol) in 1,4-dioxane (40 mL) and H2O (8 mL) was added disodium; carbonate (2.5 g, 23.5 mmol) and Pd(PPh3)4 (2.0 g, 1.76 mmol) at 20° C. under N2 and the mixture was stirred at 80° C. for 6 h under N2. LCMS showed the reaction was completed. The mixture was quenched with H2O (50 mL) and extracted with EtOAc (50 mL×3). The combined organic layers were washed with brine (150 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 4-(2-(methylthio)quinoxalin-6-yl)aniline (5) (3.0 g, yield: 76.3%). 1H-NMR (400 MHz, CDCl3) δ=8.61 (s, 1H) 8.20-8.10 (m, 1H), 7.01-8.89 (m, 2H), 7.57 (br d, J=8.4 Hz, 2H), 6.81 (br d, J=8.4 Hz, 2H), 3.83 (br s, 2H), 2.72 (s, 3H), 2.63-2.62 (m, 1H).


4-(2-(methylthio)quinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (6): To a solution of 4-(2-(methylthio)quinoxalin-6-yl)aniline (5) (1.0 g, 2.99 mmol)) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (3.5 g, 15 mmol) in DMF (10 mL) was added dicesium;carbonate (2.9 g, 8.98 mmol) at 25° C. and the mixture was stirred at 80° C. for 6 hrs under N2. LCMS showed ˜30% of starting material remained and ˜35% of desired product was detected. The mixture was poured into water (20 mL) and extracted with EtOAc (10 mL×3), the combined organic layers were washed with brine (10 mL×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 4-(2-(methylthio)quinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (6) (180 mg, yield: 15.5%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.62 (s, 1H), 8.22 (d, J=1.2 Hz, 1H), 8.03-7.93 (m, 2H), 7.71-7.56 (m, 2H), 6.83 (d, J=8.8 Hz, 2H), 2.81-2.67 (m, 3H).


3-chloro-2-(2-chloroethoxy)-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (7): To a solution of 5-bromo-3-chloro-2-(2-chloroethoxy)benzonitrile (inter Q) (244 mg, 0.8 mmol), 4-(2-(methylthio)quinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (6) (140 mg, 0.4 mol) and sodium; 2-methylpropan-2-olate (77 mg, 0.8 mmol) in toluene (3 mL) was added di-μ-bromobis(tri-tert-butylphosphine)dipalladium(I) (62 mg, 0.08 mmol) at 25° C. and the mixture was stirred at 100° C. for 3 hrs under N2 atmosphere. LCMS showed the reaction was completed. The mixture was poured into water (5 mL) and extracted with EtOAc (3 mL×3), the combined organic layers were washed with brine (3 mL×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-(2-chloroethoxy)-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (6) (200 mg, yield: 79.7%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.66 (s, 1H), 8.27 (d, J=2.0 Hz, 1H), 8.08-8.03 (m, 1H), 8.02-7.96 (m, 1H), 7.80 (d, J=8.4 Hz, 2H), 7.25 (d, J=8.8 Hz, 2H), 7.19 (d, J=3.2 Hz, 1H), 7.02 (d, J=3.2 Hz, 1H), 4.40 (t, J=6.0 Hz, 2H), 4.32 (q, J=8.4 Hz, 2H), 3.89 (t, J=6.0 Hz, 2H), 2.75 (s, 3H).


3-chloro-2-(2-chloroethoxy)-5-((4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl) (2,2,2-trifluoroethyl)amino)benzonitrile (8): To a solution of 3-chloro-2-(2-chloroethoxy)-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (7) (150 mg, 0.24 mmol) in THF (2 mL) and H2O (2 mL) was added Oxone (368 mg, 0.6 mmol) at 25° C. The mixture was stirred for 6 hrs at 25° C. under N2. LCMS showed the reaction was completed. The reaction was quenched with Na2S03 (5 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-(2-chloroethoxy)-5-((4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (8) (170 mg, yield: 95.3%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=9.54 (s, 1H), 8.44 (d, J=2.0 Hz, 1H), 8.33-8.28 (m, 1H), 8.24-8.19 (m, 1H), 7.86-7.81 (m, 2H), 7.28 (d, J=2.8 Hz, 1H), 7.26-7.21 (m, 2H), 7.11 (d, J=2.8 Hz, 1H), 4.43 (t, J=6.0 Hz, 2H), 4.38-4.30 (m, 2H), 3.90 (t, J=6.0 Hz, 2H), 3.43 (s, 3H).


N-(6-(44(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)(2,2,2-trifluoroethyl)amino) phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A22): To a solution of 3-chloro-2-(2-chloro ethoxy)-5-((4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (8) (170 mg, 0.26 mmol) in MeCN (2 mL) was added methanesulfonamide (73 mg, 0.77 mmol), dicesium;carbonate (251 mg, 0.77 mmol). The reaction was stirred at 35° C. for 6 hrs. LCMS showed the reaction was completed. The reaction was quenched with H2O (5 mL) and added HCl to adjust to pH=3-4, then the mixture was extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (neutral) to give N-(6-(4-((3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)(2,2,2-trifluoroethyl) amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A22) (44 mg, yield: 36%) as yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=8.51 (s, 1H), 8.21 (d, J=2.0 Hz, 1H), 8.09 (dd, J=2.0, 8.4 Hz, 1H), 7.90 (d, J=8.4 Hz, 2H), 7.85 (d, J=8.4 Hz, 1H), 7.43 (dd, J=2.8, 12.8 Hz, 2H), 7.30 (d, J=8.8 Hz, 2H), 4.77-4.73 (m, 2H), 4.38 (t, J=4.8 Hz, 2H), 3.96 (t, J=4.8 Hz, 2H), 3.38 (s, 3H). LCMS (220 nm): 100.00%. Exact Mass: 609.1; found 610.1/612.1.


Example 5: Synthesis of N-(1-(5-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)pyrimidin-2-yl)azetidin-3-yl)methanesulfonamide (Compound A36)



embedded image


3-chloro-2-(2-chloroethoxy)-5-(2-(4-(2-(methylthio)pyrimidin-5-yl)phenyl)propan-2-yl)benzonitrile (2): To a solution of 5-bromo-2-(methylthio)pyrimidine (1) (120 mg, 0.59 mmol) and 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propan-2-yl)benzonitrile (Inter E) (300 mg, 0.59 mmol) in 1,4-dioxane (4 mL) and H2O (1 mL) was added dipotassium;carbonate (203 mg, 1.47 mmol) and Pd(dppf)Cl2 (43 mg, 0.06 mmol) under N2 at 25° C. The mixture was stirred at 90° C. for 4 hrs under N2 atmosphere. LCMS showed the reaction was completed. The reaction solution was filtered. The filtrate was quenched with water (5 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(2-(methylthio)pyrimidin-5-yl)phenyl)propan-2-yl)benzonitrile (2) (240 mg, yield: 80.3%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.76 (s, 2H), 7.52-7.48 (m, 3H), 7.37 (d, J=2.0 Hz, 1H), 7.32 (d, J=8.4 Hz, 2H), 4.44 (t, J=6.0 Hz, 2H), 3.89 (t, J=6.0 Hz, 2H), 2.66-2.62 (m, 3H), 1.71 (s, 6H).


3-chloro-2-(2-chloroethoxy)-5-(2-(4-(2-(methylsulfonyl)pyrimidin-5-yl)phenyl)propan-2-yl)benzonitrile (3): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(2-(methylthio)pyrimidin-5-yl)phenyl)propan-2-yl)benzonitrile (2) (230 mg, 0.45 mmol) in THE (3 mL) and H2O (3 mL) was added Oxone (694 mg, 1.13 mmol) at 25° C. The mixture was stirred at 25° C. for 12 hrs under N2. LCMS showed the reaction was completed. The reaction was quenched with sat. Na2SO3 (5 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(2-(methylsulfonyl)pyrimidin-5-yl)phenyl)propan-2-yl)benzonitrile (3) (160 mg, yield: 65.0%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=9.11 (s, 2H), 7.60-7.57 (m, 2H), 7.49 (d, J=2.4 Hz, 1H), 7.40 (d, J=8.4 Hz, 2H), 7.36 (d, J=2.4 Hz, 1H), 4.47-4.44 (m, 2H), 3.92-3.87 (m, 2H), 3.42 (s, 3H), 1.73 (s, 6H).


tert-butyl (1-(5-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl) pyrimidin-2-yl)azetidin-3-yl)carbamate (5): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(2-(methyl sulfonyl)pyrimidin-5-yl)phenyl)propan-2-yl)benzonitrile (3) (160 mg, 0.29 mmol) and tert-butyl azetidin-3-ylcarbamate (4) (76 mg, 0.44 mmol) in MeCN (3 mL) was added dipotassium;carbonate (122 mg, 0.88 mmol) at 25° C. under N2. The mixture was stirred at 60° C. for 4 hrs under N2. LCMS showed the reaction was completed. The reaction was quenched with water (3 mL) and extracted with EtOAc (3 mL×3). The combined organic layers were washed with brine (5 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give tert-butyl (1-(5-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl) propan-2-yl)phenyl)pyrimidin-2-yl)azetidin-3-yl)carbamate (5) (150 mg, yield: 74.5%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.56 (s, 2H), 7.49 (d, J=2.0 Hz, 1H), 7.42 (d, J=8.4 Hz, 2H), 7.37 (d, J=2.4 Hz, 1H), 7.25 (s, 2H), 5.10-4.96 (m, 1H), 4.73-4.59 (m, 1H), 4.52 (br t, J=8.0 Hz, 2H), 4.43 (t, J=6.0 Hz, 2H), 4.00 (dd, J=4.8, 9.4 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 1.70 (s, 6H), 1.47 (s, 9H).


5-(2-(4-(2-(3-aminoazetidin-1-yl)pyrimidin-5-yl)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (6): To a mixture of tert-butyl (1-(5-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)pyrimidin-2-yl)azetidin-3-yl)carbamate (5) (150 mg, 0.26 mmol) in DCM (2 mL) was added 2,2,2-trifluoroacetic acid (0.5 mL, 6.69 mmol) at 0° C. and the mixture was stirred at 25° C. for 2 hrs. LCMS showed the reaction was completed. The mixture was quenched with sat. NaHCO3 (5 mL) and extracted with DCM (5 mL×3). The Combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 5-(2-(4-(2-(3-aminoazetidin-1-yl)pyrimidin-5-yl)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (6) (150 mg, yield: 96.6%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.58-8.53 (m, 2H), 7.48 (d, J=2.0 Hz, 1H), 7.40 (d, J=8.4 Hz, 2H), 7.37-7.35 (m, 1H), 7.24 (d, J=8.4 Hz, 2H), 4.54-4.40 (m, 4H), 4.12-4.04 (m, 1H), 3.98 (br dd, J=4.8, 8.8 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 1.71 (s, 6H).


N-(1-(5-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)pyrimidin-2-yl)azetidin-3-yl)methanesulfonamide (Compound A36): To a solution of 5-(2-(4-(2-(3-aminoazetidin-1-yl)pyrimidin-5-yl)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy) benzonitrile (6) (150 mg, 0.28 mmol) and N,N-diethylethanamine (0.1 mL, 0.56 mmol) in DCM (3 mL) was added methanesulfonyl chloride (32 mg, 0.28 mmol) at 0° C. and stirred at 20° C. for 2 hrs. LCMS showed the reaction was completed. The mixture was quenched with water (10 mL) and extracted with DCM (5 mL×3). The combined organic layers were washed with brine (10 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (neutral) to give N-(1-(5-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)pyrimidin-2-yl)azetidin-3-yl)methanesulfonamide (Compound A36) (86 mg, yield: 54.0%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=8.58 (s, 2H), 7.49 (d, J=2.4 Hz, 1H), 7.43 (d, J=8.4 Hz, 2H), 7.36 (d, J=1.2 Hz, 1H), 7.28 (s, 2H), 4.93 (br d, J=8.8 Hz, 1H), 4.59 (t, J=8.4 Hz, 2H), 4.54-4.47 (m, 1H), 4.44 (t, J=6.0 Hz, 2H), 4.10 (dd, J=5.2, 9.6 Hz, 2H), 3.89 (t, J=6.0 Hz, 2H), 3.03 (s, 3H), 1.70 (s, 6H). LCMS: (220 nm): 98.6%. Exact Mass: 559.1; found 560.1/562.1.


Example 6: Synthesis of N-((1r,3r)-3-(4-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl) propan-2-yl)phenyl)-1H-pyrazol-1-yl)cyclobutyl)methanesulfonamide (Compound A44)



embedded image


(1s,3s)-3-((tert-butoxycarbonyl)amino)cyclobutyl 4-methylbenzenesulfonate (2): To a solution of tert-butyl ((1s,3s)-3-hydroxycyclobutyl)carbamate (1) (2.0 g, 10.7 mmol) in DCM (20 mL) was added pyridine (1.7 mL, 21.4 mmol), N,N-dimethylpyridin-4-amine (0.26 g, 2.14 mmol) and 4-methylbenzenesulfonyl chloride (2.6 g, 13.9 mmol) at 0° C. and the mixture was stirred for 2 hrs at 25° C. under N2 atmosphere. TLC showed the reaction was completed. The reaction was quenched with H2O (5 mL) and extracted with DCM (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude product was purified by silica gel column chromatography to give (1s,3s)-3-((tert-butoxycarbonyl)amino)cyclobutyl 4-methylbenzenesulfonate (2) (3.0 g, yield: 74.0%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=7.78 (d, J=8.4 Hz, 2H), 7.35 (d, J=8.0 Hz, 2H), 4.62 (br s, 1H), 4.50 (t, J=7.2 Hz, 1H), 3.73 (br d, J=6.4 Hz, 1H), 2.77-2.63 (m, 2H), 2.46 (s, 3H), 2.08-1.92 (m, 2H), 1.41 (s, 9H).


tert-butyl ((1r,3r)-3-(4-bromo-1H-pyrazol-1-yl)cyclobutyl)carbamate (4): To a solution of 4-bromo-1H-pyrazole (3) (0.90 g, 6.12 mmol) in DMF (10 ml) was added NaH (60.0% purity, 0.35 g, 9.19 mmol) 0° C. under N2 and the mixture was stirred at 0° C. for 15 min under N2 atmosphere, then a solution of (1s,3s)-3-((tert-butoxycarbonyl)amino)cyclobutyl 4-methylbenzenesulfonate (2) (2.5 g, 6.74 mmol) in DMF (20 mL) was added drop-wise at 0° C. under N2 atmosphere. The mixture was stirred 50° C. under N2 for 2 hrs. TLC showed the reaction was completed. The mixture was quenched with sat. NH4Cl (20 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 ml×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by HPLC (neutral) to give tert-butyl ((1r,3r)-3-(4-bromo-1H-pyrazol-1-yl)cyclobutyl)carbamate (4) (0.40 g, yield: 18.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.48 (d, J=14.0 Hz, 2H) 4.92-4.82 (m, 1H) 4.31 (br d, J=2.4 Hz, 1H) 2.92-2.80 (m, 2H) 2.53 (br d, J=4.8 Hz, 2H) 1.46 (s, 9H).


tert-butyl ((1r,3r)-3-(4-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl) propan-2-yl)phenyl)-1H-pyrazol-1-yl)cyclobutyl)carbamate (5): To a mixture of tert-butyl ((1r,3r)-3-(4-bromo-1H-pyrazol-1-yl)cyclobutyl)carbamate (4) (1.0 g, 1.58 mmol) and 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propan-2-yl)benzonitrile (inter E) (809 mg, 1.58 mmol) in 1,4-dioxane (10 mL) and H2O (2 mL) was added dipotassium;carbonate (546 mg, 3.95 mmol) and Pd(dppf)Cl2 (116 mg, 0.16 mmol) under N2 at 25° C. The mixture was stirred for 8 hrs at 100° C. under N2 atmosphere. LCMS showed the reaction was completed. The mixture was quenched with water (10 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give tert-butyl ((1r,3 r)-3-(4-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)-1H-pyrazol-1-yl)cyclobutyl)carbamate (5) (400 mg, yield: 32.9%) as colorless oil. 1H-NMR (400 MHz, CDCl3) δ 7.79 (s, 1H), 7.68 (s, 1H), 7.46 (d, J=2.4 Hz, 1H), 7.41 (d, J=8.4 Hz, 2H), 7.36 (d, J=2.4 Hz, 1H), 7.17 (d, J=8.4 Hz, 2H), 4.96-4.87 (m, 2H), 4.42 (t, J=6.0 Hz, 2H), 3.87 (t, J=6.0 Hz, 2H), 2.94-2.89 (m, 2H), 2.56 (br s, 2H), 1.67 (s, 6H), 1.46 (s, 9H).


5-(2-(4-(1-((1r,3r)-3-aminocyclobutyl)-1H-pyrazol-4-yl)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (6): To a solution of tert-butyl ((1r,3r)-3-(4-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)-1H-pyrazol-1-yl)cyclobutyl)carbamate (5) (200 mg, 0.32 mmol) in DCM (2 mL) was added TFA (1 mL) at 25° C. and the mixture was stirred at 25° C. for 1 h. LCMS showed the starting material was consumed and the desired product was detected. The reaction was adjusted to pH=7-8 with sat. NaHCO3, and then extracted with DCM (5 mL×3). The combined organic layers were washed with brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 5-(2-(4-(1-((l r,3 r)-3-aminocyclobutyl)-1H-pyrazol-4-yl)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (6) (250 mg, yield: 92.7%) as colorless oil. 1H-NMR (400 MHz, CDCl3) δ=7.80 (s, 1H), 7.66 (s, 1H), 7.46 (d, J=2.4 Hz, 1H), 7.42 (d, J=8.4 Hz, 2H), 7.36 (d, J=2.4 Hz, 1H), 7.17 (d, J=8.4 Hz, 2H), 5.06-4.97 (m, 1H), 4.43 (t, J=6.0 Hz, 2H) 3.99-3.92 (m, 1H), 3.88 (t, J=6.0 Hz, 2H), 2.93-2.81 (m, 2H), 2.39 (ddd, J=12.8, 8.4, 4.8 Hz, 2H), 1.68 (s, 6H).


N-((1r,3r)-3-(4-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)-1H-pyrazol-1-yl)cyclobutyl)methanesulfonamide (Compound A44): To a solution of 5-(2-(4-(1-((1r,3 r)-3-aminocyclobutyl)-1H-pyrazol-4-yl)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (6) (250 mg, 0.48 mmol) in DCM (3 mL) was added N,N-diethylethanamine (1.0 mL, 7.17 mmol), methanesulfonyl chloride (49 mg, 0.43 mmol) at 0° C. The mixture was stirred for 1 h at 25° C. LCMS showed the reaction was completed. The reaction was quenched with water (5 mL), extracted with DCM (5 mL×3). The combined organic layers were washed with brine (2 mL×5), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (FA) to give N-((1r,3r)-3-(4-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenyl)-1H-pyrazol-1-yl)cyclobutyl)methanesulfonamide (Compound A44) (60 mg, yield: 59%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=7.82 (s, 1H), 7.66 (s, 1H), 7.47 (d, J=2.4 Hz, 1H), 7.42 (d, J=8.4 Hz, 2H), 7.36 (d, J=2.4 Hz, 1H), 7.18 (d, J=8.4 Hz, 2H), 4.96-4.86 (m, 1H), 4.78 (d, J=7.2 Hz, 1H), 4.49-4.33 (m, 3H), 3.88 (t, J=6.0 Hz, 2H), 3.11-2.93 (m, 5H), 2.70-2.60 (m, 2H), 1.68 (s, 6H). LCMS (220 nm): 99.01%. Exact Mass: 546.1; found 547.1/549.1.


Example 7: Synthesis of N-(4-amino-5-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (Compound A118)



embedded image


embedded image


methyl 2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidine-5-carboxylate (2): To a solution of methyl 2,4-dichloropyrimidine-5-carboxylate (60.0 g, 290 mmol) in THF (200 mL) was added (2,4-dimethoxyphenyl)methanamine (58.2 g, 348 mmol) and DIEA (99.2 mL, 580 mmol). The mixture was stirred at 15° C. for 12 hours. LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to remove solvent. The crude product was purified by column chromatography on silica gel to give methyl 2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidine-5-carboxylate (97.0 g, yield: 99.1%) as a white solid.


[2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidin-5-yl]methanol (3): To a solution of methyl 2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidine-5-carboxylate (56 g, 14.8 mmol) in THF (1000 mL) was added LiAlH4 (0.843 g, 22.2 mmol). The mixture was stirred at −78° C. for 2 hours. LCMS showed the reaction was completed. The reaction mixture was quenched with water (200 mL) and extracted with EA (800 mL). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude was purified by silica gel column to give [2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidin-5-yl]methanol (27 g, yield: 52.6%) as a yellow solid.


2-chloro-5-(chloromethyl)-N-[(2,4-dimethoxyphenyl)methyl]pyrimidin-4-amine (4): To a solution of [2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidin-5-yl]methanol (22.0 g, 71.0 mmol) in DCM (300 mL) was added 4-methylbenzenesulfonyl chloride (16.2 g, 85.2 mmol) and TEA (14.8 mL, 107 mmol). The mixture was stirred at 25° C. for 12 hours. LCMS showed the reaction was completed. The reaction mixture was quenched with water (200 mL) and extracted with EA (300 mL). The combined organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude was purified by silica gel column to give 2-chloro-5-(chloromethyl)-N-[(2,4-dimethoxyphenyl)methyl]pyrimidin-4-amine (9.0 g, yield: 30.0%) as a yellow solid.


3-chloro-5-[1-[4-[[2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidin-5-yl]methoxy]phenyl]-1-methyl-ethyl]-2-(2-chloroethoxy)benzonitrile (6): To a solution of 2-chloro-5-(chloromethyl)-N-[(2,4-dimethoxyphenyl)methyl]pyrimidin-4-amine (9.0 g, 27.4 mmol) and 3-chloro-2-(2-chloroethoxy)-5-[1-(4-hydroxyphenyl)-1-methyl-ethyl]benzonitrile (11.5 g, 32.9 mmol) in CH3CN (200 mL) was added K2CO3 (5.68 g, 41.1 mmol) at 25° C. Then the mixture was stirred at 25° C. for 3 hours. LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to remove solvent. The reaction mixture was diluted with water (200 mL) and extracted with EA (300 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude was purified by silica gel column to give 3-chloro-5-[1-[4-[[2-chloro-4-[(2,4-dimethoxyphenyl)methyl amino]pyrimidin-5-yl]methoxy]phenyl]-1-methyl-ethyl]-2-(2-chloroethoxy)benzonitrile (11.0 g, yield: 62.5%) as a yellow solid.


tert-butyl N-[2-chloro-5-[[4-[1-[3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl]-1-methyl-ethyl]phenoxy]methyl]pyrimidin-4-yl]-N-[(2,4-dimethoxyphenyl)methyl]carbamate (7): To a solution of 3-chloro-5-[1-[4-[[2-chloro-4-[(2,4-dimethoxyphenyl)methylamino]pyrimidin-5-yl]methoxy]phenyl]-1-methyl-ethyl]-2-(2-chloroethoxy)benzonitrile (11.0 g, 17.1 mmol), TEA (5.20 g, 51.4 mmol), DMAP (1.05 g, 8.57 mmol) in THE (200 mL) was added Di-tert-butyl dicarbonate (18.7 g, 85.7 mmol) at 0° C. Then the mixture was stirred at 25° C. for 12 hours. LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to remove solvent. The crude was purified by silica gel column to give tert-butyl N-[2-chloro-5-[[4-[1-[3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl]-1-methyl-ethyl]phenoxy]methyl]pyrimidin-4-yl]-N-[(2,4-dimethoxyphenyl)methyl]carbamate (12.0 g, yield: 94.4%) as a yellow oil.


tert-butyl N-[5-[[4-[1-[3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl]-1-methyl-ethyl]phenoxy]methyl]-2-(methanesulfonamido)pyrimidin-4-yl]-N-[(2,4-dimethoxyphenyl)methyl]carbamate (8): To a solution of tert-butyl N-[2-chloro-5-[[4-[1-[3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl]-1-methyl-ethyl]phenoxy]methyl]pyrimidin-4-yl]-N-[(2,4-dimethoxyphenyl)methyl]carbamate (9.0 g, 12.1 mmol) in dioxane (200.0 mL) was added methanesulfonamide (3.46 g, 36.4 mmol), Cs2CO3 (11.9 g, 36.4 mmol), Xantphos (1.05 g, 1.82 mmol) and Tris(dibenzylideneacetone)dipalladium(0) (2.09 g, 3.64 mmol). The mixture was stirred at 100° C. for 12 hours. LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure. The crude was purified by silica gel column to give tert-butyl N-[5-[[4-[l-[3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl]-1-methyl-ethyl]phenoxy]methyl]-2-(methanesulfonamido)pyrimidin-4-yl]-N-[(2,4-dimethoxyphenyl)methyl]carbamate (7.5 g, yield: 77.2%) as a yellow oil.


N-(4-amino-5-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (Compound A118): To a solution of tert-butyl N-[5-[[4-[1-[3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl]-1-methyl-ethyl]phenoxy]methyl]-2-(methanesulfonamido)pyrimidin-4-yl]-N-[(2,4-dimethoxyphenyl)methyl]carbamate (3.0 g, 3.75 mmol) in DCM (30.0 mL) was added TFA (10.0 mL) at 25° C. Then the mixture was stirred at 40° C. for 18 hours. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure. The impure product was purified by Prep-HPLC (FA) to give N-(4-amino-5-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (900 mg, yield: 46.3%) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=7.84-7.81 (m, 1H), 7.63-7.61 (m, 1H), 7.57-7.54 (m, 1H), 7.21-7.15 (m, 2H), 6.96-6.89 (m, 2H), 4.87-4.74 (m, 2H), 4.45-4.37 (m, 2H), 3.99-3.91 (m, 2H), 3.12-2.99 (m, 3H), 1.69-1.56 (m, 6H). M+H: 550.1 (calc.); found 550.3 (LC-MS).


Example 8: Synthesis of N-(6-(4-((3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)(cyclopropyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A80)



embedded image


embedded image


embedded image


Synthesis of 5-bromo-3-chloro-2-(2-chloroethoxy)benzonitrile (Inter Q):


5-bromo-3-chloro-2-hydroxybenzaldehyde (2a): To a solution of 4-bromo-2-chloro-phenol (1a) (150 g, 0.72 mol) in TFA (1500 mL) was added Hexamethylenetetramine (203 g, 1.45 mol) at 20° C. The mixture was stirred at 100° C. for 16 h under N2 atmosphere. The reaction was quenched with water (5 L) and 50% H2SO4 (3 L) and stirred at 20° C. for 30 min. The mixture was filtered and the filter cake was concentrated under reduced pressure to give 5-bromo-3-chloro-2-hydroxybenzaldehyde (2a) (152 g, yield: 80.0%) as yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=11.27-11.20 (m, 1H), 10.11 (s, 1H), 7.98 (d, J=2.4 Hz, 1H), 7.83 (d, J=2.4 Hz, 1H).


5-bromo-3-chloro-2-hydroxybenzonitrile (3a): To a solution of 5-bromo-3-chloro-2-hydroxybenzaldehyde (2a) (152 g, 0.65 mol) in HCOOH (1500 mL) was added HCl-NH2OH (89.7 g, 1.29 mol) and HCOONa (87.8 g, 1.29 mol) at 20° C. and stirred at 100° C. for 16 h. The resulting mixture was quenched with H2O (5000 mL) (lots of solid appeared). The suspension was filtered and the filter cake was concentrated under reduced pressure to give 5-bromo-3-chloro-2-hydroxybenzonitrile (3a) (106.5 g, yield: 63.7%) as an off-white solid. 1H-NMR (400 MHz, CDCl3) δ=7.71 (s, 1H), 7.61 (s, 1H).


5-bromo-3-chloro-2-(2-chloroethoxy)benzonitrile (inter Q): To a solution of 5-bromo-3-chloro-2-hydroxybenzonitrile (3a) (188 g, 0.81 mol) in DMF (2000 mL) was added 1-bromo-2-chloroethane (4a) (348 g, 2.43 mol) and Cs2O3 (659 g, 2.02 mol) at 20° C. and stirred at 80° C. for 16 h under N2. The mixture was quenched with water (5000 mL) and extracted with EtOAc (2000 mL×3). The combined organic layers were washed with brine (2000 mL×5), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with PE/MTBE=3/1 (500 mL) to give 5-bromo-3-chloro-2-(2-chloroethoxy)benzonitrile (inter Q) (215 g, yield: 90.6%) as an off-white solid. 1H-NMR (400 MHz, CDCl3) δ=7.78 (d, J=2.4 Hz, 1H), 7.64 (d, J=2.4 Hz, 1H), 4.46 (t, J=6.0 Hz, 2H), 3.89 (t, J=6.0 Hz, 2H).


Synthesis of Compound A80:


6-bromo-2-methylsulfanyl-quinoxaline (inter N): To a solution of 6-bromo-2-chloro-quinoxaline (8.0 g, 0.0312 mol) in DMF (100 mL) was added NaSMe (2.63 g, 0.0375 mol) at 20° C. The reaction was stirred at 20° C. for 2 h. The mixture was quenched with water (100 mL) and extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (200×2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column to give 6-bromo-2-methylsulfanyl-quinoxaline (6.0 g, yield: 77.5%) as a white solid. 1H-NMR (400 MHz, CDCl3) δ=8.59 (s, 1H), 8.18 (d, J=2.0 Hz, 1H), 7.83-7.69 (m, 2H), 2.70 (s, 3H).


4-(2-(methylthio)quinoxalin-6-yl)phenol (3): To a solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (2) (4.3 g, 17.6 mmol) and 6-bromo-2-methylsulfanyl-quinoxaline (inter N) (5 g, 17.6 mmol) in 1,4-dioxane (50 mL) and H2O (12.5 mL) was added K2CO3 (4.88 g, 35.3 mmol) and Pd(dppf)Cl2 (1.29 g, 1.76 mmol) at 25° C. under N2 and the mixture was stirred at 90° C. for 3 h under N2. The reaction was quenched with water (60 mL) and extracted with EtOAc (15 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 4-(2-methylsulfanylquinoxalin-6-yl)phenol (3.3 g, yield: 54.5%) as a yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=9.70 (s, 1H), 8.78 (s, 1H), 8.14 (s, 1H), 8.04 (br d, J=8.8 Hz, 1H), 7.93 (br d, J=8.4 Hz, 1H), 7.68 (br d, J=8.4 Hz, 2H), 6.91 (br d, J=8.4 Hz, 2H), 2.67 (s, 3H).


4-(2-(methylthio)quinoxalin-6-yl)phenyl trifluoromethanesulfonate (4): To a solution of 4-(2-(methylthio)quinoxalin-6-yl)phenol (3) (1.3 g, 2.91 mmol) and Py (460 mg, 5.81 mmol) in DCM (10 mL) was added Tf2O (1.23 g, 4.36 mmol) at 0° C. and the mixture was stirred at 20° C. for 1 h. The mixture was poured into sat. aq. NH4Cl (10 mL) and extracted with DCM (10 mL×3). The combined organic layers were washed with brine (10 mL×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column chromatography to give 4-(2-(methylthio)quinoxalin-6-yl)phenyl trifluoromethanesulfonate (4) (1.0 g, yield: 85.1%) as a yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=8.87 (s, 1H), 8.34 (d, J=2.4 Hz, 1H), 8.16 (dd, J=2.4, 8.8 Hz, 1H), 8.09-8.06 (d, J=8.8 Hz, 2H, 8.05-8.01 (m, 1H), 7.65 (d, J=8.8 Hz, 2H), 2.70 (s, 3H).


N-cyclopropyl-4-(2-(methylthio)quinoxalin-6-yl)aniline (6): To a solution of 4-(2-(methylthio)quinoxalin-6-yl)phenyl trifluoromethanesulfonate (4) (1 g, 2.5 mmol) and cyclopropanamine (5) (1.25 g, 21.9 mmol) in 1,4-dioxane (10 mL) was added Cs2CO3 (3.05 g, 9.37 mmol) and t-BuBrettPhos-Pd-G3 (213 mg, 0.25 mmol) at 20° C. under N2. The mixture was stirred at 70° C. under N2 atmosphere for 8 h. The reaction mixture was diluted with H2O (30 mL) and extracted with EtOAc (15 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give N-cyclopropyl-4-(2-(methylthio)quinoxalin-6-yl)aniline (6) (1.20 g, yield: 93.8%) as a yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ=8.78 (s, 1H), 8.11 (d, J=2.0 Hz, 1H), 8.08-8.04 (m, 1H), 7.93 (d, J=8.8 Hz, 1H), 7.65 (d, J=8.8 Hz, 2H), 6.83 (d, J=8.8 Hz, 2H), 6.39 (s, 1H), 2.68 (s, 3H), 2.44-2.35 (m, 1H), 0.75-0.70 (m, 2H), 0.43-0.40 (m, 2H).


3-chloro-2-(2-chloroethoxy)-5-(cyclopropyl(4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino) benzonitrile (7): To a solution of N-cyclopropyl-4-(2-(methylthio)quinoxalin-6-yl)aniline (6) (300 mg, 0.976 mmol), 5-bromo-3-chloro-2-(2-chloroethoxy)benzonitrile (inter Q) (576 mg, 1.95 mmol) and t-BuONa (281 mg, 2.93 mmol) in toluene (3 mL) was added (t-Bu3PPdBr)2 (152 mg, 0.2 mmol) at 25° C. and the mixture was stirred at 100° C. for 3 h under N2. The reaction mixture was diluted with H2O (10 mL). The resulting aqueous phase was extracted with EtOAc (3×5 mL). The combined organic layers were washed with brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-(2-chloroethoxy)-5-(cyclopropyl(4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (7) (100 mg, yield: 17.7%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.65 (s, 1H), 8.24 (s, 1H), 8.06-7.95 (m, 2H), 7.77 (d, J=8.4 Hz, 2H), 7.30 (d, J=3.2 Hz, 1H), 7.23 (d, J=8.4 Hz, 2H), 7.16 (d, J=2.8 Hz, 1H), 4.37 (t, J=6.4 Hz, 2H), 3.90 (t, J=6.4 Hz, 2H), 2.82-2.77 (m, 1H), 2.74 (s, 3H), 1.02-0.97 (m, 2H), 0.72-0.70 (m, 2H).


3-chloro-2-(2-chloroethoxy)-5-(cyclopropyl(4-(2-(methylsulfonyl)quinoxalin-6-yl) phenyl) amino)benzonitrile (8): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(cyclopropyl(4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (7) (100 mg, 0.18 mmol) in THE (2 mL) and H2O (2 mL) was added Oxone (560 mg, 0.911 mmol) at 25° C. The mixture was stirred for 6 h at 35° C. The reaction was quenched with water (5 mL), extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-(2-chloroethoxy)-5-(cyclopropyl(4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)amino)benzonitrile (8) (100 mg, yield: 89.3%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=9.53 (s, 1H), 8.43 (d, J=1.6 Hz, 1H), 8.33-8.26 (m, 1H), 8.26-8.19 (m, 1H), 7.81 (d, J=6.8 Hz, 2H), 7.37 (d, J=3.2 Hz, 1H), 7.30-7.25 (m, 2H), 7.21 (d, J=2.8 Hz, 1H), 4.40 (t, J=6.4 Hz, 2H), 3.91 (t, J=6.4 Hz, 2H), 3.42 (s, 3H), 2.86-2.78 (m, 1H), 1.07-0.99 (m, 2H), 0.77-0.68 (m, 2H).


N-(6-(4-((3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)(cyclopropyl)amino)phenyl) quinoxalin-2-yl)methanesulfonamide (Compound A80): To a solution of 3-chloro-2-(2-chloroethoxy)-5-(cyclopropyl(4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)amino)benzonitrile (8) (100 mg, 0.16 mmol) and MsNH2 (61.9 mg, 0.65 mmol) in MeCN (3 mL) was added Cs2CO3 (159 mg, 0.49 mmol) at 20° C. under N2. The mixture was stirred for 8 h at 35° C. The mixture was quenched with water (10 mL), extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (neutral) to give N-(6-(4-((3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)(cyclopropyl)amino)phenyl)quinoxalin-2-yl) methanesulfonamide (21.4 mg, yield: 14.4%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.45-8.40 (m, 1H), 8.18 (br s, 1H), 7.92 (br d, J=8.8 Hz, 1H), 7.70 (br d, J=8.4 Hz, 2H), 7.62-7.40 (m, 1H), 7.31 (d, J=2.8 Hz, 1H), 7.22 (d, J=8.8 Hz, 2H), 7.15 (d, J=2.8 Hz, 1H), 4.37 (t, J=6.4 Hz, 2H), 3.89 (t, J=6.4 Hz, 2H), 3.26 (br s, 3H), 2.81-2.78 (m, 1H), 1.00-0.97 (m, 2H), 0.72-0.68 (m, 2H). LCMS (220 nm): 96.93%. Exact Mass: 567.09; found 568.0/570.0.


Example 9: Synthesis of N-(6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy) phenyl) quinoxalin-2-yl) methanesulfonamide (Compound A97)



embedded image


embedded image


4-(4-bromophenoxy)-2-chloro-1-fluorobenzene (3): To a solution of (3-chloro-4-fluorophenyl) boronic acid (1) (100 g, 0.57 mol), 4-bromophenol (2) (119 g, 0.69 mol), Pyridine (136 g, 1.72 mol) and 4 A molecular sieve (101 g) in DCE (1500 mL) was added Cu(OAc)2 (156 g, 0.86 mol) at 25° C. The mixture was stirred at 70° C. under 02 ball for 6 h. The reaction was filtered by Celite and to the filtrate was added water (1000 mL) then extracted with DCM (800 mL×3). The combined organic layers were washed with brine (1000 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 4-(4-bromophenoxy)-2-chloro-1-fluorobenzene (3) (55.0 g, yield: 28.6%) as a yellow oil. 1H-NMR (400 MHz, CDCl3) δ=7.46 (d, J=8.8 Hz, 2H), 7.12 (t, J=8.8 Hz, 1H), 7.05 (dd, J=28.0, 6.0 Hz, 1H), 6.92-6.85 (m, 3H).


5-(4-bromophenoxy)-3-chloro-2-fluorobenzaldehyde (4): To a solution of 4-(4-bromophenoxy)-2-chloro-1-fluorobenzene (3) (55 g, 0.16 mol) in THE (550 mL) was added LDA (2 mol/L, 98.5 mL, 0.19 mol) drop wise at −70° C. under N2. The mixture was stirred at 70° C. for 45 min. Then DMF (24 g, 0.33 mol) was added dropwise into the reaction and the mixture was stirred for another 1 h at −70° C. under N2. The mixture was quenched with sat. NH4Cl (200 mL) and extracted with EtOAc (200 mL×3). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 5-(4-bromophenoxy)-3-chloro-2-fluorobenzaldehyde (4) (13.0 g, yield: 21.6%) as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ=10.13 (s, 1H), 7.76 (dd, J=3.2, 6.0 Hz, 1H), 7.61 (d, J=8.8 Hz, 2H), 7.33 (dd, J=3.2, 4.8 Hz, 1H), 7.08 (d, J=8.8 Hz, 2H).


5-(4-bromophenoxy)-3-chloro-2-fluorobenzonitrile (inter Z): To a solution of 5-(4-bromophenoxy)-3-chloro-2-fluorobenzaldehyde (4) (13 g, 0.04 mol) in HCOOH (130 mL) was added HCOONa (5.10 g, 0.08 mol) and HCl—NH2OH (5.21 g, 0.08 mol) at 20° C. The mixture was stirred at 100° C. for 8 h under N2. The resulting mixture was quenched with H2O (100 mL) and extracted with EtOAc (50 mL×3). The combined organic layers were washed with sat. NaHCO3 (50 mL×2) and brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 5-(4-bromophenoxy)-3-chloro-2-fluorobenzonitrile (inter Z) (12.5 g, yield: 91.9%) as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ=7.79 (dd, J=3.2, 6.0 Hz, 1H), 7.73 (dd, J=3.2, 4.8 Hz, 1H), 7.60 (d, J=8.8 Hz, 2H), 7.07 (d, J=8.8 Hz, 2H).


5-(4-bromophenoxy)-3-chloro-2-cyclopropoxybenzonitrile (6): To a solution of cyclopropanol (a) (1.34 g, 23.2 mmol) in DMF (100 ml) was added NaH (60.0% purity, 1.48 g, 38.6 mmol) at 0° C. under N2 and the mixture was stirred at 0° C. for 30 min, then a solution of 5-(4-bromophenoxy)-3-chloro-2-fluorobenzonitrile (inter Z) (7.0 g, 19.3 mmol) in DMF (100 mL) was added dropwise at 0° C. under N2. The mixture was stirred 20° C. under N2 for 2 h. The mixture was quenched with sat. NH4Cl (200 mL) and then extracted with EtOAc (200 mL×3). The combined organic layers were washed with brine (500 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column chromatography to give 5-(4-bromophenoxy)-3-chloro-2-cyclopropoxybenzonitrile (6) (4.80 g, yield: 61.4%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.53-7.50 (m, 2H), 7.25 (d, J=3.2 Hz, 1H), 7.08 (d, J=3.2 Hz, 1H), 6.91 (dd, J=2.4, 7.2 Hz, 2H), 4.46 (t d, J=3.6, 6.4 Hz, 1H), 1.03 (d, J=2.8 Hz, 2H), 0.71 (d, J=7.2 Hz, 2H).


3-chloro-2-cyclopropoxy-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy) benzonitrile (7): To a solution of 5-(4-bromophenoxy)-3-chloro-2-cyclopropoxybenzonitrile (6) (4.3 g, 10.6 mmol) and Pin2B2 (2.97 g, 11.7 mmol)) in 1,4-dioxane (100 mL) was added Pd(dppf)Cl2 (0.777 g, 1.06 mmol) and AcOK (2.08 g, 21.2 mmol) at 25° C. under N2. The mixture was stirred at 90° C. under N2 for 3 h. The reaction was quenched with H2O (10 mL) and extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-cyclopropoxy-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenoxy) benzonitrile (7) (3.14 g, yield: 64.7%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.85 (d, J=8.4 Hz, 2H), 7.26 (d, J=2.8 Hz, 1H), 7.09 (d, J=2.8 Hz, 1H), 7.01 (d, J=8.8 Hz, 2H), 4.45 (tt, J=2.8, 6.0 Hz, 1H), 1.36 (s, 12H), 1.09-0.99 (m, 2H), 0.73-0.65 (m, 2H).


3-chloro-2-cyclopropoxy-5-(4-(2-(methylthio)quinoxalin-6-yl)phenoxy)benzonitrile (8): To a solution of 3-chloro-2-cyclopropoxy-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)benzonitrile (7) (2.6 g, 5.68 mmol) and 6-bromo-2-(methylthio)quinoxaline (inter N) (1.93 g, 6.82 mmol) in 1,4-dioxane (40 mL) and H2O (8 mL) was added K2CO3 (2.36 g, 17.1 mmol) and Pd(dppf)Cl2 (0.625 g, 0.855 mmol) under N2 at 20° C. The mixture was stirred for 6 h at 90° C. under N2. The reaction was quenched with H2O (5 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-cyclopropoxy-5-(4-(2-(methylthio)quinoxalin-6-yl)phenoxy)benzonitrile (8) (1.50 g, yield: 51.6%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.64 (s, 1H), 8.20 (d, J=2.0 Hz, 1H), 8.01 (d, J=8.8 Hz, 2H), 7.95 (dd, J=2.0, 8.8 Hz, 2H), 7.76 (d, J=8.4 Hz, 2H), 7.34 (d, J=3.2 Hz, 1H), 7.17-7.13 (m, 3H), 4.47 (tt, J=2.8, 6.0 Hz, 1H), 2.74 (s, 3H), 1.09-1.02 (m, 2H), 0.72 (d, J=7.2 Hz, 2H).


3-chloro-2-cyclopropoxy-5-(4-(2-(methylsulfonyl)quinoxalin-6-yl)phenoxy)benzonitrile (9): To a solution of 3-chloro-2-cyclopropoxy-5-(4-(2-(methylthio) quinoxalin-6-yl) phenoxy) benzonitrile (8) (1.5 g, 2.61 mmol) in THE (15 mL) and H2O (15 mL) was added Oxone (8.02 g, 13.0 mmol) at 20° C. and the mixture was stirred at 35° C. for 8 h. The mixture was poured into saturated aq. Na2SO3 (5 mL) and then extracted with EtOAc (5 mL×3), the organic layers were washed with brine (10 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-cyclopropoxy-5-(4-(2-(methylsulfonyl)quinoxalin-6-yl)phenoxy)benzonitrile (9) (2.10 g, yield: 98.2%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=9.54 (s, 1H), 8.42 (s, 1H), 8.30 (d, J=8.8 Hz, 1H), 8.20 (d, J=9.2 Hz, 1H), 7.83 (d, J=8.4 Hz, 2H), 7.36 (d, J=2.4 Hz, 1H), 7.22-7.17 (m, 3H), 4.48 (tt, J=2.4, 6.4 Hz, 1H), 3.42 (s, 3H), 1.06 (br s, 2H), 0.73 (br d, J=6.4 Hz, 2H).


N-(6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl) methanesulfonamide (Compound A97): To a solution of 3-chloro-2-cyclopropoxy-5-(4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenoxy)benzonitrile (9) (1.60 g, 2.93 mmol) and methanesulfonamide (0.835 g, 8.78 mmol) in MeCN (20 mL) was added Cs2CO3 (2.86 g, 8.78 mmol) at 25° C. and the mixture was stirred at 25° C. for 5 h. The mixture was poured into water (10 mL) and adjusted to pH=5-6 with 1M aq. HCl solution. The mixture was extracted with EtOAc (10 mL×3), the organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) give N-(6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A97) (0.789 g, yield: 48.8%) as a yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.44 (s, 1H), 8.15 (s, 1H), 7.89 (br d, J=8.8 Hz, 1H), 7.70 (d, J=8.8 Hz, 2H), 7.64-7.46 (m, 1H), 7.34 (d, J=3.2 Hz, 1H), 7.18-7.12 (m, 3H), 4.47 (tt, J=3.2, 6.4 Hz, 1H), 3.27 (br s, 3H), 1.09-1.03 (m, 2H), 0.72 (dd, J=0.8, 6.0 Hz, 2H). LCMS (220 nm): 98.25%. Exact Mass: 506.1; found 506.9/508.9.


Example 10: Synthesis of N-(6-(4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(methyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A164)



embedded image


6-bromo-2-methylsulfanyl-quinoxaline (inter N): To a solution of 6-bromo-2-chloro-quinoxaline (8.0 g, 0.0312 mol) in DMF (100 mL) was added NaSMe (2.63 g, 0.0375 mol) at 20° C. The reaction was stirred at 20° C. for 2 h. The mixture was quenched with water (100 mL) and extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (50×2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column to give 6-bromo-2-methylsulfanyl-quinoxaline (6.0 g, yield: 77.5%) as a white solid. 1H-NMR (400 MHz, CDCl3) δ=8.59 (s, 1H), 8.18 (d, J=2.0 Hz, 1H), 7.83-7.69 (m, 2H), 2.70 (s, 3H).


4-(2-(methylthio)quinoxalin-6-yl)aniline (3): To a solution of 6-bromo-2-(methylthio)quinoxaline (inter N) (1.0 g, 3.53 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (2) (0.927 g, 4.23 mmol) in 1,4-dioxane (20 mL) and H2O (4 mL) was added K2CO3 (1.46 g, 10.6 mmol) and Pd(dppf)Cl2 (0.516 g, 0.706 mmol) under N2 at 20° C. The mixture was stirred for 6 h at 80° C. under N2. The reaction was quenched with H2O (10 mL), then extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 4-(2-(methylthio)quinoxalin-6-yl)aniline (3) (0.95 g, yield: 87.6%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.53 (s, 1H), 8.06 (d, J=1.2 Hz, 1H), 7.86 (dd, J=1.2, 2.8 Hz, 2H), 7.49 (m, J=6.8 Hz, 2H), 6.74 (d, J=6.8 Hz, 2H), 2.64 (s, 3H).


3-chloro-2-fluoro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (5): To a solution of 4-(2-(methylthio)quinoxalin-6-yl)aniline (3) (1.0 g, 3.37 mmol) and 5-bromo-3-chloro-2-fluorobenzonitrile (4) (0.947 g, 4.04 mmol) in 1,4-Dioxane (20 mL) was added Cs2CO3 (2.74 g, 8.42 mmol) and SPhos Pd G3 (0.263 g, 0.337 mmol) under N2 at 20° C. The mixture was stirred for 8 h at 90° C. under N2. The reaction was quenched with H2O (10 mL), and then extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL×3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-fluoro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (5) (0.70 g, yield: 44.5%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.64 (s, 1H), 8.19 (s, 1H), 8.03-7.98 (m, 1H), 7.97-7.92 (m, 1H), 7.72 (d, J=8.4 Hz, 2H), 7.35 (dd, J=2.8, 6.0 Hz, 1H), 7.22-7.13 (m, 3H), 5.96 (s, 1H), 2.73 (s, 3H).


3-chloro-2-fluoro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (6): To a solution of 3-chloro-2-fluoro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (5) (0.650 g, 1.20 mmol) and Cs2CO3 (0.805 g, 2.47 mmol) in DMF (7 mL) was added MeI (0.263 g, 1.85 mmol) at 25° C. The mixture was stirred at 25° C. for 4 h. The reaction was quenched with H2O (10 mL), and then extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-fluoro-5-((4-(2-(methylthio) quinoxalin-6-yl) phenyl) amino) benzonitrile (6) (0.290 g, yield: 43.2%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.65 (s, 1H), 8.25-8.21 (m, 1H), 8.04-8.00 (m, 1H), 7.99-7.95 (m, 1H), 7.77 (d, J=8.4 Hz, 2H), 7.24 (d, J=8.4 Hz, 2H), 7.18 (dd, J=2.8, 6.0 Hz, 1H), 7.00 (dd, J=3.2, 4.0 Hz, 1H), 3.37 (s, 3H), 2.74 (s, 3H).


3-chloro-2-cyclopropoxy-5-(methyl(4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino) benzonitrile (8): To a solution of cyclopropanol (7) (57.7 mg, 0.993 mmol) in DMF (1 mL) was added NaH (60.0% purity, 38 mg, 0.993 mmol) at 0° C. under N2 and the mixture was stirred at 0° C. for 15 min, then 3-chloro-2-fluoro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (6) was added at 0° C. under N2. The mixture was stirred at 25° C. under N2 for 2 h. The mixture was quenched with sat. NH4Cl (5 mL) and then extracted with EtOAc (5 mL×3), the organic layers were washed with brine (5 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-chloro-2-cyclopropoxy-5-(methyl(4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (8) (0.27 g, yield: 92.0%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.64 (s, 1H), 8.25-8.21 (m, 1H), 8.05-8.00 (m, 1H), 7.98 (dd, J=1.6, 6.4 Hz, 1H), 7.75 (d, J=8.4 Hz, 2H), 7.23-7.19 (m, 3H), 7.07-7.03 (m, 1H), 4.45-4.38 (m, 1H), 3.36 (s, 3H), 2.74 (s, 3H), 1.08-1.03 (m, 2H), 0.68 (br dd, J=0.8, 6.4 Hz, 2H).


3-chloro-2-cyclopropoxy-5-(methyl(4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)amino) benzonitrile (9): To a solution of 3-chloro-2-cyclopropoxy-5-(methyl(4-(2-(methylthio)quinoxalin-6-yl)phenyl)amino)benzonitrile (8) (0.260 g, 0.440 mmol) in THE (2 mL) and H2O (2 mL) was added Oxone (1.08 g, 1.76 mmol) at 20° C. and the mixture was stirred at 35° C. for 8 h. LCMS showed the reaction was completed. The mixture was poured into saturated Na2SO3 solution (5 mL) and then extracted with EtOAc (5 mL×3), the organic layers were washed with brine (10 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-cyclopropoxy-5-(methyl(4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)amino)benzonitrile (9) (0.220 g, yield: 69.3%) as a yellow solid.


N-(6-(4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(methyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A164): To a solution of 3-chloro-2-cyclopropoxy-5-(methyl(4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)amino)benzonitrile (9) (220 mg, 0.349 mmol) and methanesulfonamide (99.5 mg, 1.05 mmol) in MeCN (2 mL) was added Cs2CO3 (341 mg, 1.05 mmol) at 25° C. and the mixture was stirred at 25° C. for 8 h under N2. The mixture was poured into water (3 mL) and then adjusted to pH=5-6 with 1M aq. HCl solution. The mixture was extracted with EtOAc (5 mL×3), the organic layers were washed with brine (2 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give N-(6-(4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(methyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A164) (33.5 mg, yield: 23.0%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.46-8.44 (m, 1H), 8.16 (s, 1H), 7.98-7.84 (m, 1H), 7.68 (d, J=8.4 Hz, 2H), 7.61-7.43 (m, 1H), 7.22 (d, J=3.2 Hz, 1H), 7.20 (d, J=8.4 Hz, 2H), 7.06 (d, J=2.8 Hz, 1H), 4.42 (tt, 6.0 Hz, 1H), 3.36 (s, 3H), 3.26 (br s, 3H), 1.08-1.03 (m, 2H), 0.72-0.65 (m, 2H). LCMS (220 nm): 97.19%. Exact Mass: 519.1; found 520.0/522.0.


Example 11: Synthesis of N-(6-(4-((3-chloro-5-cyano-4-cyclopropoxyphenyl) (2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A186)



embedded image


4-(2-(methylthio)quinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (3): To a solution of 4-(2-(methylthio)quinoxalin-6-yl)aniline (1) (1.20 g, 4.04 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (2) (2.81 g, 12.1 mmol) in THF (10 mL) was added TEA (1.69 mL, 12.1 mmol) at 25° C. and the mixture was stirred at 80° C. for 8 h under N2. LCMS showed the 30% starting material remained and 50% desired was detected. The reaction mixture was quenched with H2O (10 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude. The crude was purified by silica gel column chromatography (petroleum either/EtOAc=10/to 1/1) to give 4-(2-(methylthio)quinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (3) (700 mg, yield: 44.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.62 (s, 1H), 8.15 (d, J=2.0 Hz, 1H), 7.96 (s, 1H), 7.94 (d, J=2.0 Hz, 1H), 7.65-7.62 (m, 2H), 6.84-6.82 (m, 2H), 4.31-3.95 (m, 2H), 2.73 (s, 3H).


3-chloro-2-cyclopropoxy-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (5): To a solution of 4-(2-(methylthio)quinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (3) (400 mg, 1.03 mmol) and 5-bromo-3-chloro-2-cyclopropoxybenzonitrile (4) (281 mg, 1.03 mmol) in 1,4-dioxane (4 mL) was added Cs2CO3 (1.01 g, 3.09 mmol) and Ruphos Pd G3 (172 mg, 0.21 mmol) at 20° C. under N2. The mixture was stirred at 90° C. under N2 atmosphere for 8 h. LCMS showed the starting material was consumed and ˜30% desired was detected. The reaction mixture was quenched with H2O (10 mL) and extracted with EtOAc (5×3 mL). The combined organic layers were washed with brine (3 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum either/EtOAc=3/1 to 1/1) and then further purified by p-HPLC (FA) to give 3-chloro-2-cyclopropoxy-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (5) (150 mg, yield: 24.2%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.65 (s, 1H), 8.23 (d, J=2.0 Hz, 1H), 8.03 (d, J=8.8 Hz, 1H), 7.97 (dd, J=2.0 Hz, 8.4 Hz, 1H), 7.80-7.76 (m, 2H), 7.23-7.20 (m, 3H), 7.05 (d, J=3.2 Hz, 1H), 4.47-4.42 (m, 1H), 4.32 (q, J=8.4 Hz, 2H), 2.74 (s, 3H), 1.09-1.01 (m, 2H), 0.75-0.66 (m, 2H).


3-chloro-2-cyclopropoxy-5-((4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (6): To a solution of 3-chloro-2-cyclopropoxy-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (5) (150 mg, 0.25 mol) in THE (1 mL) and H2O (1 mL) was added Oxone (460 mg, 0.75 mol) at 25° C. The mixture was stirred for 8 h at 25° C. under N2. LCMS showed the reaction was completed. The reaction was quenched with H2O (2 mL) and extracted with EtOAc (2 mL×3). The combined organic layers were washed with brine (5 mL), dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-cyclopropoxy-5-((4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (6) (150 mg, yield: 79.7%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.42 (d, J=2.0 Hz, 1H), 8.29 (d, J=8.4 Hz, 1H), 8.21 (dd, J=1.6 Hz, 8.8 Hz, 1H), 7.82 (br d, J=8.4 Hz, 2H), 7.32 (d, J=2.8 Hz, 1H), 7.24-7.17 (m, 3H), 7.15 (d, J=2.8 Hz, 1H), 4.49-4.46 (m, 1H), 4.37-4.31 (m, 2H), 3.42 (s, 3H), 1.09-0.98 (m, 2H), 0.81-0.69 (m, 2H).


N-(6-(4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A186): To a solution of 3-chloro-2-cyclopropoxy-5-((4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (6) (150 mg, 0.24 mmol) in DMF (2 mL) was added MsNH2 (112 mg, 1.18 mmol), Cs2CO3 (230 mg, 0.71 mmol) at 25° C. The reaction was stirred at 25° C. for 2 h. LCMS showed the reaction was completed. The mixture was quenched with H2O (2 mL) and extracted with EtOAc (1 mL×3), then the EtOAc phases were washed with brine (2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by p-HPLC (FA) to give N-(6-(4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl) methanesulfonamide (Compound A186) (85.8 mg, yield: 60.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.43 (br d, J=6.4 Hz, 1H), 8.16 (s, 1H), 7.90 (br d, J=8.0 Hz, 1H), 7.71 (br d, J=8.4 Hz, 2H), 7.62-7.44 (m, 1H), 7.24-7.18 (m, 3H), 7.05 (d, J=2.8 Hz, 1H), 4.46-4.43 (m, 1H), 4.35-4.28 (m, 2H), 3.27 (br s, 3H), 1.09-1.01 (m, 2H), 0.76-0.67 (m, 2H) LCMS (220 nm): 97.76%. Exact Mass: 587.10; found 588.0/590.0.


Example 12: Synthesis of 3-chloro-2-cyclopropoxy-5-(2-(4-((2-morpholinopyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (Compound A201)



embedded image


3-chloro-2-fluoro-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy) phenyl)propan-2-yl)benzonitrile (2): To a solution of 3-chloro-2-fluoro-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (inter U) (2.0 g, 6.21 mmol) and (2-(methylthio)pyrimidin-4-yl)methanol (1) (1.29 g, 7.46 mmol) in THF (30 mL) was added PPh3 (2.4 g, 9.32 mmol) and DIAD (2.5 g, 12.4 mmol) at 0° C. under N2 and the mixture was stirred at 20° C. for 8 h. LCMS showed the starting material was consumed and ˜32% desired was detected. The reaction mixture was diluted with H2O (35 mL). The resulting aqueous mixture were extracted with EtOAc (3×15 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=20:1 to 5:1) to give 3-chloro-2-fluoro-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (2) (2.2 g, yield: 70.3%) as colorless oil. 1H-NMR (400 MHz, CDCl3) δ=8.54 (d, J=5.2 Hz, 1H), 7.51-7.44 (m, 1H), 7.34 (dd, J=2.4, 5.2 Hz, 1H), 7.21 (d, J=4.8 Hz, 1H), 7.14-7.07 (m, 2H), 6.93-6.87 (m, 2H), 5.09 (s, 2H), 2.59 (s, 3H), 1.66 (s, 6H).


3-chloro-2-cyclopropoxy-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy) phenyl)propan-2-yl)benzonitrile (4): To a solution of cyclopropanol (3) (339 mg, 5.83 mmol) in DMF (15 mL) was added sodium hydride (79 mg, 60% purity, 7.29 mmol) at 0° C. under N2. The mixture was stirred 0° C. under N2 for 0.5 h. Then was added 3-chloro-2-fluoro-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (2) (2.6 g, 4.86 mmol) in DMF (15 mL) at 20° C. under N2. The mixture was stirred 25° C. under N2 for 2 h. LCMS showed the reaction was completed. The reaction mixture was quenched with sat.NH4Cl (45 mL) and extracted with EtOAc (3×15 mL). The combined organic layers were washed with brine (3×15 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=20:1 to 5:1) to give 3-chloro-2-cyclopropoxy-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy) phenyl)propan-2-yl)benzonitrile (4) (1.9 g, yield: 67.1%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.54 (d, J=5.2 Hz, 1H), 7.42 (d, J=2.0 Hz, 1H), 7.31 (d, J=2.0 Hz, 1H), 7.22 (d, J=5.2 Hz, 1H), 7.15-7.09 (m, 2H), 6.89 (d, J=8.8 Hz, 2H), 5.09 (s, 2H), 4.49-4.45 (m, 1H), 2.59 (s, 3H), 1.64 (s, 6H), 1.02 (br d, J=1.2 Hz, 2H), 0.71 (br d, J=7.2 Hz, 2H).


3-chloro-2-cyclopropoxy-5-(2-(4-((2-(methylsulfonyl)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (5): To a solution of 3-chloro-2-cyclopropoxy-5-(2-(4-((2-(methylthio)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (4) (1.65 g, 3.19 mmol) in THE (20 mL) and H2O (20 mL) was added Oxone (5.88 g, 9.56 mmol) at 20° C. and the mixture was stirred at 35° C. for 8 h. LCMS showed the reaction was completed. The mixture was poured into sat.Na2SO3 (40 mL) and extracted with EtOAc (15 mL×3), the organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-cyclopropoxy-5-(2-(4-((2-(methylsulfonyl)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (5) (1.7 g, yield: 81.4%) as colorless oil. 1H-NMR (400 MHz, CDCl3) δ=8.94 (d, J=5.2 Hz, 1H), 7.85 (d, J=4.8 Hz, 1H), 7.42 (d, J=2.4 Hz, 1H), 7.30 (d, J=2.0 Hz, 1H), 7.15 (d, J=8.4 Hz, 2H), 6.91 (d, J=9.2 Hz, 2H), 5.29 (s, 2H), 4.52-4.42 (m, 1H), 3.39 (s, 3H), 1.70-1.63 (m, 6H), 1.07-0.96 (m, 2H), 0.73-0.68 (m, 2H).


tert-butyl 3-chloro-2-cyclopropoxy-5-(2-(4-((2-morpholinopyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (Compound A201): To a solution of 3-chloro-2-cyclopropoxy-5-(2-(4-((2-(methyl sulfonyl)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (5) (100 mg, 0.19 mmol) in DMSO (1 mL) was added morpholine (6) (79 mg, 0.90 mmol) and DIEA (47 mg, 0.36 mmol) at 20° C. and stirred at 80° C. for 2 h. LCMS showed the starting material was consumed and 78% desired was detected. The reaction mixture was quenched with H2O (2 mL) and extracted with EtOAc (3×2 mL). The combined organic layers were washed with brine (2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (Neutral) to give tert-butyl 3-chloro-2-cyclopropoxy-5-(2-(4-((2-morpholinopyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (Compound A201) (44 mg, yield: 46%) as off-white solid. 1H-NMR (400 MHz, CDCl3) δ=8.34 (d, J=4.8 Hz, 1H), 7.42 (d, J=2.0 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.11 (br d, J=8.8 Hz, 2H), 6.89 (br d, J=8.8 Hz, 2H), 6.77 (d, J=4.8 Hz, 1H), 4.96 (s, 2H), 4.51-4.44 (m, 1H), 3.80 (br dd, J=4.4, 14.0 Hz, 8H), 1.64 (s, 6H), 1.07-0.98 (m, 2H), 0.76-0.67 (m, 2H). LCMS (220 nm): 98.97%. Exact Mass: 504.2, found: 505.3/506.8.


Example 13: Synthesis of N-(6-(4-((5-cyano-6-cyclopropoxypyridin-3-yl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A203)



embedded image


5-bromo-2-cyclopropoxynicotinonitrile (3): To a solution of cyclopropanol (2) (468 mg, 10.0 mmol) in THF (10 mL) was added sodium hydride (515 mg, 10.0 mmol) at 0° C. under N2 and the mixture was stirred at 0° C. for 30 min, then 5-bromo-2-fluoronicotinonitrile (1) (1.5 g, 10.0 mmol) in THF (5 mL) was added at 0° C. under N2. The mixture was stirred 25° C. under N2 for 2 h. LCMS showed the reaction was completed. The mixture was quenched with aq. NH4Cl (10 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (PE:EtOAc=20:1 to 3:1) to give 5-bromo-2-cyclopropoxynicotinonitrile (3) (1.6 g, yield: 80.71%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.45 (d, J=2.4 Hz, 1H), 7.96 (d, J=2.8 Hz, 1H), 4.45-4.33 (m, 1H), 0.89-0.84 (m, 4H).


2-cyclopropoxy-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)nicotinonitrile (5): To a solution of 5-bromo-2-cyclopropoxynicotinonitrile (3) (68.4 mg, 0.26 mmol), 4-(2-methylsulfanylquinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (4) (100 mg, 0.26 mmol) and t-BuONa (0.4 mL) in t-Amyl-OH (2 mL) was added t-BuXphos Pd G3 (21 mg, 0.03 mmol) at 20° C. under N2 atmosphere. The mixture was stirred at 90° C. under N2 for 12 h. LCMS showed the starting material remained and ˜40% desired was detected. The reaction mixture was quenched with H2O (2 mL) and extracted with EtOAc (2 mL×3). The combined organic layers were washed with brine (2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=20:1 to 3:1) to give 2-cyclopropoxy-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)nicotinonitrile (5) (490 mg, yield: 95.1%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.63 (s, 1H), 8.30 (d, J=3.2 Hz, 1H), 8.18 (d, J=1.6 Hz, 1H), 8.01-7.94 (m, 2H), 7.71-7.69 (m, 3H), 7.02 (d, J=8.8 Hz, 2H), 4.42-4.40 (m, 1H), 4.35-4.29 (m, 2H), 2.73 (s, 3H), 0.91-0.86 (m, 4H).


2-cyclopropoxy-5-((4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)nicotinonitrile (6): To a solution of 2-cyclopropoxy-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)nicotinonitrile (5) (200 mg, 0.37 mmol) in THF (2 mL) and H2O (2 mL) was added Oxone (575 mg, 0.94 mmol) at 25° C. The mixture was stirred at 35° C. under N2 for 6 h. LCMS showed the reaction was completed. The reaction mixture was quenched with Na2SO3 (5 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL×3), dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 2-cyclopropoxy-5-((4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)nicotinonitrile (6) (160 mg, yield: 63.4%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=9.52 (s, 1H), 8.38 (d, J=1.6 Hz, 1H), 8.36-8.34 (m, 1H), 8.19-8.15 (m, 2H), 7.76-7.75 (m, 2H), 7.74 (s, 1H), 7.02-6.99 (m, 2H), 4.45-4.38 (m, 1H), 4.36-4.33 (m, 2H), 3.42 (s, 3H), 0.93-0.88 (m, 4H).


N-(6-(4-((5-cyano-6-cyclopropoxypyridin-3-yl)(2,2,2-trifluoroethyl)amino)phenyl) quinoxalin-2-yl)methanesulfonamide (Compound A203): To a solution of 2-cyclopropoxy-5-((4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)nicotinonitrile (6) (110 mg, 0.18 mmol) and MsNH2 (53 mg, 0.55 mmol) in DMF (3 mL) was added Cs2CO3 (179 mg, 0.55 mmol) at 25° C. The mixture was stirred at 50° C. for 3 h. LCMS showed the starting material was consumed and ˜85% desired was detected. The reaction mixture was quenched with H2O (3 mL). The resulting aqueous mixture were extracted with EtOAc (1 mL×3). The combined organic layers were washed with brine (3 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (Neutral) to give N-(6-(4-((5-cyano-6-cyclopropoxypyridin-3-yl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A203) (68.3 mg, yield: 39.3%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.48 (s, 1H), 8.30 (d, J=2.8 Hz, 1H), 8.12 (s, 1H), 7.86 (br d, J=8.4 Hz, 1H), 7.69 (d, J=3.2 Hz, 1H), 7.63 (br d, J=8.4 Hz, 2H), 7.69-7.48 (m, 1H), 7.01 (d, J=8.8 Hz, 2H), 4.45-4.42 (m, 1H), 4.32 (q, J=8.4, 16.8 Hz, 2H), 3.26 (br s, 3H), 0.92-0.86 (m, 4H). LCMS: (220 nm): 97.77%. Exact Mass: 554.1; found 555.0/556.0.


Example 14: Synthesis of N-((6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl) quinoxalin-2-yl)methyl)methanesulfonamide (Compound A207)



embedded image


6-bromo-2-vinylquinoxaline (2): To a solution of potassium trifluoro(vinyl)borate (a) (2.8 g, 18.5 mmol) and 6-bromo-2-chloroquinoxaline (1) (5.0 g, 18.5 mmol) in 1,4-dioxane (70 mL) was added TEA (3.8 g, 37.0 mmol) and Pd(dppf)Cl2 (1.35 g, 1.85 mmol) under N2 at 20° C. The mixture was stirred for 6 h at 90° C. under N2. LCMS showed the starting material was consumed and the product was detected. The reaction mixture was quenched with H2O (20 mL). Then the mixture was extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (30 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduce pressure. The crude was purified by silica gel column chromatography (PE:EtOAc from 1:0 to 1:2) to give 6-bromo-2-vinylquinoxaline (2) (5.0 g, yield: 92.1%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.96 (s, 1H), 8.23 (d, J=2.0 Hz, 1H), 7.89 (d, J=8.8 Hz, 1H), 7.80 (dd, J=2.0, 8.8 Hz, 1H), 7.00 (dd, J=10.8, 17.6 Hz, 1H), 6.48 (d, J=17.6 Hz, 1H), 5.81 (d, J=11.2 Hz, 1H).


6-bromoquinoxaline-2-carboxylic acid (3): To a mixture of 6-bromo-2-vinylquinoxaline (2) (5.0 g, 19.1 mmol) in THF/H2O (80 mL, 3/1) was added NaIO4 (12.3 g, 57.4 mmol) and K2O4.2H2O (3.5 g, 9.57 mmol) at 25° C. and the mixture was stirred at 25° C. for 2 h. LCMS showed the reaction was completed. The reaction mixture was poured into water (20 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (40 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc from 1:0 to 1:1) to give 6-bromoquinoxaline-2-carboxylic acid (3) (2.0 g, yield: 37.2%) as white solid. 1H-NMR (400 MHz, DMSO-d6) δ=9.42 (s, 1H), 8.42 (d, J=2.0 Hz, 1H), 8.16 (d, J=8.8 Hz, 1H), 8.08 (dd, J=2.4, 9.2 Hz, 1H).


Methyl 6-bromoquinoxaline-2-carboxylate (4): To a solution of 6-bromoquinoxaline-2-carboxylic acid (3) (2.0 g, 7.1 mmol) in DMF (20 mL) was added Cs2CO3 (4.6 g, 14.2 mmol) and MeI (2.0 g, 14.2 mmol) at 35° C. under N2 and the mixture was stirred at 35° C. for 8 h under N2. LCMS showed the reaction was completed. The reaction mixture was quenched with water (10 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (15 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc from 1:0 to 1:2) to give methyl 6-bromoquinoxaline-2-carboxylate (4) (1.6 g, yield: 75.8%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.80 (s, 1H), 7.64 (d, J=2.0 Hz, 1H), 7.42 (d, J=9.2 Hz, 1H), 7.21 (dd, J=2.0, 9.2 Hz, 1H), 3.38 (s, 3H).


(6-bromoquinoxalin-2-yl)methanol (5): To a solution of 6-bromoquinoxaline-2-carboxylate (4) (1.3 g, 4.87 mmol) in MeOH (10 mL) and DCM (5 mL) was added NaBH4 (0.37 g, 9.73 mmol) at 0° C. under N2. The mixture was stirred at 20° C. for 1 h. The resulting mixture was quenched with sat. NH4Cl (5 mL) and then extracted with EtOAc (3×5 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc from 1:0 to 2:1) to give (6-bromoquinoxalin-2-yl)methanol (5) (0.46 g, yield: 35.6%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=9.55 (s, 1H), 8.39 (d, J=2.0 Hz, 1H), 8.17 (d, J=8.8 Hz, 1H), 7.96 (dd, J=2.0, 9.2 Hz, 1H), 4.13 (s, 2H).


Tert-butyl ((6-bromoquinoxalin-2-yl)methyl)(methylsulfonyl)carbamate (6): To a solution of (6-bromoquinoxalin-2-yl)methanol (5) (200 mg, 0.753 mmol) and MsNHBoc (0.16 g, 0.753 mmol) in THE (2 mL) was added PPh3 (0.30 g, 1.13 mmol) and DIAD (0.30 g, 1.51 mmol) at 0° C. under N2 and the mixture was stirred at 20° C. for 8 h. LCMS showed the starting material was consumed and the product was detected. The reaction was quenched with H2O (1 mL). Then the mixture was extracted with EtOAc (1 mL×3). The combined organic layers were washed with brine (2 mL×2), dried over Na2SO4, filtered and concentrated under reduce pressure. The crude was purified by silica gel column chromatography (PE:EtOAc from 1:0 to 2:1) to give tert-butyl ((6-bromoquinoxalin-2-yl)methyl)(methylsulfonyl)carbamate (6) (0.200 g, yield: 54.2%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.81 (s, 1H), 8.32 (s, 1H), 7.86 (s, 2H), 6.35 (br s, 1H), 5.26 (s, 2H), 3.56 (s, 3H), 1.42 (s, 9H).


Tert-butyl ((6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl)methyl)(methylsulfonyl)carbamate (8): To a solution of 3-chloro-2-cyclopropoxy-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)benzonitrile (7) (150 mg, 0.328 mmol) and tert-butyl ((6-bromoquinoxalin-2-yl)methyl)(methylsulfonyl)carbamate (6) (152 mg, 0.328 mmol) in 1,4-Dioxane (2.0 mL) and H2O (0.4 mL) was added K2CO3 (0.14 g, 0.984 mmol) and Pd(dppf)Cl2 (0.048 g, 0.066 mmol) at 25° C. under N2 and the mixture was stirred at 90° C. for 2 h under N2. LCMS showed the reaction was completed. The reaction was quenched with water (2 mL) and extracted with EtOAc (1 mL×3). The combined organic layers were washed with brine (1 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc from 1:0 to 1:1) to give tert-butyl ((6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl)methyl)(methylsulfonyl)carbamate (8) (0.14 g, yield: 61.9%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.83 (d, J=11.2 Hz, 1H), 8.32 (s, 1H), 8.08-8.03 (m, 1H), 8.02-7.96 (m, 1H), 7.80-7.77 (m, 2H), 7.35 (d, J=2.8 Hz, 1H), 7.20-7.14 (m, 3H), 5.31 (d, J=4.4 Hz, 2H), 4.50-4.45 (m, 1H), 3.60 (d, J=4.4 Hz, 3H), 1.73-1.56 (m, 9H), 1.08-1.01 (m, 2H), 0.75-0.69 (m, 2H).


N-((6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl)methyl)methanesulfonamide (Compound A207): To a solution of tert-butyl ((6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl)methyl) (methylsulfonyl)carbamate (8) (140 mg, 0.180 mmol) in DCM (2 mL) was added TFA (1 mL, 9.02 mmol) at 0° C. and the mixture was stirred at 20° C. for 1 h. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC (Waters xbridge 150*25 mm 10 um, water (NH4HCO3)-ACN from 34% to 64%) to give N-((6-(4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenyl)quinoxalin-2-yl)methyl)methanesulfonamide (Compound A207) (95.0 mg, yield: 15.0%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=8.89 (s, 1H), 8.33 (d, J=1.6 Hz, 1H), 8.14 (d, J=8.8 Hz, 1H), 8.08 (d, J=2.0 Hz, 1H), 7.79 (d, J=8.4 Hz, 2H), 7.35 (d, J=3.2 Hz, 1H), 7.22-7.14 (m, 3H), 5.77 (br s, 1H), 4.76 (d, J=5.2 Hz, 2H), 4.48 (tt, J=2.8, 6.0 Hz, 1H), 3.07 (s, 3H), 1.11-1.02 (m, 2H), 0.76-0.68 (m, 2H). LCMS (220 nm): 99.5%. Exact Mass: 520.1; found Ms+18: 521.0/523.0.


Example 15: Synthesis of N-(6-(4-((3-chloro-5-cyano-4-morpholinophenyl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A224)



embedded image


3-chloro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)-2-morpholinobenzonitrile (4): To a solution of 4-(2-methylsulfanylquinoxalin-6-yl)-N-(2,2,2-trifluoroethyl)aniline (400 mg, 1.14 mmol) and 5-bromo-3-chloro-2-morpholinobenzonitrile (460 mg, 1.37 mmol) in 1,4-Dioxane (6.0 mL) was added Cs2CO3 (933 mg, 2.86 mmol) and Sphos-Pd-G3 (26.8 mg, 0.34 mmol) under N2 at 20° C. The mixture was stirred for 3 h at 90° C. under N2. LCMS showed the reaction was completed. The reaction was filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=10:13:1) to give 3-chloro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)-2-morpholinobenzonitrile (4) (200 mg, yield: 27.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.65 (s, 1H), 8.23 (d, J=2.0 Hz, 1H), 8.05-8.01 (m, 1H), 7.78 (d, J=8.4 Hz, 2H), 7.33 (d, J=8.0 Hz, 1H), 7.24 (d, J=8.4 Hz, 2H), 7.15 (d, J=2.8 Hz, 1H), 7.03 (d, J=2.8 Hz, 1H), 4.31 (q, J=8.4 Hz, 2H), 3.88-3.85 (m, 4H), 3.31 (s, 4H), 2.74 (s, 3H).


3-chloro-5-((4-(2-(methylsulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)-2-morpholinobenzonitrile (5): To a solution of 3-chloro-5-((4-(2-(methylthio)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)-2-morpholinobenzonitrile (4) (200 mg, 0.32 mmol) in THE (2.0 mL) and H2O (2.0 mL) was added Oxone (971 mg, 1.58 mmol) at 25° C. The mixture was stirred for 8 h at 25° C. under N2. LCMS showed the reaction was completed. The reaction was quenched with sat.Na2SO3 (10 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (10 mL), dried with anhydride Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-5-((4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)-2-morpholinobenzonitrile (5) (250 mg, yield: 98.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=9.54 (s, 1H), 8.43 (s, 1H), 8.30 (d, J=8.8 Hz, 1H), 8.21 (d, J=8.8 Hz, 1H), 7.82 (d, J=8.4 Hz, 2H), 7.25-7.20 (m, 3H), 7.12 (d, J=2.4 Hz, 1H), 4.38-4.30 (m, 2H), 3.90-3.86 (m, 4H), 3.43 (s, 3H), 3.34 (s, 4H).


N-(6-(4-((3-chloro-5-cyano-4-morpholinophenyl)(2,2,2-trifluoroethyl)amino) phenyl) quinoxalin-2-yl)methanesulfonamide (Compound A224): To a solution of 3-chloro-5-((4-(2-(methyl sulfonyl)quinoxalin-6-yl)phenyl)(2,2,2-trifluoroethyl)amino)-2-morpholinobenzonitrile (5) (250 mg, 0.37 mmol) and MsNH2 (71.1 mg, 0.75 mmol) in DMF (3 mL) was added Cs2CO3 (365 mg, 1.12 mmol) at 25° C. The reaction was stirred at 25° C. for 2 h. LCMS showed the reaction was completed. The mixture was poured into water (5 mL) and extracted with EtOAc (3 mL×3), then the EtOAc phases were washed with brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude was purified by perp-HPLC (Column: xbridge 150*25 mm 10 um; mobile phase: water (NH4HCO3)-ACN, gradient: 25%-55% B over 10 min) to give N-(6-(4-((3-chloro-5-cyano-4-morpholinophenyl)(2,2,2-trifluoroethyl)amino)phenyl)quinoxalin-2-yl)methanesulfonamide (Compound A224) (40 mg, yield: 17.3%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=8.44 (s, 1H), 8.17 (s, 1H), 7.91 (d, J=7.6 Hz, 1H), 7.72 (d, J=7.6 Hz, 2H), 7.53 (s, 1H), 7.22 (d, J=8.4 Hz, 2H), 7.16 (s, 1H), 7.03 (s, 1H), 4.31 (q, J=7.6 Hz, 2H), 3.87 (s, 4H), 3.39-3.19 (m, 7H). LCMS: (220 nm): 97.9%. Exact Mass: 616.1, found 617.0/619.0.


Example 16: Synthesis of N-(3-((4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)methanesulfonamide (Compound A228)



embedded image


3-chloro-2-cyclopropoxy-5-(4-hydroxyphenoxy)benzonitrile (1): To a 3-chloro-2-cyclopropoxy-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)benzonitrile (inter Z1) (400 mg, 0.923 mmol) in THF (5 mL) and H2O (5 mL) was added Oxone (1.13 g, 1.185 mmol) at 35° C. and the mixture was stirred at 35° C. for 2 h. LCMS showed the reaction was completed. The mixture was poured into saturated Na2SO3 (5 mL) and extracted with EtOAc (mL×3), the organic layers were washed with brine (10 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 3-chloro-2-cyclopropoxy-5-(4-hydroxyphenoxy)benzonitrile (1) (0.48 g, yield: 94.8%) as yellow solid. LCMS (220 nm): 88.2%. Exact Mass: 301.0; found Mass+1: 302.0/304.0


Tert-butyl (3-((4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)carbamate (3): To a solution of 3-chloro-2-cyclopropoxy-5-(4-hydroxyphenoxy)benzonitrile (1) (200 mg, 0.597 mmol) and tert-butyl (3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)carbamate (2) (153 mg, 0.716 mmol) in Tol (2 mL) was added CMBP (280 mg, 1.19 mmol) at 25° C. and the mixture was stirred at 110° C. for 2 h under N2. LCMS showed the starting material was consumed and the desired product was detected. The reaction was quenched with H2O (2 mL), and extracted with EtOAc (1 mL×3). The combined organic layers were washed with brine (3 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE-EA from 0%-50%) to give tert-butyl (3-((4 (3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)carbamate (3) (280 mg, yield: 85.0%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=7.18 (d, J=2.8 Hz, 1H), 7.04-6.97 (m, 1H), 6.97-6.85 (m, 4H), 4.97 (br s, 1H), 4.44-4.40 (m, 1H), 4.08 (s, 2H), 2.06 (br d, J=4.8 Hz, 6H), 1.46 (s, 9H), 1.09-0.96 (m, 2H), 0.76-0.60 (m, 2H).


5-(4-((3-aminobicyclo[1.1.1]pentan-1-yl)methoxy)phenoxy)-3-chloro-2-cyclopropoxybenzonitrile (4): To a mixture of tert-butyl (3-((4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)carbamate (3) (280 mg, 0.507 mmol) in DCM (3 mL) was added TFA (1.5 mL, 0.507 mmol) at 0° C. Then the reaction was stirred at 20° C. for 2 h. LCMS showed the starting material was consumed and desired was detected. The mixture was quenched with sat. NaHCO3 (3 mL) and extracted with DCM (2 mL×3). The combined organic layers was dried over anhydrous Na2SO4, filtered and concentrated under reduce pressure to give 5-(4-((3-aminobicyclo[1.1.1]pentan-1-yl)methoxy)phenoxy)-3-chloro-2-cyclopropoxybenzonitrile (4) (200 mg, yield: 69.6%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.19 (d, J=2.8 Hz, 1H), 6.98 (d, J=3.2 Hz, 1H), 6.96-6.91 (m, 4H), 4.44-4.39 (m, 1H), 4.05 (s, 2H), 1.86 (s, 6H), 1.05-1.00 (m, 2H), 0.71-0.65 (m, 2H).


N-(3-((4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenoxy)methyl) bicyclo[1.1.1.]pentan-1-yl)methanesulfonamide (Compound A228): To a solution of 5-(4-((3-aminobicyclo[1.1.1]pentan-1-yl)methoxy)phenoxy)-3-chloro-2-cyclopropoxybenzonitrile (4) (200 mg, 0.454 mmol) in DCM (3 mL) was added TEA (140 mg, 1.36 mmol) and methanesulfonyl chloride (52 mg, 0.454 mmol) at 0° C. The mixture was stirred at 20° C. for 1 h. LCMS showed the reaction was completed. The reaction was quenched with ice water (5 mL), extracted with DCM (3 mL×3). The combined organic layers were washed with brine (3 mL×2), dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (Phenomenex luna C18 150*25 mm*10 um, water (FA)-ACN from 37% to 67%) to give N-(3-((4-(3-chloro-5-cyano-4-cyclopropoxyphenoxy)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl) methanesulfonamide (Compound A228) (61 mg, yield: 46.2%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=7.19 (d, J=2.0 Hz, 1H), 6.97 (br d, J=8.4 Hz, 3H), 6.90 (d, J=8.8 Hz, 2H), 4.95 (br s, 1H), 4.47-4.39 (m, 1H), 4.09 (s, 2H), 3.02 (s, 3H), 2.15 (s, 6H), 1.03 (br s, 2H), 0.68 (q, J=2.0 Hz, 2H). LCMS (220 nm): 99.5%. Exact Mass: 474.1; found Ms+18: 492.1/494.0.


Example 17: Synthesis of N-(3-((4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)methanesulfonamide (Compound A236)



embedded image


5-((4-((tert-butyldimethylsilyl)oxy)phenyl)(2,2,2-trifluoroethyl)amino)-3-chloro-2-cyclopropoxybenzonitrile (4): To a solution of 4-((tert-butyldimethylsilyl)oxy)-N-(2,2,2-trifluoroethyl)aniline (2) (0.5 g, 1.64 mmol) and 5-bromo-3-chloro-2-cyclopropoxybenzonitrile (3) (0.6 g, 1.96 mmol) in 1,4-Dioxane (7 mL) was added Cs2CO3 (1.6 g, 4.91 mmol) and SPhos-Pd-G3 (0.26 g, 0.33 mmol) under N2 at 20° C. The mixture was stirred at 90° C. for 8 h under N2. LCMS showed the reaction was completed. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=20:13:1) to give 5-((4-((tert-butyldimethylsilyl)oxy)phenyl)(2,2,2-trifluoroethyl)amino)-3-chloro-2-cyclopropoxybenzonitrile (4) (0.85 g, yield: 73.1%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=7.05 (d, J=8.8 Hz, 1H), 6.93-6.89 (m, 2H), 6.73 (dd, J=2.8, 6.0 Hz, 2H), 6.60 (d, J=8.4 Hz, 1H), 4.36 (tt, J=2.8, 6.0 Hz, 1H), 3.75-3.66 (m, 2H), 1.02 (s, 9H), 0.98 (s, 6H), 0.26 (s, 2H), 0.17 (s, 2H).


3-chloro-2-cyclopropoxy-5-((4-hydroxyphenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (5): To a solution of 5-((4-((tert-butyldimethylsilyl)oxy)phenyl)(2,2,2-trifluoroethyl)amino)-3-chloro-2-cyclopropoxybenzonitrile (4) (850 mg, 1.20 mmol) in THE (9 mL) was added TBAF (1 mol/L, 1.80 mL, 1.80 mmol) at 0° C. and the mixture was stirred at 25° C. for 1 h. LCMS showed the reaction was completed. The mixture was quenched with water (15 mL) and extracted with EtOAc (6 mL×3). The combined organic layers were washed with brine (10 mL), dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=5:11:1) to give 3-chloro-2-cyclopropoxy-5-((4-hydroxyphenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (5) (500 mg, yield: 87.3%) as yellow solid. 1H-NMR (400 MHz, CDCl3) δ=7.08 (d, J=8.4 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 6.74 (d, J=8.8 Hz, 1H), 6.62 (d, J=8.4 Hz, 1H), 4.39-4.35 (m, 1H), 3.71 (q, J=8.8 Hz, 2H), 1.04-0.98 (m, 2H), 0.69-0.61 (m, 2H).


tert-butyl (3-((4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenoxy)methyl)bicyclo[1.1.1]pentan-l-yl)carbamate (7): To a solution of 3-chloro-2-cyclopropoxy-5-((4-hydroxyphenyl)(2,2,2-trifluoroethyl)amino)benzonitrile (5) (200 mg, 0.52 mmol) and tert-butyl (3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)carbamate (6) (111 mg, 0.52 mmol) in toluene (4.0 mL) was added CMBP (252 mg, 1.05 mmol) at 25° C. under N2 and the mixture was stirred at 110° C. for 1 h. LCMS showed the reaction was completed The mixture was quenched with H2O (10 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL), dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EtOAc=10:13:1) to give tert-butyl (3-((4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)carbamate (7) (300 mg, yield: 79.5%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=7.10 (d, J=8.8 Hz, 2H), 6.98-6.93 (m, 2H), 6.89 (d, J=3.2 Hz, 1H), 6.70 (d, J=3.2 Hz, 1H), 4.99 (s, 1H), 4.37-4.34 (m, 1H), 4.21-4.15 (m, 2H), 4.10 (s, 2H), 2.08 (s, 6H), 1.47 (s, 9H), 1.04-0.98 (m, 2H), 0.65-0.63 (m 2H).


5-((4-((3-aminobicyclo[1.1.1]pentan-1-yl)methoxy)phenyl)(2,2,2-trifluoroethyl)amino)-2-(cyclopropylamino)nicotinonitrile (8): To a solution of tert-butyl (5-((4-((3-((tert-butoxycarbonyl)amino)bicyclo[1.1.1]pentan-1-yl)methoxy)phenyl)(2,2,2-trifluoroethyl)amino)-3-cyanopyridin-2-yl)(cyclopropyl)carbamate (7) (250 mg, 0.39 mmol) in DCM (3 mL) was added TFA (443 mg, 3.88 mmol) at 0° C. under N2 and the mixture was stirred at 25° C. for 1 h. LCMS showed the reaction was successful. The mixture was concentrated under reduced pressure to give 5-((4-((3-aminobicyclo[1.1.1]pentan-1-yl)methoxy)phenyl)(2,2,2-trifluoroethyl)amino)-2-(cyclopropylamino)nicotinonitrile (8) (200 mg, yield: 92.9%) as yellow oil. 1H-NMR (400 MHz, CDCl3) δ=8.15 (s, 1H), 7.30-7.27 (m, 1H), 6.91-6.86 (m, 2H), 6.86-6.81 (m, 2H), 5.26 (s, 1H), 4.19-4.12 (m, 2H), 4.03 (s, 2H), 2.82 (s, 1H), 1.95 (s, 6H), 0.87 (d, J=6.4 Hz, 2H), 0.60 (br s, 2H).


N-(3-((4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)methanesulfonamide (Compound A236): To a solution of 5-((4-((3-aminobicyclo[1.1.1]pentan-1-yl)methoxy)phenyl)(2,2,2-trifluoroethyl)amino)-2-(cyclopropylamino)nicotinonitrile (8) (200 mg, 0.42 mmol) and TEA (0.36 mL, 2.1 mmol) in DCM (4 mL) was added MsCl (43.5 mg, 0.38 mmol) at 0° C. and stirred at 20° C. for 1 h. LCMS showed the reaction was completed. The mixture was quenched with H2O (5 mL) and extracted with DCM (3 mL×3). The combined organic layers were washed with brine (3 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude was purified by perp-HPLC (Column: xbridge 150*25 mm 10 um; mobile phase: water (NH4HCO3)-ACN, gradient: 25%-55% B over 10 min) give N-(3-((4-((3-chloro-5-cyano-4-cyclopropoxyphenyl)(2,2,2-trifluoroethyl)amino)phenoxy)methyl)bicyclo[1.1.1]pentan-1-yl)methanesulfonamide (Compound A236) (121 mg, yield: 82.1%) as white solid. 1H-NMR (400 MHz, CDCl3) δ=7.11 (d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 6.91 (d, J=3.2 Hz, 1H), 6.69 (d, J=3.2 Hz, 1H), 4.95 (s, 1H), 4.35 (tt, J=2.8, 6.0 Hz, 1H), 4.18 (q, J=8.4 Hz, 2H), 4.12 (s, 2H), 3.02 (s, 3H), 2.16 (s, 6H), 1.05-0.98 (m, 2H), 0.62-0.67 (m, 2H). LCMS: (220 nm): purity: 98.6%. Exact Mass: 555.1, found 556.1/558.0.


Example 18: Synthesis of Compounds of Table A

Compounds of Table A were synthesized according to modified procedures based on the examples provided herein and/or with organic chemistry reactions known to one skilled in the art. The characterization of the synthesized compounds is provided in Table 1.









TABLE 1







Characterization of Compounds of Table A (M + H by LC-MS)












M + H
M + H


Comp ID

1H-NMR

(calc)
(found)













A1

1H-NMR (400 MHz, CDCl3) δ = 8.81 (s, 1H), 8.02 (d, J = 8.4 Hz, 1H), 7.55-7.49 (m, 4H), 7.36 (d,

518.1
518.0



J = 2.4 Hz, 1H), 7.33 (d, J = 8.0 Hz, 2H), 5.87 (s, 1H), 4.58 (d, J = 4.0 Hz, 2H), 4.45 (t, J = 6.0 Hz,





2H), 3.89 (t, J = 6.0 Hz, 2H), 2.99 (s, 3H), 1.72 (s, 6H).




A2

1H-NMR (400 MHz, CDCl3) δ = 8.97 (s, 1H), 8.67 (s, 1H), 7.98 (d, J = 8.0 Hz, 2H), 7.47 (d, J =

519.1
518.9



2.0 Hz, 1H), 7.37 (d, J = 2.0 Hz, 1H), 7.35 (d, J = 8.4 Hz, 2H), 5.35 (s, 1H), 4.56 (d, J = 5.6 Hz,





2H), 4.44 (t, J = 6.0 Hz, 2H), 3.89 (t, J = 6.0 Hz, 2H), 3.01 (s, 3H), 1.72 (br s, 6H)




A3

1H-NMR (400 MHz, CDCl3) δ = 8.58 (s, 1H), 7.87 (d, J = 8.4 Hz, 2H), 7.75 (d, J = 8.4 Hz, 1H),

518.1
518.1



7.68 (d, J = 1.2 Hz, 1H), 7.39 (d, J = 2.4 Hz, 1H), 7.30 (d, J = 2.4 Hz, 1H), 7.23 (d, J = 8.4 Hz, 2H),





4.59 (br d, J = 8.8 Hz, 1H), 4.39-4.31 (m, 4H), 3.81 (t, J = 6.0 Hz, 2H), 2.89 (s, 3H), 1.64 (s, 6H)




A4

1H-NMR (400 MHz, CDCl3) δ = 8.97 (s, 2H), 7.55 (d, J = 8.0 Hz, 2H), 7.49 (d, J = 2.4 Hz, 1H),

519.1
518.9



7.39-7.32 (m, 3H), 5.84-5.73 (m, 1H), 4.69 (s, 2H), 4.45 (t, J = 6.4 Hz, 2H), 3.89 (t, J = 6.0 Hz,





2H), 3.04 (s, 3H), 1.73 (s, 6H)




A5

1H-NMR (400 MHz, CDCl3) δ = 8.82 (s, 2H), 8.38 (br d, J = 8.4 Hz, 2H), 7.46 (d, J = 2.0 Hz, 1H),

519.1
519.0



7.38 (d, J = 2.0 Hz, 1H), 7.32 (br d, J = 8.4 Hz, 2H), 4.77 (s, 1H), 4.43 (q, J = 6.4, 12.8 Hz, 4H),





3.88 (t, J = 6.0 Hz, 2H), 3.00 (s, 3H), 1.74-1.70 (m, 6H)




A6

1H-NMR (400 MHz, CDCl3) δ = 8.74 (d, J = 2.0 Hz, 1H), 7.88 (dd, J = 2.4, 8.0 Hz, 1H), 7.55-7.51

532.1
532.0



(m, 2H), 7.50 (d, J = 2.4 Hz, 1H), 7.37 (d, J = 2.4 Hz, 1H), 7.34-7.28 (m, 3H), 5.79 (t, J = 5.2 Hz,





1H), 4.45 (t, J = 6.0 Hz, 2H), 3.89 (t, J = 6.0 Hz, 2H), 3.63 (q, J = 6.0 Hz, 2H), 3.17 (t, J = 6.0





Hz, 2H), 2.97 (s, 3H), 1.72 (s, 6H)




A7

1H-NMR (400 MHz, MeOD) δ = 7.37-7.31 (m, 3H), 7.20 (d, J = 2.8 Hz, 1H), 7.04 (d, J = 8.4 Hz,

484.1
484.0



2H), 4.41 (t, J = 5.6 Hz, 2H), 4.26 (q, J = 7.2, 14.4 Hz, 1H), 4.11 (br d, J = 5.2 Hz, 2H), 3.91 (t,





J = 5.6 Hz, 2H), 3.55 (br s, 2H), 3.23-3.05 (m, 2H), 2.95 (s, 3H), 2.82 (t, J = 7.6 Hz, 2H)




A8

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.4 Hz, 1H), 7.35-7.30 (m, 3H), 7.16 (d, J = 8.0 Hz,

524.1
524.0



2H), 4.48 (br d, J = 5.2 Hz, 1H), 4.43 (t, J = 6.0 Hz, 2H), 3.89 (t, J = 6.0 Hz, 2H), 3.67 (s, 2H),





3.44 (br dd, J = 3.2, 7.2 Hz, 1H), 3.02-3.01 (m, 1H), 3.03 (br s, 1H), 2.99 (s, 3H), 2.39-2.29 (m,





2H), 2.07 (br d, J = 11.6 Hz, 2H), 1.82 (br d, J = 10.4 Hz, 2H), 1.67 (s, 6H)




A9

1H-NMR (400 MHz, CDCl3) δ = 7.37 (br d, J = 8.0 Hz, 2H), 7.28 (br s, 1H), 7.07 (d, J = 3.2 Hz,

498.1
498.3



1H), 6.98 (d, J = 8.8 Hz, 2H), 4.41 (t, J = 6.4 Hz, 2H), 4.30 (br d, J = 2.8 Hz, 1H), 3.89 (t, J =





6.0 Hz, 2H), 3.55 (br s, 2H), 3.48-3.31 (m, 1H), 3.00 (s, 3H), 2.89 (br d, J = 10.4 Hz, 2H), 2.21 (br





s, 2H), 2.03 (br d, J = 11.6 Hz, 2H), 1.74-1.60 (m, 2H)




A10

1H-NMR (400 MHz, CDCl3) δ = 7.71-7.69 (m, 1H), 7.63-7.61 (m, 1H), 7.53-7.51 (m, 3H), 7.42

571.1
571.0



(s, 1H), 7.37 (s, 1H), 7.28 (br s, 1H), 7.26 (br s, 1H), 5.20 (s, 2H), 4.46-4.43 (m, 2H), 4.35 (d,





J = 8.4 Hz, 2H), 4.25-4.23 (m, 2H), 3.89 (t, J = 6.0 Hz, 2H), 3.00 (s, 3H), 1.72 (s, 6H).




A11

1H-NMR (400 MHz, CDCl3) δ = 8.37 (d, J = 1.6 Hz, 1H), 7.79 (dd, J = 2.4, 8.8 Hz, 1H), 7.49 (d,

559.1
559.0



J = 2.4 Hz, 1H), 7.46 (d, J = 8.4 Hz, 2H), 7.37 (d, J = 2.4 Hz, 1H), 7.25 (d, J = 8.8 Hz, 2H), 6.46 (d,





J = 8.4 Hz, 1H), 5.41 (s, 1H), 4.57-4.48 (m, 3H), 4.44 (t, J = 6.4 Hz, 2H), 4.11 (br d, J = 2.0 Hz,





2H), 3.89 (t, J = 6.4 Hz, 2H), 3.03 (s, 3H), 1.70 (s, 6H)




A12

1H-NMR (400 MHz, CDCl3) δ = 7.80 (s, 1H), 7.69 (s, 1H), 7.47 (d, J = 2.4 Hz, 1H), 7.42 (d, J = 8.4

521.1
521.0



Hz, 2H), 7.36 (d, J = 2.4 Hz, 1H), 7.19 (d, J = 8.4 Hz, 2H), 5.07 (s, 1H), 4.43 (t, J = 6.4 Hz, 2H), 4.35





(t, J = 5.2 Hz 2H), 3.88 (t, J = 6.4 Hz, 2H), 3.66 (dd, J = 6.0, 10.8 Hz, 2H), 2.94 (s, 3H), 1.69 (s, 6H)




A13

1H-NMR (400 MHz, CDCl3) δ = 7.44 (d, J = 2.4 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.09 (d, J = 8.4

539.2
539.1



Hz, 2H), 6.86 (d, J = 8.8 Hz, 2H), 4.42 (t, J = 6.0 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.43 (br t, J = 5.2





Hz, 2H), 3.40-3.34 (m, 4H), 3.01 (s, 3H), 2.97 (br s, 4H), 2.91 (br t, J = 5.6 Hz, 2H), 1.64 (s, 6H)




A14

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.10 (br d, J =

526.1
526.4



7.6 Hz, 2H), 6.88 (br s, 2H), 4.68 (br t, J = 6.4 Hz, 1H), 4.42 (t, J = 6.0 Hz, 2H), 4.05 (br dd, J = 2.0,





11.2 Hz, 1H), 3.95-3.79 (m, 4H), 3.51-3.35 (m, 3H), 3.28-3.15 (m, 1H), 3.01 (s, 3H), 2.92-2.82 (m,





1H), 2.74-2.62 (m, 1H), 1.64 (s, 6H)




A20

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.0 Hz, 1H), 7.28 (d, J = 2.4 Hz, 1H), 7.17-7.09 (m,

510.1
510.0



4H), 4.42 (t, J = 6.0 Hz, 2H), 4.22 (t, J = 5.6 Hz, 1H), 3.88 (t, J = 6.4 Hz, 2H), 3.80 (t, J = 8.4 Hz,





2H), 3.50-3.41 (m, 2H), 2.95 (s, 3H), 2.92 (d, J = 8.0 Hz, 2H), 2.77-2.71 (m, 2H), 1.65 (s, 6H)




A21

1H-NMR (400 MHz, CDCl3) δ = 8.79 (s, 1H), 7.94 (dd, J = 2.0, 8.0 Hz, 1H), 7.63-7.61 (m, 2H),

492.0
492.0



7.45 (d, J = 8.0 Hz, 1H), 7.33 (d, J = 2.8 Hz, 1H), 7.16-7.14 (m, 3H), 5.71 (s, 1H), 4.55 (d, J = 5.6





Hz, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.91 (t, J = 6.0 Hz, 2H), 2.99 (s, 3H).




A23

1H-NMR (400 MHz, DMSO-d6) δ = 11.64 (s, 1H), 8.64 (d, J = 1.6 Hz, 1H), 8.62 (s, 1H), 8.59 (d,

530.0
530.2



J = 2.8 Hz, 1H), 8.51 (dd, J = 1.6, 8.4 Hz, 1H), 8.26 (d, J = 8.8 Hz, 1H), 7.94 (d, J = 8.8 Hz, 1H),





7.80 (d, J = 2.8 Hz, 1H), 7.74 (d, J = 3.2 Hz, 1H), 7.68 (dd, J = 2.8, 8.4 Hz, 1H), 4.44 (t, J = 4.8





Hz, 2H), 3.99 (t, J = 5.6 Hz 2H), 3.47 (s, 3H)




A24

1H-NMR (400 MHz, CDCl3) δ = 8.89 (br s, 1H), 8.11 (s, 1H), 7.90 (br d, J = 9.2 Hz, 1H), 7.66 (br

529.0
528.9



d, J = 8.4 Hz, 2H), 7.34 (d, J = 2.8 Hz, 1H), 7.19 (s, 1H), 7.18-7.13 (m, 2H), 7.01 (d, J = 6.8 Hz,





1H), 4.44 (t, J = 6.0 Hz, 2H), 3.91 (t, J = 6.0 Hz, 2H), 3.16 (s, 3H)




A25

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.4 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.14-7.08 (m,

525.1
525.3



2H), 6.89-6.83 (m, 2H), 4.49 (br d, J = 8.8 Hz, 1H), 4.42 (t, J = 6.0 Hz, 2H), 4.07-3.96 (m, 1H), 3.88





(t, J = 6.0 Hz, 2H), 3.80 (s, 2H), 2.96-2.92 (m, 3H), 2.62 (ddd, J = 2.8, 8.0, 10.4 Hz, 2H), 1.84 (ddd,





J = 2.8, 8.0, 10.4 Hz, 2H), 1.65 (s, 6H), 1.27 (s, 3H).




A26

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.0 Hz, 1H), 7.31 (d, J = 2.0 Hz, 1H), 7.14 (d, J = 8.4

529.1
529.0



Hz, 2H), 6.90 (d, J = 8.4 Hz, 2H), 4.89 (d, J = 8.8 Hz, 1H), 4.43 (t, J = 6.0 Hz, 2H), 4.27-4.16 (m,





1H), 4.05 (d, J = 12.0 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 2.97 (s, 3H), 2.95-2.82 (m, 2H), 2.53-2.41





(m, 2H), 1.65 (s, 6H)




A27

1H-NMR (400 MHz, CDCl3) δ = 8.43 (br d, J = 2.0 Hz, 1H), 8.13 (s, 1H), 7.89 (br d, J = 8.4 Hz,

528.1
528.0



1H), 7.65 (br d, J = 8.4 Hz, 2H), 7.59-7.42 (m, 1H), 7.33 (d, J = 2.8 Hz, 1H), 7.19-7.17 (m, 3H),





5.89 (s, 1H), 4.39 (t, J = 6.0 Hz, 2H), 3.90 (t, J = 6.0 Hz, 2H), 3.27 (br s, 3H).




A28

1H-NMR (400 MHz, CDCl3) δ = 8.44 (s, 1H), 8.17 (s, 1H), 7.91 (br d, J = 8.4 Hz, 1H), 7.66 (br d,

584.1
584.1



J = 8.0 Hz, 2H), 7.53 (br s, 1H), 7.19-7.17 (m, 3H), 7.00 (d, J = 2.4 Hz, 1H), 4.36 (t, J = 6.0 Hz,





2H), 3.87 (t, J = 6.0 Hz, 2H), 3.26 (br s, 3H), 1.45 (s, 9H).




A29

1H-NMR (400 MHz, MeOD) δ = 7.51 (d, J = 2.4 Hz, 1H), 7.47 (d, J = 2.4 Hz, 1H), 7.18 (s, 4H),

538.2
538.0



4.42 (t, J = 5.6 Hz, 2H), 3.90 (t, J = 5.6 Hz, 2H), 3.31-3.25 (m, 1H), 2.99 (d, J = 12.0 Hz, 2H), 2.95





(s, 3H), 2.82-2.78 (m, 2H), 2.62-2.58 (m, 2H), 2.22 (t, J = 11.6 Hz, 2H), 1.98 (d, J = 11.2 Hz, 2H),





1.66 (s, 6H), 1.64-1.60 (m, 2H)




A30

1H-NMR (400 MHz, CDCl3) δ = 7.28 (d, J = 8.8 Hz, 2H), 7.26 (s, 1H), 7.04 (d, J = 2.8 Hz, 1H),

512.1
512.0



6.98 (d, J = 8.4 Hz, 2H), 5.12 (d, J = 2.0 Hz, 1H), 4.42 (t, J = 6.0 Hz, 2H), 3.91 (t, J = 6.0 Hz, 2H),





3.51 (br s, 1H), 3.31 (d, J = 11.6 Hz, 2H), 3.03 (s, 3H), 3.00-2.90 (m, 4H), 2.54 (t, J = 10.8 Hz, 2H),





2.16 (d, J = 11.6 Hz, 2H), 1.96 (q, J = 10.0 Hz, 2H).




A31

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.35-7.30 (m, 1H), 7.10 (br d, J = 8.4

540.1
540.0



Hz, 2H), 6.95-6.81 (m, 2H), 4.94 (br dd, J = 4.8, 6.4 Hz, 1H), 4.42 (t, J = 6.0 Hz, 2H), 4.04 (dd,





J = 2.4, 11.2 Hz, 1H), 3.88 (t, J = 6.4 Hz, 2H), 3.85-3.74 (m, 2H), 3.49-3.36 (m, 3H), 3.35-3.25





(m, 1H), 2.97 (s, 3H), 2.93-2.81 (m, 1H), 2.67-2.54 (m, 1H), 1.88-1.79 (m, 2H), 1.64 (s, 6H)




A32

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.0 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 7.09 (d, J =

566.2
566.4



8.8 Hz, 2H), 6.86 (d, J = 8.8 Hz, 2H), 4.42 (t, J = 6.4 Hz, 2H), 3.88 (t, J = 6.0 Hz, 4H), 3.61 (br d,





J = 11.6 Hz, 2H), 3.19-3.12 (m, 2H), 3.08-3.01 (m, 2H), 2.99 (s, 2H), 2.81 (s, 3H), 2.23 (br d,





J = 13.6 Hz, 2H), 1.72-1.66 (m, 2H), 1.64 (s, 6H)




A33

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.4 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.08 (d, J =

525.1
525.0



6.4 Hz, 2H), 6.81 (d, J = 6.8 Hz, 2H), 4.42 (t, J = 6.0 Hz, 2H), 4.15 (d, J = 7.2 Hz, 1H), 4.22-





4.14 (m, 1H), 3.88 (t, J = 6.0 Hz, 2H), 3.45-3.43 (m, 1H), 3.01 (s, 3H), 2.20-2.13 (m, 4H),





1.64 (s, 6H), 1.60-1.59 (m, 2H), 1.45-1.42 (m, 2H)




A34

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.4 Hz, 1 H), 7.34 (d, J = 2.0 Hz, 1 H), 7.27-7.24

560.1
560.0



(m, 2 H), 7.15 (d, J = 8.0 Hz, 2 H), 4.60 (d, J = 8.0 Hz, 1 H), 4.43 (t, J = 6.0 Hz, 2 H), 3.88 (t, J =





6.0 Hz, 2 H), 3.67-3.64 (m, 2 H), 3.51-3.60 (m, 1 H), 3.58-3.55 (m, 1 H), 3.23-3.17 (m, 1 H),





3.06 (s, 3 H), 2.95 (d, J = 10.8 Hz, 1 H), 2.33-2.27 (m, 1 H), 2.24 (t, J = 11.2 Hz, 1 H), 2.10-2.07





(m, 1 H), 1.84-1.82 (m, 1 H), 1.67 (s, 6 H).




A35

1H-NMR (400 MHz, CDCl3) δ = 7.36 (br d, J = 8.4 Hz, 2H), 7.28 (d, J = 2.8 Hz, 1H), 7.08 (d, J =

534.1
533.9



3.2 Hz, 1H), 6.99 (d, J = 8.4 Hz, 2H), 4.6 (br s, 1H), 4.41 (t, J = 6.0 Hz, 2H), 3.90 (t, J = 6.0 Hz,





2H), 3.75-3.55 (m, 3H), 3.26-3.15 (m, 1H), 3.07 (s, 3H), 2.96 (br d, J = 10.8 Hz, 1H), 2.47-2.21





(m, 2H), 2.15-2.06 (m, 1H), 1.91-1.78 (m, 1H)




A37

1H-NMR (400 MHz, CDCl3) δ = 8.50 (s, 1H), 7.90 (s, 1H), 7.83 (d, J = 8.4 Hz, 2H), 7.47 (s, 1H),

560.1
560.0



7.38 (d, J = 1.6 Hz, 1H), 7.30-7.27 (m, 2H), 4.94 (d, J = 6.4 Hz, 1H), 4.55 (d, J = 5.6 Hz, 3H),





4.43 (t, J = 6.0 Hz, 2H), 4.08-4.05 (m, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.04 (s, 3H), 1.70 (s, 6H).




A38

1H-NMR (400 MHz, CDCl3) δ = 8.39 (d, J = 2.0 Hz, 1H), 7.70 (dd, J = 2.4, 8.4 Hz, 1H), 7.54-

533.1
533.0



7.51 (m, 2H), 7.30 (d, J = 2.8 Hz, 1H), 7.11 (d, J = 3.2 Hz, 1H), 7.11-7.07 (m, 2H), 6.42 (d, J =





8.8 Hz, 1H), 4.92 (d, J = 8.0 Hz, 1H), 4.49-4.47 (m, 3H), 4.41 (t, J = 6.0 Hz, 2H), 3.97-3.96 (m,





2H), 3.90 (t, J = 6.4 Hz, 2H), 3.03 (s, 3H)




A39

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.0 Hz, 1H), 7.32 (d, J = 2.0 Hz, 1H), 7.12 (d, J =

529.1
529.0



8.4 Hz, 2H), 6.88 (d, J = 8.4 Hz, 2H), 4.58 (br d, J = 8.8 Hz, 1H), 4.43 (t, J = 6.0 Hz, 2H),





4.05 (d, J = 18.8 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.70 (dq, J = 3.6, 8.0 Hz, 1H), 3.00-2.93 (m,





5H), 2.49-2.36 (m, 2H), 1.65 (s, 6H)




A40

1H-NMR (400 MHz, CDCl3) δ = 8.39 (d, J = 5.2 Hz, 1H), 7.99 (d, J = 8.0 Hz, 2H), 7.45 (s,

560.1
560.0



1H), 7.36 (d, J = 1.2 Hz, 1H), 7.30 (d, J = 8.0 Hz, 2H), 7.04 (d, J = 5.2 Hz, 1H), 4.89 (d, J = 9.2





Hz, 1H), 4.61-4.57 (m, 2H), 4.45-4.42 (m, 1H), 4.43 (t, J = 6.0 Hz, 2H), 4.09 (dd, J = 4.8, 9.2 Hz,





2H), 3.88 (t, J = 6.0 Hz, 2H), 3.02 (s, 3H), 1.71 (s, 6H).




A41

1H-NMR (400 MHz, CDCl3) δ = 8.37 (s, 1H), 7.93 (d, J = 8.0 Hz, 2H), 7.76 (s, 1H), 7.47 (d, J =

560.1
560.0



2.4 Hz, 1H), 7.37 (d, J = 2.4 Hz, 1H), 7.30 (d, J = 8.4 Hz, 2H), 5.18 (br d, J = 6.4 Hz, 1H), 4.57 (t,





J = 4.8 Hz, 3H), 4.44 (t, J = 6.0 Hz, 2H), 4.09 (d, J = 4.4 Hz, 2H), 3.89 (t, J = 6.0 Hz, 2H),





3.04 (s, 3H), 1.71 (s, 6H).




A43

1H-NMR (400 MHz, CDCl3) δ = 8.44 (br s, 1H), 8.15 (s, 1H), 7.89 (br d, J = 8.0 Hz, 1H), 7.70 (d,

481.1
481.0



J = 8.8 Hz, 2H), 7.54 (br d, J = 10.8 Hz, 1H), 7.34 (d, J = 3.2 Hz, 1H), 7.19-7.12 (m, 3H), 4.06 (s,





3H), 3.28 (br s, 3H).




A46

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.4 Hz, 1H), 7.33 (d, J = 2.0 Hz, 1H), 7.26-7.22 (m,

560.1
560.0



2H), 7.19-7.13 (m, 2H), 5.02 (d, J = 9.6 Hz, 1H), 4.45 (t, J = 6.0 Hz, 2H), 3.90 (t, J = 6.0 Hz, 3H),





3.65 (d, J = 0.8 Hz, 2H), 3.07-3.03 (m, 1H), 2.90 (s, 3H), 2.85 (d, J = 12.0 Hz, 1H), 2.48-2.40 (m,





2H), 2.28-2.26 (m, 1H), 2.03-2.01 (m, 1H), 1.68 (s, 6H)




A47

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.33 (d, J = 2.4 Hz, 1H), 7.04 (d, J =

565.2
565.1



11.6 Hz, 2H), 6.86 (d, J = 8.8 Hz, 2H), 4.87-4.71 (br s, 1H), 4.41 (t, J = 6.0 Hz, 2H), 4.10 (br s,





1H), 3.88 (t, J = 6.0 Hz, 2H), 3.71 (q, J = 6.8, 8.4 Hz, 2H), 3.59 (td, J = 3.6, 12.4 Hz, 2H), 2.97 (t,





J = 8.4 Hz, 5H), 2.83-2.75 (m, 2H), 2.20-2.12 (m, 1H), 1.78 (br dd, J = 3.2, 12.8 Hz, 2H), 1.63 (s,





6H), 1.50-1.38 (m, 2H)




A48

1H-NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.4 Hz, 1H), 7.37-7.32 (m, 1H), 7.09 (br d, J = 8.8

565.2
565.1



Hz, 2H), 6.88 (br d, J = 8.8 Hz, 2H), 4.94-4.81 (m, 0.2H), 4.50 (br d, J = 7.2 Hz, 1H), 4.43 (t,





J = 6.0 Hz, 2H), 4.16-4.03 (m, 1H), 3.89 (t, J = 6.0 Hz, 2H), 3.85-3.74 (m, 0.2H), 3.31-3.20 (m,





4H), 3.04-2.94 (m, 4H), 2.79-2.68 (m, 1H), 2.66-2.60 (m, 1H), 2.56 (br d, J = 4.4 Hz, 4H), 2.52-





2.46 (m, 1H), 2.20-2.12 (m, 2H), 1.65 (s, 6H)




A50

1H-NMR (400 MHz, CDCl3) δ = 8.20 (s, 1H), 7.48 (d, J = 2.4 Hz, 1H), 7.44-7.32 (m, 3H), 7.37

577.1
577.0



(d, J = 2.0 Hz, 1H), 7.25 (d, J = 8.4 Hz, 2H), 4.87 (d, J = 8.8 Hz, 1H), 4.56-4.54 (m, 3H), 4.43 (t,





J = 6.0 Hz, 2H), 4.06 (dd, J = 5.2, 8.4 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.03 (s, 3H), 1.70 (s, 6H).




A52

1H-NMR (400 MHz, CDCl3) δ = 9.26-9.18 (m, 1H), 8.66 (d, J = 5.2 Hz, 1H), 7.41 (d, J = 2.4 Hz,

527.1
527.1



1H), 7.30-7.29 (m, 2H), 7.13 (d, J = 4.8 Hz, 2H), 6.90 (d, J = 4.8 Hz, 2H), 5.12 (s, 2H), 4.80-4.75





(m, 1H), 3.47 (s, 3H), 2.41-2.35 (m, 4H), 1.82-1.78 (m, 1H), 1.64 (s, 6H), 1.50-1.48 (m, 1H)




A53

1H-NMR (400 MHz, CDCl3) δ = 8.35 (d, J = 2.0 Hz, 1H), 7.68 (dd, J = 2.4, 8.4 Hz, 1H), 7.49 (d,

559.1
559.0



J = 2.4 Hz, 1H), 7.47-7.43 (m, 2H), 7.37 (d, J = 2.4 Hz, 1H), 7.26-7.22 (m, 2H), 6.50 (d, J = 8.8





Hz, 1H), 4.82-4.71 (m, 2H), 4.44 (t, J = 6.0 Hz, 2H), 4.28 (dd, J = 7.2, 8.4 Hz, 2H), 3.94 (dd, J =





6.0, 8.8 Hz, 2H), 3.89 (t, J = 6.4 Hz, 2H), 2.93 (s, 3H), 1.70 (s, 6H)




A54

1H-NMR (400 MHz, CDCl3) δ = 8.43 (d, J = 2.4 Hz, 1H), 7.72 (dd, J = 2.4, 8.8 Hz, 1H), 7.50 (d,

573.1
573.1



J = 2.4 Hz, 1H), 7.46 (d, J = 8.4 Hz, 2H), 7.38 (d, J = 2.4 Hz, 1H), 7.23 (d, J = 8.4 Hz, 2H), 6.47





(d, J = 8.8 Hz, 1H), 4.54 (d, J = 13.6 Hz, 1H), 4.44 (t, J = 6.0 Hz, 2H), 4.44-4.42 (m, 1H), 3.90-





3.85 (m, 3H), 3.71-3.64 (m, 1H), 3.61-3.51 (m, 2H), 3.05 (s, 3H), 2.45-2.38 (m, 1H), 2.15-





2.10 (m, 1H), 1.70 (s, 6H).




A58

1H-NMR (400 MHz, CDCl3) δ = 8.62 (d, J = 1.6 Hz, 1H), 7.85 (dd, J = 8.4, 2.4 Hz, 1H), 7.51 (d,

587.2
587.1



J = 8.4 Hz, 2H), 7.48 (d, J = 2.0 Hz, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.35 (d, J = 2.4 Hz, 1H), 7.30





(d, J = 8.4 Hz, 2H), 4.36 (t, J = 5.2 Hz, 2H), 3.49-3.38 (m, 1H), 3.08-3.18 (m, 2H), 3.07-2.95 (m,





5H), 2.50-2.41 (m, 2H), 2.06 (d, J = 12.0 Hz, 2H), 1.71 (s, 8H).




A60

1H-NMR (400 MHz, CDCl3) δ = 8.46 (d, J = 2.4 Hz, 1H), 7.77 (dd, J = 2.4, 8.8 Hz, 1H), 7.50-

573.1
575.1



7.47 (m, 3H), 7.38 (d, J = 2.4 Hz, 1H), 7.25 (d, J = 8.4 Hz, 2H), 6.77 (d, J = 8.8 Hz, 1H), 4.44 (t,





J = 6.0 Hz, 2H), 3.89 (t, J = 6.0 Hz, 2H), 3.76 (t, J = 5.2 Hz, 4H), 3.42-3.34 (t, J = 5.2, 4H), 2.83





(s, 3H), 1.70 (s, 6H)




A62

1H-NMR (400 MHz, CDCl3) δ = 9.42-9.37 (m, 1H), 8.66 (d, J = 5.2 Hz, 1H), 7.42 (d, J = 2.4 Hz,

513.1
513.3



1H), 7.31 (d, J = 2.0 Hz, 1H), 7.29 (d, J = 5.2 Hz, 1H), 7.14 (d, J = 8.8 Hz, 2H), 6.91 (d, J = 8.8





Hz, 2H), 5.12 (s, 2H), 4.50-4.45 (m, 1H), 3.47 (s, 3H), 1.65 (s, 6H), 1.02-1.01 (m, 2H), 0.73-





0.71 (m, 2H).




A63

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.09 (d, J =

539.1
539.1



8.8 Hz, 2H), 6.82 (d, J = 8.8 Hz, 2H), 4.42 (t, J = 6.4 Hz, 2H), 4.13 (d, J = 7.6 Hz, 1H), 3.88 (t,





J = 6.0 Hz, 2H), 3.76 (d, J = 6.0 Hz, 2H), 3.34-3.30 (m, 1H), 3.00 (s, 3H), 2.17-2.13 (m, 2H),





2.00-1.97 (m, 2H), 1.82-1.72 (m, 1H), 1.64 (s, 6H), 1.34-1.20 (m, 4H)




A64

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.0 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.09 (d, J =

525.1
525.0



8.8 Hz, 2H), 6.85 (d, J = 8.8 Hz, 2H), 4.42 (t, J = 6.0 Hz, 2H), 4.19 (br t, J = 5.2 Hz, 1H), 3.98





(d, J = 6.4 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.25 (t, J = 6.4 Hz, 2H), 2.98 (s, 3H), 2.77-2.75 (m,





1H), 2.57 (td, J = 7.2, 14.4 Hz, 1H), 2.09-2.06 (m, 2H), 1.98-1.94 (m, 2H), 1.65 (s, 6H)




A67

1H-NMR (400 MHz, CDCl3) δ = 8.44 (br s, 1H), 8.16 (s, 1H), 7.90 (br d, J = 8.8 Hz, 1H), 7.71

562.1
562.0



(d, J = 8.4 Hz, 2H), 7.54-7.53 (m, 1H), 7.22 (t, J = 2.8 Hz, 2H) 7.19 (s, 1H), 7.05 (d, J = 2.8 Hz,





1H), 4.31 (q, J = 8.4 Hz, 2H), 4.04 (s, 3H), 3.27 (br s, 3H)




A68

1H-NMR (400 MHz, CDCl3) δ = 8.59 (s, 2H), 7.52 (d, J = 8.8 Hz, 2H), 7.20 (d, J = 8.8 Hz, 2H),

615.1
615.0



7.15 (d, J = 2.8 Hz, 1H), 6.96 (d, J = 3.2 Hz, 1H), 4.87 (br d, J = 8.8 Hz, 1H), 4.60-4.58 (m, 2H),





4.55-4.45 (m, 1H), 4.39 (t, J = 6.4 Hz, 2H), 4.28 (q, J = 8.8 Hz, 2H), 4.11 (dd, J = 4.8, 10.0 Hz,





2H), 3.88 (t, J = 6.0 Hz, 2H), 3.04 (s, 3H).




A69

1H-NMR (400 MHz, CDCl3) δ = 7.91 (d, J = 8.4 Hz, 2H), 7.66 (d, J = 9.2 Hz, 1H), 7.47 (d, J =

560.1
560.1



2.0 Hz, 1H), 7.37 (d, J = 2.0 Hz, 1H), 7.30 (br d, J = 8.4 Hz, 2H), 6.67 (br d, J = 9.2 Hz, 1H),





5.74 (br d, J = 1.2 Hz, 1H), 4.59-4.56 (m, 3H), 4.44 (t, J = 6.0 Hz, 2H), 4.19 (br d, J = 4.0 Hz,





2H), 3.89 (t, J = 6.0 Hz, 2H), 3.03 (s, 3H), 1.71 (s, 6H)




A71

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 2.0 Hz, 1H), 7.10 (d, J =

537.1
537.1



6.4 Hz, 2H), 6.83 (d, J = 6.8 Hz, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.94 (d, J = 8.4 Hz, 4H), 3.89-3.86





(m, 4H), 2.85 (s, 3H), 2.69-2.63 (m, 1H), 2.43-2.38 (m, 2H), 2.20-2.15 (m, 2H), 1.65 (s, 6H)




A72

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 7.07 (d, J =

523.1
523.0



6.4 Hz, 2H), 6.70 (d, J = 6.4 Hz, 2H), 4.59 (quin, J = 6.4 Hz, 1H), 4.42 (t, J = 6.0 Hz, 2H), 3.98





(s, 2H), 3.96 (s, 2H), 3.88 (t, J = 6.0 Hz, 2H), 2.86 (s, 3H), 2.78-2.72 (m, 2H), 2.42-2.37 (m, 2H),





1.64 (s, 6H)




A74

1H-NMR (400 MHz, CDCl3) δ = 7.45 (s, 1H), 7.32 (s, 1H), 7.09 (d, J = 8.8 Hz, 2H), 6.84 (d,

525.1
525.0



J = 8.8 Hz, 2H), 4.42 (t, J = 6.0 Hz, 2H), 4.15-4.14 (m, 1H), 3.90-3.87 (m, 4H), 3.18 (t, J = 6.4





Hz, 2H), 2.97 (s, 3H), 2.80-3.65 (m, 1H), 2.55-2.45 (m, 1H), 2.28-2.25 (m, 2H), 1.71-1.68 (m,





2H), 1.64 (s, 6H)




A76
NA
560.1
560.3


A78
NA
521.1
521.1


A79
NA
521.1
520.9


A87
NA
576.1
576


A100

1H-NMR (400 MHz, CDCl3) δ = 8.67 (d, J = 5.2 Hz, 1H), 7.92 (d, J = 4.0 Hz, 1H), 7.63 (d, J =

603.1
603.1



5.2 Hz, 1H), 7.45 (d, J = 1.6 Hz, 1H), 7.32 (d, J = 2.0 Hz, 1H), 7.26 (s, 1H), 7.10 (d, J = 4.0 Hz,





2H), 1H), 7.09 (t, J = 1.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 2H), 5.12 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H),





3.88 (t, J = 6.0 Hz, 2H), 1.65 (s, 6H)




A101

1H-NMR (400 MHz, CDCl3) δ = 10.05 (s, 1H), 8.66 (d, J = 4.8 Hz, 1H), 8.31 (d, J = 1.6 Hz, 1H),

603.1
603.0



7.59 (d, J = 4.4 Hz, 1H), 7.45 (d, J = 6.0 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 1.6 Hz,





1H), 7.23 (d, J = 5.2 Hz, 1H), 7.11 (d, J = 8.8 Hz, 2H), 6.87 (d, J = 8.8 Hz, 2H), 5.06 (s, 2H),





4.43 (t, J = 6.0 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 1.65 (s, 6H)




A102

1H-NMR (400 MHz, CDCl3) δ = 8.21 (d, J = 6.4 Hz, 1H), 7.49 (d, J = 2.4 Hz, 1H), 7.42 (d, J =

499.1
499.0



8.4 Hz, 2H), 7.37 (d, J = 2.4 Hz, 1H), 7.22 (d, J = 8.4 Hz, 2H), 6.66 (dd, J = 4.8, 14.0 Hz, 1H),





6.33 (d, J = 6.4 Hz, 1H), 5.48 (s, 2H), 4.46-4.43 (m, 1H), 4.38 (dd, J = 2.8, 13.6 Hz, 1H), 3.22 (s,





3H), 1.69 (s, 6H)




A103

1H-NMR (400 MHz, DMSO-d6) δ = 8.37 (br s, 1H), 8.24-8.19 (m, 1H), 7.65-7.62 (m, 2H), 7.56

587.1
587.0



(d, J = 2.0 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.92 (d, J = 8.8 Hz, 2H), 6.83-6.79 (m, 2H), 4.98 (s,





2H), 4.41 (t, J = 5.2 Hz, 2H), 3.95 (t, J = 5.2 Hz, 2H), 1.62 (s, 6H).




A104

1H-NMR (400 MHz, CDCl3) δ = 9.10 (s, 1H), 8.82 (d, J = 5.2 Hz, 1H), 8.14 (s, 1H), 7.64 (d,

587.1
587.0



J = 4.8 Hz, 1H), 7.46 (d, J = 2.0 Hz, 1H), 7.32 (d, J = 2.0 Hz, 1H), 7.15 (d, J = 8.8 Hz, 2H), 6.93





(d, J = 9.2 Hz, 2H), 5.27 (s, 2H), 4.98 (s, 2H, 4.43 (t, J = 6.0 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H),





1.66 (s, 6H).




A105

1H-NMR (400 MHz, CDCl3) δ = 9.41 (s, 1H), 8.63 (d, J = 5.2 Hz, 1H), 8.04 (d, J = 3.2 Hz, 2H),

601.1
601.0



7.45 (s, 1H), 7.32 (s, 1H), 7.23 (d, J = 5.2 Hz, 1H), 7.13 (d, J = 8.4 Hz, 2H), 6.89 (d, J = 8.4 Hz,





2H), 5.09 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.93 (s, 3H), 3.88 (t, J = 6.0 Hz, 2H), 1.65 (s, 6H).




A106

1H-NMR (400 MHz, DMSO-d6) δ = 8.93 (d, J = 5.2 Hz, 1H), 8.67 (s, 1H), 8.22 (s, 1H), 7.64 (d,

587.1
587.1



J = 5.2 Hz, 2H), 7.57 (d, J = 2.4 Hz, 1H), 7.46 (s, 2H), 7.21 (d, J = 8.8 Hz, 2H), 7.02 (d, J = 8.8





Hz, 2H), 5.30 (s, 2H), 4.41 (t, J = 5.2 Hz, 2H), 3.95 (t, J = 5.2 Hz, 2H), 1.64 (s, 6H).




A107

1H-NMR (400 MHz, CDCl3) δ = 9.66 (s, 1H), 8.58 (d, J = 5.2 Hz, 1H), 7.74 (s, 1H), 7.47 (s, 1H),

601.1
601.1



7.44 (d, J = 2.0 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 7.17 (d, J = 2.0 Hz, 1H), 7.10 (d, J = 8.8 Hz,





2H), 6.88 (d, J = 8.4 Hz, 2H), 5.05 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88 (d, J = 6.0 Hz, 2H),





3.75 (s, 3H), 1.64 (s, 6H).




A108

1H-NMR (400 MHz, CDCl3) δ = 9.33 (s, 1H), 8.88 (d, J = 4.8 Hz, 1H), 7.73 (d, J = 5.2 Hz, 1H),

588.1
588.1



7.46 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 2.0 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.94 (d, J = 8.8 Hz,





2H), 5.61 (s, 2H), 5.29 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88 (d, J = 6.0 Hz, 2H), 1.66 (s, 6H).




A109

1H-NMR (400 MHz, CDCl3) δ = 12.29 (s, 1H), 9.15 (d, J = 2.0 Hz, 1H), 8.79 (d, J = 3.6 Hz),

598.1
598.2



8.51 (d, J = 5.2 Hz, 1H), 8.38 (d, J = 8.4 Hz, 1H), 7.64-7.62 (m, 2H), 7.56 (d, J = 2.0 Hz, 1H),





7.16 (d, J = 8.8 Hz, 2H), 7.13 (d, J = 5.6 Hz, 1H), 6.90 (d, J = 8.8 Hz, 2H), 5.04 (s, 2H), 4.41 (t,





J = 5.2 Hz, 2H), 3.95 (d, J = 5.2 Hz, 2H), 1.62 (s, 6H)




A110

1H-NMR (400 MHz, CDCl3) δ = 10.13 (s, 1H), 8.69 (d, J = 4.0 Hz, 1H), 8.56 (d, J = 5.2 Hz, 1H),

598.1
598.1



8.34 (d, J = 8.0 Hz, 1H), 7.96-7.95 (m, 1H), 7.53-7.51 (m, 1H), 7.44 (d, J = 2.4 Hz, 1H), 7.31 (d,





J = 2.0 Hz, 1H), 7.18 (d, J = 5.2 Hz, 1H), 7.09 (d, J = 8.8 Hz, 2H), 6.83 (d, J = 8.8 Hz, 2H), 4.97





(s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88 (d, J = 6.0 Hz, 2H), 1.64 (s, 6H).




A111

1H NMR (400 MHz, DMSO-d6) δ = 8.30-8.37 (m, 1H), 8.17-8.23 (m, 1H), 7.62-7.67 (m, 1H),

564.1
564.3



7.54-7.60 (m, 1H), 7.26-7.32 (m, 1H), 7.19-7.25 (m, 1H), 7.07-7.17 (m, 1H), 6.66-6.74 (m, 1H),





5.22-5.27 (m, 2H), 4.37-4.47 (m, 2H), 3.99-4.03 (m, 2H), 3.93-3.98 (m, 2H), 2.95 (s, 3H),





1.64 (s, 6H)




A112

1H NMR (400 MHz, DMSO-d6) δ = 11.19-11.67 (m, 1H), 8.60-8.68 (m, 1H), 7.61-7.67 (m, 1H),

733.2
733.1



7.53-7.60 (m, 1H), 7.23-7.30 (m, 1H), 7.14-7.19 (m, 1H), 7.08-7.13 (m, 1H), 6.93-7.00 (m, 1H),





5.11-5.19 (m, 2H), 4.38-4.49 (m, 2H), 3.92-4.01 (m, 2H), 3.53-3.60 (m, 2H), 3.36 (s, 3H), 3.23-





3.29 (m, 4H), 2.28-2.35 (m, 4H), 1.63 (s, 6H), 1.40 (s, 9H)




A113

1H NMR (400 MHz, DMSO-d6) δ = 8.45 (d, J = 5.2 Hz, 1H), 8.20-8.26 (m, 1H), 7.60-7.65 (m,

633.2
633.1



1H), 7.50-7.58 (m, 1H), 7.08-7.17 (m, 2H), 6.90-7.04 (m, 2H), 4.98-5.07 (m, 2H), 4.36-4.45 (m,





2H), 3.92-3.97 (m, 2H), 3.57 (s, 2H), 3.14 (s, 3H), 2.85-2.93 (m, 4H), 2.52-2.54 (m, 4H), 1.63





(s, 6H)




A114

1H NMR (400 MHz, METHANOL) δ = 7.54-7.45 (m, 2H), 7.22-7.14 (m, 2H), 6.98-6.92 (m,

618.2
618.1



2H), 6.74-6.68 (m, 1H), 5.00 (s, 2H), 4.47-4.39 (m, 2H), 3.94-3.88 (m, 2H), 3.20 (s, 3H), 3.15-





3.09 (m, 2H), 2.73-2.63 (m, 3H), 1.89-1.82 (m, 2H), 1.71-1.67 (m, 2H), 1.67 (s, 6H)




A115

1H NMR (400 MHz, DMSO-d6) δ = 11.32-11.22 (m, 1H), 7.67-7.58 (m, 1H), 7.58-7.50 (m,

718.2
718.1



1H), 7.22-7.10 (m, 3H), 7.02-6.93 (m, 2H), 5.11-5.03 (m, 2H), 4.46-4.35 (m, 2H), 4.08-3.97 (m,





2H), 3.96-3.92 (m, 2H), 3.35 (s, 3H), 2.90-2.72 (m, 3H), 1.84-1.76 (m, 2H), 1.62 (s, 6H), 1.56-





1.49 (m, 2H), 1.40 (s, 9H)




A116

1H NMR (400 MHz, DMSO-d6) δ = 8.12-8.25 (m, 1H), 7.61-7.65 (m, 1H), 7.54-7.58 (m, 1H),

619.2
619.1



7.17-7.22 (m, 2H), 6.93-7.01 (m, 2H), 6.47-6.51 (m, 1H), 4.84-4.89 (m, 2H), 4.38-4.45 (m,





2H), 3.93-3.98 (m, 2H), 3.56-3.63 (m, 4H), 3.15 (s, 3H), 2.74-2.79 (m, 4H), 1.63 (s, 6H)




A117

1H NMR (400 MHz, CDCl3) δ = 9.11-9.76 (m, 1H), 7.32-7.38 (m, 1H), 7.22-7.26 (m, 1H),

719.2
719.1



7.00-7.08 (m, 2H), 6.81-6.90 (m, 2H), 6.30-6.35 (m, 1H), 4.90-5.04 (m, 2H), 4.23-4.39 (m,





2H), 3.80 (t, J = 6.4 Hz, 2H), 3.63 (br s, 4H), 3.40-3.53 (m, 4H), 3.30 (s, 3H), 1.56 (s, 6H),





1.38-1.44 (m, 9H)




A119

1H NMR (400 MHz, DMSO-d6) δ = 11.36 (br s, 1H), 8.63 (d, J = 5.2 Hz, 1H), 7.62 (d, J = 2.4

565.1
547.2



Hz, 1H), 7.56 (d, J = 2.4 Hz, 1H), 7.30 (d, J = 2.4 Hz, 1H), 7.22 (br d, J = 4.8 Hz, 1H), 7.08 (dd,

(M + H − H2O)



J = 2.4, 8.4 Hz, 1H), 6.90 (d, J = 8.6 Hz, 1H), 5.14 (s, 2H), 5.05 (t, J = 5.6 Hz, 1H), 4.60 (d,





J = 5.6 Hz, 2H), 4.47-4.37 (m, 2H), 4.00-3.91 (m, 2H), 3.35 (s, 3H), 1.63 (s, 6H)




A120

1H-NMR (400 MHz, CDCl3) δ = 7.62-7.57 (m, 2H), 7.43 (dd, J = 1.6, 8.0 Hz, 1H), 7.38 (s, 1H),

517.1
517.0



7.31 (d, J = 2.8 Hz, 1H), 7.20 (d, J = 8.0 Hz, 1H), 7.12 (d, J = 2.8 Hz, 1H), 7.11-7.07 (m, 2H),





4.53 (s, 2H), 4.42 (t, J = 6.0 Hz, 2H), 3.90 (t, J = 6.0 Hz, 2H), 3.62 (t, J = 6.0 Hz, 2H), 3.07 (t,





J = 6.0 Hz, 2H), 2.89 (s, 3H)




A121
NA
563.1
563.0


A122

1H-NMR (400 MHz, CDCl3) δ = 8.66 (d, J = 4.8 Hz, 1H), 8.63-8.56 (m, 1H), 7.30 (d, J = 5.2 Hz,

509.0
508.7



1H), 7.23-7.21 (m, 1H), 7.04-6.96 (m, 5H), 5.12 (s, 2H), 4.39 (t, J = 6.0 Hz, 2H), 3.89 (t, J = 6.0





Hz, 2H), 3.49 (s, 3H).




A123
NA
531.0
530.9


A124
NA
555.1
554.9


A125

1H-NMR (400 MHz, CDCl3) δ = 7.48-7.41 (m, 2H), 7.31 (d, J = 2.0 Hz, 1H), 7.11 (d, J = 8.8 Hz,

552.1
552.0



3H), 6.91 (d, J = 8.8 Hz, 2H), 5.12 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H),





3.50 (s, 3H), 1.65 (s, 6H).




A126
NA
554.2
554.0


A128
NA
521.0
520.9


A129
NA
501.1
501.0


A130

1H-NMR (400 MHz, CDCl3) δ = 7.61-7.54 (m, 1H), 7.46 (d, J = 2.0 Hz, 1H), 7.39-7.31 (m, 2H),

551.1
551.0



7.24-7.17 (m, 1H), 7.13 (d, J = 8.8 Hz, 2H), 6.93 (d, J = 8.8 Hz, 2H), 6.58 (br s, 1H), 5.13 (s,





2H), 4.43 (t, J = 6.4 Hz, 2H), 3.88 (t, J = 6.4 Hz, 2H), 3.07 (s, 3H), 1.65 (s, 6H).




A131
NA
551.1
551.0


A132

1H-NMR (400 MHz, CDCl3) δ = 9.03 (br s, 1H), 8.64 (d, J = 5.2 Hz, 1H), 7.39 (d, J = 2.4 Hz,

576.2
576.1



1H), 7.32-7.28 (m, 2H), 7.13 (d, J = 8.8 Hz, 2H), 6.93-6.87 (m, 2H), 5.11 (s, 2H), 3.47 (s, 3H),





3.41 (br t, J = 5.2 Hz, 4H), 2.24-2.10 (m, 4H), 1.64 (s, 6H).




A133
NA
566.2
566.2


A134
NA
623.2
623.1


A135

1H-NMR (400 MHz, MeOD) δ = 8.44 (d, J = 5.6 Hz, 1H), 7.55 (s, 2H), 7.34 (d, J = 8.8 Hz,

535.1
534.7



2H), 7.15 (d, J = 8.8 Hz, 2H), 6.73 (d, J = 6.0 Hz, 1H), 4.44 (t, J = 5.6 Hz, 2H), 3.90 (t, J = 5.6





Hz, 2H), 3.35 (s, 3H), 3.00 (s, 3H), 1.71 (s, 6H).




A136

1H-NMR (400 MHz, CDCl3) δ = 9.77 (s, 1H), 8.68 (d, J = 5.2 Hz, 1H), 7.46 (d, J = 2.0 Hz, 1H),

519.1
519.1



7.40 (d, J = 2.0 Hz, 1H), 7.30 (d, J = 4.8 Hz, 1H), 7.13 (d, J = 8.8 Hz, 2H), 6.91 (d, J = 9.2 Hz,





2H), 5.13 (s, 2H), 3.78 (t, J = 7.6 Hz, 2H), 3.47 (s, 3H), 3.43 (t, J = 7.2 Hz, 2H), 1.66 (s, 6H).




A137
NA
497.1
518.9





(M + Na+)


A138
NA
586.1
586.1


A140
NA
530.2
530.2


A141

1H-NMR (400 MHz, CDCl3) δ = 7.42 (d, J = 2.0 Hz, 1H), 7.30 (d, J = 2.4 Hz, 1H), 7.08 (d,

501.2
523.0



J = 8.8 Hz, 2H), 6.74-6.67 (m, 2H), 4.60-4.57 (m, 1H), 4.49-4.45 (m, 1H), 3.97 (d, J = 11.2

(M + Na+)



Hz, 4H), 2.86 (s, 3H), 2.79-2.71 (m, 2H), 2.44-2.34 (m, 2H), 1.64 (s, 6H), 1.05-0.98 (m, 2H),





0.72-0.69 (m, 2H).




A142

1H-NMR (400 MHz, CDCl3) δ = 7.37 (d, J = 2.4 Hz, 1H), 7.29 (d, J = 2.4 Hz, 1H), 7.09 (d,

530.2
530.2



J = 8.8 Hz, 2H), 6.81 (d, J = 8.8 Hz, 2H), 5.09 (s, 1H), 4.07 (s, 2H), 3.85 (t, J = 4.4 Hz, 4H),





3.33 (t, J = 4.4 Hz, 4H), 3.00 (s, 3H), 2.12 (s, 6H), 1.62 (s, 6H).




A143

1H-NMR (400 MHz, CDCl3) δ = 7.41 (d, J = 2.4 Hz, 1H), 7.29 (d, J = 2.4 Hz, 1H), 7.09 (d, J =

501.2
523.1



8.8 Hz, 2H), 6.82 (d, J = 8.8 Hz, 2H), 4.95 (s, 1H), 4.49-4.45 (m, 1H), 4.08 (s, 2H), 3.01 (s,

(M + Na+)



3H), 2.12 (s, 6H), 1.64 (s, 6H), 1.05-0.98 (m, 2H), 0.74-0.67 (m, 2H).




A145

1H-NMR (400 MHz, CDCl3) δ = 8.44 (br s, 1H), 8.16 (d, J = 1.2 Hz, 1H), 7.91 (br d, J = 8.4 Hz,

545.1
545.0



1H), 7.69 (d, J = 8.8 Hz, 2H), 7.54 (s, 1H), 7.22 (d, J = 8.4 Hz, 2H), 7.18 (d, J = 2.8 Hz, 1H),





7.02 (d, J = 3.2 Hz, 1H), 4.37 (t, J = 6.0 Hz, 2H), 3.89 (t, J = 6.0 Hz, 2H), 3.27 (br s, 3H)




A146
NA
541.2
541.1


A147
NA
527.1
527.0


A148

1H-NMR (400 MHz, CDCl3) δ = 8.64 (d, J = 5.2 Hz, 1H), 7.38 (d, J = 2.4 Hz, 1H), 7.31 (d, J =

568.2
568.1



2.0 Hz, 1H), 7.30 (br s, 1H), 7.13 (d, J = 8.8 Hz, 2H), 6.89 (d, J = 8.8 Hz, 2H), 5.11 (s, 2H),





3.93 (t, J = 4.4 Hz, 2H), 3.47 (s, 3H), 3.43-3.38 (m, 2H), 3.28 (s, 2H), 1.64 (s, 6H), 0.91-0.85





(m, 2H), 0.72-0.63 (m, 2H).




A149
NA
554.2
554.0


A150

1H-NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 2.0 Hz, 1H), 7.06 (d, J =

537.1
536.9



8.4 Hz, 2H), 6.71 (d, J = 8.8 Hz, 2H), 4.57 (q, J = 6.8 Hz, 1H), 4.46-4.35 (m, 3H), 3.96-3.84 (m,





3H), 2.93 (s, 3H), 2.68-2.44 (m, 4H), 2.27-2.20 (m, 2H), 2.05 (br t, J = 9.6 Hz, 2H), 1.63 (s, 6H).




A151

1H-NMR (400 MHz, CDCl3) δ = 8.52 (s, 2H), 7.49 (br s, 1H), 7.42 (br d, J = 8.0 Hz, 2H), 7.36

588.2
588.0



(br s, 1H), 7.31-7.28 (m, 1H), 7.26-7.23 (m, 1H), 5.37 (br d, J = 6.8 Hz, 1H), 4.44 (br t, J = 6.0





Hz, 2H), 4.10-3.97 (m, 1H), 3.88 (br t, J = 6.0 Hz, 2H), 3.78 (br d, J = 12.0 Hz, 2H), 2.97 (br t,





J = 10.8 Hz, 2H), 2.82 (s, 3H), 2.22 (br d, J = 10.8 Hz, 2H), 1.70 (br s, 8H).




A152

1H-NMR (400 MHz, CDCl3) δ = 8.95 (s, 1H), 8.64 (d, J = 5.2 Hz, 1H), 7.40 (d, J = 2.0 Hz, 1H),

576.2
576.1



7.32-7.28 (m, 2H), 7.13 (d, J = 8.8 Hz, 2H), 6.90 (d, J = 8.8 Hz, 2H), 5.11 (s, 2H), 3.56-3.49 (m,





2H), 3.47 (s, 3H), 3.29-3.24 (m, 2H), 2.12-1.95 (m, 4H), 1.64 (s, 6H).




A153
NA
558.2
558.2


A154

1H-NMR (400 MHz, CDCl3) δ = 8.46 (s, 1H), 8.16 (s, 1H), 7.90 (br d, J = 7.6 Hz, 1H), 7.71 (d,

491.1
491.0



J = 8.4 Hz, 2H), 7.54 (s, 1H), 7.17 (d, J = 8.8 Hz, 2H), 7.10 (dd, J = 2.8, 12.0 Hz, 1H), 7.01 (dd,





J = 1.6, 2.8 Hz, 1H), 4.38 (s, 1H), 3.28 (br s, 3H), 1.03-0.96 (m, 2H), 0.76-0.68 (m, 2H).




A155

1H-NMR (400 MHz, CDCl3) δ = 8.44 (br s, 1H), 8.15 (s, 1H), 7.89 (br d, J = 8.0 Hz, 1H), 7.71

513.1
513.0



(br d, J = 8.8 Hz, 2H), 7.62-7.47 (m, 1H), 7.15 (d, J = 8.8 Hz, 2H), 7.09 (dd, J = 2.8, 12.0 Hz,





1H), 7.02 (br s, 1H), 4.48 (t, J = 5.6 Hz, 2H), 3.86 (t, J = 6.0 Hz, 2H), 3.28 (br s, 3H).




A157

1H-NMR (400 MHz, DMSO-d6) δ = 11.15 (s, 1H), 8.23 (d, J = 10.8 Hz, 1H), 8.18 (s, 1H), 8.02

546.0 5
545.8



(d, J = 8.8 Hz, 1H), 7.96 (d, J = 8.8 Hz, 1H), 7.85 (d, J = 8.8 Hz, 2H), 7.66 (d, J = 2.8 Hz, 1H),





7.62 (d, J = 2.8 Hz, 1H), 7.23 (d, J = 8.8 Hz, 2H), 4.42 (t, J = 5.2 Hz, 2H), 3.98 (t, J = 5.2 Hz,





2H), 3.52 (s, 3H).




A158

1H-NMR (400 MHz, CDCl3) δ = 9.06 (s, 1H), 8.64 (d, J = 5.2 Hz, 1H), 7.29 (d, J = 5.2 Hz, 1H),

565.1
565.0



7.18-7.12 (m, 4H), 6.89 (d, J = 8.8 Hz, 2H), 5.11 (s, 2H), 3.69 (d, J = 10.4 Hz, 6H), 3.47 (s, 3H),





2.58 (s, 4H), 1.64 (s, 6H).




A160

1H-NMR (400 MHz, CDCl3) δ = 9.05 (s, 1H), 8.64 (d, J = 5.2 Hz, 1H), 7.29 (d, J = 5.2 Hz, 1H),

599.1
599.1



7.18-7.13 (m, 4H), 6.89 (d, J = 8.8 Hz, 2H), 5.11 (s, 2H), 3.78 (s, 2H), 3.47 (s, 3H), 2.79 (t, J =





11.2 Hz, 2H), 2.56 (t, J = 4.4 Hz, 2H), 1.93-1.82 (m, 2H), 1.77-1.70 (m, 2H), 1.64 (s, 6H).




A161
NA
565.1
565.0


A162

1H-NMR (400 MHz, CDCl3) δ = 8.64 (d, J = 5.2 Hz, 1H), 8.25 (s, 1H), 7.30 (d, J = 4.8 Hz, 1H),

522.1
522.0



7.11 (d, J = 8.8 Hz, 2H), 7.01 (d, J = 6.4 Hz, 2H), 6.90 (d, J = 2.8 Hz, 1H), 6.72 (d, J = 3.2 Hz,





1H), 5.13 (s, 2H), 4.31 (t, J = 6.4 Hz, 2H), 3.86 (t, J = 6.4 Hz, 2H), 3.49 (s, 3H), 3.24 (s, 3H)




A163
NA
520.1
519.8


A165
NA
610.1
610.1


A166
NA
571.1
571.0


A167

1H-NMR (400 MHz, CDCl3) δ = 8.63 (d, J = 5.2 Hz, 1H), 7.29 (d, J = 5.2 Hz, 1H), 7.17-7.14 (m,

542.2
542.0



2H), 7.12 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 8.8 Hz, 2H), 5.10 (s, 2H), 4.77-4.72 (m, 2H), 4.35 (dd,





J = 4.4, 9.6 Hz, 2H), 4.22-4.15 (m, 1H), 3.47 (s, 3H), 3.33 (s, 3H), 1.59 (s, 6H).




A168
NA
576.2
576.1


A169

1H-NMR (400 MHz, CDCl3) δ = 8.61 (d, J = 5.2 Hz, 1H), 7.41 (s, 1H), 7.34 (d, J = 1.2 Hz, 1H),

556.2
556.0



7.26 (br s, 1H), 7.12 (d, J = 8.8 Hz, 2H), 6.89 (d, J = 8.4 Hz, 2H), 5.10 (s, 2H), 4.50-4.46 (m, 1H),





3.78 (br t, J = 6.8 Hz, 2H), 3.46 (s, 3H), 3.07 (br t, J = 6.4 Hz, 2H), 3.00 (br t, J = 7.2 Hz, 2H),





2.77 (br t, J = 8.4 Hz, 2H), 1.64 (s, 6H).




A170

1H-NMR (400 MHz, CDCl3) δ = 8.63 (d, J = 5.2 Hz, 1H), 7.63 (d, J = 2.4 Hz, 1H), 7.44 (d, J =

502.1
501.9



2.4 Hz, 1H), 7.28 (s, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.92 (d, J = 8.8 Hz, 2H), 5.12 (s, 2H), 4.28 (t,





J = 4.8 Hz, 2H), 3.92 (t, J = 5.6 Hz, 2H), 3.48 (s, 3H), 1.61 (s, 6H)




A171
NA
489.1
489.0


A177

1H-NMR (400 MHz, CDCl3) δ = 8.46 (s, 1H), 8.18 (s, 1H), 7.92 (d, J = 8.8 Hz, 1H), 7.69 (d,

546.1
546.0



J = 8.8 Hz, 2H), 7.56 (s, 1H), 7.32 (d, J = 3.2 Hz, 1H), 7.22 (d, J = 8.4 Hz, 2H), 7.17 (d, J = 2.8





Hz, 1H), 4.44-4.39 (m, 1H), 3.28 (s, 3H), 2.82-2.77 (m, 1H), 1.07-1.03 (m, 2H), 1.01-0.95 (m,





2H), 0.72-0.66 (m, 4H).




A178

1H-NMR (400 MHz, CDCl3) δ = 8.65 (br d, J = 4.8 Hz, 1H), 7.42 (d, J = 2.4 Hz, 1H), 7.31-7.28

515.1
515.0



(m, 2H), 7.13 (d, J = 8.8 Hz, 2H), 6.90 (d, J = 8.8 Hz, 2H), 5.11 (s, 2H), 4.15 (t, J = 6.4 Hz, 2H),





3.47 (s, 3H), 1.94-1.84 (m, 2H), 1.64 (s, 6H), 1.09 (t, J = 7.6 Hz, 3H).




A179

1H-NMR (400 MHz, CDCl3) δ = 9.95 (s, 1H), 8.68 (d, J = 5.2 Hz, 1H), 7.45 (d, J = 2.0 Hz, 1H),

483.1
483.1



7.42 (d, J = 2.0 Hz, 1H), 7.30 (d, J = 5.6 Hz, 1H), 7.14 (d, J = 9.2 Hz, 2H), 6.92-6.87 (m, 3H),





6.04 (d, J = 17.6 Hz, 1H), 5.78 (d, J = 11.6 Hz, 1H), 5.13 (s, 2H), 3.47 (s, 3H), 1.66 (s, 6H)




A180
NA
612.2
612.0


A181
NA
565.1
565.0


A182

1H-NMR (400 MHz, CDCl3) δ = 8.94 (br s, 1H), 8.64 (d, J = 5.2 Hz, 1H), 7.59-7.53 (m, 2H),

563.1
563.0



7.46 (d, J = 2.0 Hz, 1H), 7.44-7.36 (m, 3H), 7.32 (d, J = 2.4 Hz, 1H), 7.29 (d, J = 5.2 Hz, 1H),





7.15-7.10 (m, 2H), 6.93-6.87 (m, 2H), 5.20 (s, 2H), 5.11 (s, 2H), 3.47 (s, 3H), 1.65 (s, 6H).




A183
NA
594.1
594.0


A184
NA
585.2
585.0


A185

1H-NMR (400 MHz, CDCl3) δ = 8.61 (d, J = 4.8 Hz, 1H), 8.21 (s, 1H), 7.42 (s, 1H), 7.28 (s, 1H),

507.1
506.9



7.10 (br d, J = 8.4 Hz, 2H), 6.93 (d, J = 8.8 Hz, 2H), 5.10 (s, 2H), 4.42 (t, J = 6.0 Hz, 2H), 3.90-





3.86 (m, 4H), 3.48 (s, 3H).




A187

1H-NMR (400 MHz, CDCl3) δ = 8.44 (br s, 1H), 8.16 (d, J = 1.2 Hz, 1H), 7.94-7.87 (m, 1H), 7.67

523.1
523.0



(d, J = 8.4 Hz, 2H), 7.54 (br d, J = 5.2 Hz, 1H), 7.24-7.16 (m, 3H), 7.06 (d, J = 2.8 Hz, 1H), 4.45-





4.37 (m, 1H), 3.27 (s, 3H), 1.11-1.00 (m, 2H), 0.74-0.59 (m, 2H).




A188

1H-NMR (400 MHz, CDCl3) δ = 8.54-8.30 (m, 1H), 8.15 (br s, 1H), 7.92-7.79 (m, 1H), 7.70 (br

509.1
509.0



d, J = 8.0 Hz, 2H), 7.62-7.42 (m, 1H), 7.33 (d, J = 2.8 Hz, 1H), 7.17-7.12 (m, 3H), 4.15 (t, J = 6.4





Hz, 2H), 3.26 (br s, 3H), 1.97-1.87 (m, 2H), 1.12 (t, J = 7.2 Hz, 3H).




A189
NA
563.1
562.9


A190

1H-NMR (400 MHz, CDCl3) δ = 8.44 (s, 1H), 8.15 (d, J = 1.2 Hz, 1H), 7.88 (d, J = 1.6 Hz, 1H),

541.1
541.0



7.71 (d, J = 8.8 Hz, 2H), 7.52 (d, J = 2.8 Hz, 1H), 7.39 (d, J = 3.2 Hz, 1H), 7.15 (d, J = 8.4 Hz,





2H), 4.65-4.62 (m, 1H), 3.27 (s, 3H), 1.03-0.94 (m, 2H), 0.88-0.81 (m, 2H).




A191

1H-NMR (400 MHz, DMSO-d6) δ = 11.27 (br s, 1H), 8.22 (d, J = 10.8 Hz, 1H), 8.17 (s, 1H),

524.1
524.0



8.04-7.98 (m, 1H), 7.97-7.92 (m, 1H), 7.84 (d, J = 8.4 Hz, 2H), 7.65 (d, J = 2.8 Hz, 1H), 7.60 (d,





J = 2.8 Hz, 1H), 7.24 (d, J = 8.8 Hz, 2H), 4.45-4.37 (m, 1H), 3.48 (br s, 3H), 0.94-0.86 (m, 2H),





0.74-0.67 (m, 2H).




A192
NA
507.1
507.0


A193

1H-NMR (400 MHz, MeOD) δ = 8.22 (s, 1H), 8.01 (d, J = 2.0 Hz, 1H), 7.88-7.82 (m, 3H), 7.71

507.1
507.1



(dd, J = 2.0, 8.4 Hz, 1H), 7.45 (d, J = 2.8 Hz, 1H), 7.33 (d, J = 2.8 Hz, 1H), 7.22-7.17 (m, 2H),





4.50-4.44 (m, 1H), 3.27 (s, 3H), 1.02-0.96 (m, 2H), 0.73-0.67 (m, 2H)




A194

1H-NMR (400 MHz, CDCl3) δ = 8.41 (s, 1H), 8.14 (s, 1H), 7.89 (dd, J = 1.6, 8.8 Hz, 1H), 7.78

429.1
429.0



(d, J = 8.4 Hz, 1H), 7.73 (d, J = 8.4 Hz, 2H), 7.33 (d, J = 2.8 Hz, 1H), 7.16-7.13 (m, 3H), 5.74 (s,





2H), 4.45-4.49 (m, 1H), 1.06-1.03 (m, 2H), 0.74-0.69 (m, 2H)




A195
NA
521.1
521.0


A196

1H-NMR (400 MHz, CDCl3) δ = 9.97 (s, 1H), 8.86 (s, 2H), 7.57 (br d, J = 8.8 Hz, 2H), 7.33 (d,

457.1
457.0



J = 2.8 Hz, 1H), 7.16 (br d, J = 8.0 Hz, 3H), 4.51-4.45 (m, 1H), 3.53 (s, 3H), 1.05-1.02 (m, 2H),





0.75-0.70 (m, 2H).




A197
NA
483.1
483.2


A198

1H-NMR (400 MHz, CDCl3) δ = 8.44 (br s, 1H), 8.15 (s, 1H), 7.89 (br d, J = 8.8 Hz, 1H), 7.72

498.1
498.1



(br d, J = 8.8 Hz, 2H), 7.63-7.48 (m, 1H), 7.45 (s, 2H), 7.15 (d, J = 8.8 Hz, 2H), 4.62-4.56 (m,





1H), 3.27 (br s, 3H), 1.11-1.04 (m, 2H), 0.94-0.87 (m, 2H)




A199

1H-NMR (400 MHz, CDCl3) δ = 8.45 (br s, 1H), 8.16 (d, J = 1.2 Hz, 1H), 7.90 (d, J = 8.8 Hz,

572.1
572.2



1H), 7.71 (d, J = 8.8 Hz, 2H), 7.57 (br s, 1H), 7.21 (d, J = 8.8 Hz, 2H), 6.97 (dd, J = 2.8, 12.8 Hz,





1H), 6.92-6.87 (m, 1H), 4.36-4.27 (m, 3H), 3.27 (br s, 3H), 1.01-0.95 (m, 2H), 0.73-0.67 (m, 2H).




A200

1H-NMR (400 MHz, CDCl3) δ = 8.26 (s, 1H), 8.09 (d, J = 2.0 Hz, 1H), 7.86-7.83 (m, 1H), 7.81-

443.1
443.0



7.78 (m, 1H), 7.76-7.71 (m, 2H), 7.33 (d, J = 3.2 Hz, 1H), 7.15 (d, J = 2.8 Hz, 1H), 7.14-7.11 (m,





2H), 5.09 (br s, 1H), 4.49-4.44 (m, 1H), 3.18 (d, J = 5.2 Hz, 3H), 1.07-1.05 (m, 2H), 0.72-





0.70 (m, 2H).




A202

1H-NMR (400 MHz, CDCl3) δ = 8.67 (d, J = 5.2 Hz, 1H), 7.31 (d, J = 5.2 Hz, 1H), 7.15 (d, J = 8.8

568.1
568.2



Hz, 2H), 7.05 (d, J = 8.8 Hz, 2H), 6.94 (d, J = 3.2 Hz, 1H), 6.72 (d, J = 3.2 Hz, 1H), 5.15 (s, 2H),





4.40-4.34 (m, 1H), 4.19 (q, J = 8.4 Hz, 2H), 3.49 (s, 3H), 1.04-0.99 (m, 2H), 0.69-0.62 (m, 2H)




A204
NA
499.1
499.2


A205
NA
485.1
485.0


A206

1H-NMR (400 MHz, CDCl3) δ = 8.71 (s, 1H), 8.16 (s, 1H), 7.92 (d, J = 8.4 Hz, 1H), 7.82 (d,

547.1
547.0



J = 8.0 Hz, 1H), 7.75 (d, J = 8.0 Hz, 2H), 7.34 (s, 1H), 7.19-7.12 (m, 3H), 4.47 (s, 1H), 4.39 (s,





4H), 3.20 (s, 4H), 1.07-1.05 (m, 2H), 0.73-0.71 (m, 2H).




A208

1H-NMR (400 MHz, CDCl3) δ = 8.44 (s, 1H), 8.15 (s, 1H), 7.90 (d, J = 8.4 Hz, 1H), 7.67 (d,

504.1
504.0



J = 8.8 Hz, 2H), 7.53 (s, 1H), 7.21 (d, J = 8.8 Hz, 2H), 6.96 (dd, J = 2.8, 13.2 Hz, 1H), 6.90 (s,





1H), 4.32-4.29 (m, 1H), 3.36 (s, 3H), 3.27 (s, 3H), 1.02-0.95 (m, 2H), 0.70-0.65 (m, 2H).




A209
NA
509.1
509.2


A210

1H-NMR (400 MHz, CDCl3) δ = 8.05 (br s, 1H), 7.72-7.63 (m, 3H), 7.44 (d, J = 8.8 Hz, 1H),

509.1
509.1



7.31 (d, J = 3.2 Hz, 1H), 7.13 (d, J = 2.8 Hz, 1H), 7.13-7.05 (m, 2H), 6.96 (br s, 1H), 4.49-4.44





(m, 1H), 4.08 (s, 3H), 3.20 (s, 3H), 1.11-1.00 (m, 2H), 0.76-0.64 (m, 2H).




A211

1H-NMR (400 MHz, CDCl3) δ = 9.23 (d, J = 2.0 Hz, 1H), 8.05 (dd, J = 2.0, 9.2 Hz, 1H), 7.68 (d,

523.1
523.0



J = 8.4 Hz, 2H), 7.61 (d, J = 9.2 Hz, 1H), 7.34 (d, J = 2.8 Hz, 1H), 7.18-7.15 (m, 3H), 6.27 (s,





1H), 4.51-4.46 (m, 1H), 3.37 (s, 3H), 1.08-1.04 (m, 2H), 0.76-0.71 (m, 2H).




A212
NA
470.1
470.0


A213

1H-NMR (400 MHz, CDCl3) δ = 8.97 (s, 1H), 8.67 (s, 1H), 8.07 (d, J = 8.8 Hz, 2H), 7.33 (d, J =

471.1
471.1



2.8 Hz, 1H), 7.17-7.13 (m, 3H), 5.38 (br s, 1H), 4.56 (d, J = 5.6 Hz, 2H), 4.51-4.45 (m, 1H),





3.02 (s, 3H), 1.09-1.02 (m, 2H), 0.75-0.70 (m, 2H).




A214
NA
471.1
471.1


A215

1H-NMR (400 MHz, CDCl3) δ = 8.26 (s, 1H), 8.11 (s, 1H), 7.91-7.84 (m, 1H), 7.78 (br d, J = 8.8

561.1
561.0



Hz, 1H), 7.73 (d, J = 8.4 Hz, 2H), 7.33 (d, J = 2.8 Hz, 1H), 7.17-7.11 (m, 3H), 5.03-4.80 (m, 1H),





4.53-4.37 (m, 2H), 3.33-3.14 (m, 4H), 2.64-2.53 (m, 2H), 2.38-2.22 (m, 2H), 1.11-1.01 (m, 2H),





0.77-0.67 (m, 2H)




A216
NA
533.1
533.0


A217
NA
474.1
474.0


A218

1H-NMR (400 MHz, CDCl3) δ = 8.43 (br s, 1H), 8.12 (d, J = 2.0 Hz, 1H), 8.00 (d, J = 2.4 Hz,

483.1
483.1



1H), 7.87 (dd, J = 1.6, 8.8 Hz, 1H), 7.65 (d, J = 8.4 Hz, 2H), 7.59-7.49 (m, 1H), 7.46 (d, J = 2.8





Hz, 1H), 7.15-7.07 (m, 2H), 4.42-4.27 (m, 1H), 3.27 (s, 3H), 0.88-0.83 (m, 4H).




A219
NA
508.1
508.0


A221

1H-NMR (400 MHz, CDCl3) δ = 8.79 (d, J = 5.2 Hz, 1H), 7.55 (d, J = 5.6 Hz, 1H), 7.18-7.14 (m,

582.1
582.2



2H), 7.08-7.04 (m, 2H), 6.95 (d, J = 2.8 Hz, 1H), 6.73 (d, J = 3.2 Hz, 1H), 5.61 (br s, 1H), 5.19 (s,





2H), 4.62 (d, J = 5.2 Hz, 2H), 4.40-4.33 (m, 1H), 4.23-4.16 (m, 2H), 3.02 (s, 3H), 1.03-0.99 (m,





2H), 0.68-0.63 (m, 2H).




A222

1H-NMR (400 MHz, CDCl3) δ = 8.36 (br s, 1H), 8.08 (s, 1H), 7.82 (br d, J = 8.8 Hz, 1H), 7.62 (d,

570.1
570.0



J = 8.4 Hz, 2H), 7.47 (br s, 1H), 7.17 (d, J = 2.8 Hz, 1H), 7.12 (d, J = 8.4 Hz, 2H), 7.00 (d, J = 2.8





Hz, 1H), 6.09-5.79 (m, 1H), 4.38-4.35 (m, 1H), 4.05-3.99 (m, 2H), 3.27 (br s, 3H), 0.99-0.95 (m,





2H), 0.65-0.61 (m, 2H).




A223

1H-NMR (400 MHz, CDCl3) δ = 8.91 (br s, 1H), 8.68 (d, J = 5.2 Hz, 1H), 7.31 (d, J = 5.2 Hz,

597.1
597.0



1H), 7.15 (d, J = 8.8 Hz, 2H), 7.05 (d, J = 9.2 Hz, 2H), 6.89 (d, J = 3.2 Hz, 1H), 6.73 (d, J = 3.2





Hz, 1H), 5.16 (s, 2H), 4.25-4.14 (m, 2H), 3.84 (t, J = 4.8 Hz, 4H), 3.48 (s, 3H), 3.24 (s, 4H).




A225

1H-NMR (400 MHz, CDCl3) δ = 8.68 (s, 1H), 8.06 (s, 1H), 7.67 (s, 1H), 7.46 (d, J = 2.8 Hz, 1H),

510.1
510.1



7.29 (br s, 2H), 6.97 (d, J = 8.8 Hz, 2H), 6.86-6.80 (m, 2H), 5.66 (s, 2H), 4.46-4.35 (m, 1H), 3.04





(s, 3H), 1.00-0.94 (m, 2H), 0.91-0.85 (m, 2H).




A226
NA
506.1
506.1


A227

1H-NMR (400 MHz, CDCl3) δ = 9.09 (s, 1H), 7.94 (s, 1H), 7.56 (s, 1H), 7.49 (d, J = 2.8 Hz,

510.1
510.1



1H), 7.36 (d, J = 8.8 Hz, 2H), 7.02 (d, J = 8.4 Hz, 2H), 6.88 (d, J = 2.8 Hz, 1H), 6.85 (s, 1H),





5.64 (s, 2H), 4.45-4.37 (m, 1H), 3.05 (s, 3H), 1.00-0.95 (m, 2H), 0.92-0.87 (m, 2H).




A229

1H-NMR (400 MHz, CDCl3) δ = 7.95 (s, 1H), 7.77 (s, 1H), 7.22 (d, J = 2.8 Hz, 1H), 7.00 (s,

541.1
541.5



5H), 6.82 (s, 1H), 4.45-4.40 (m, 1H), 3.8 (t, J = 4.8 Hz 4H), 3.34-3.27 (m, 7H), 1.07-1.00 (m,





2H), 0.72-0.64 (m, 2H).




A230
NA
541.1
541.1


A231

1H-NMR (400 MHz, CDCl3) δ = 9.05 (s, 1H), 7.23 (d, J = 2.8 Hz, 1H), 7.01 (s, 5H), 6.15 (s,

500.1
500.1



1H), 4.48-4.41 (m, 1H), 4.40 (s, 2H), 4.30 (t, J = 5.6 Hz, 2H), 3.74 (t, J = 5.2 Hz, 2H), 3.05 (s,





3H), 1.06-0.99 (m, 2H), 0.71-0.66 (m, 2H)




A232
NA
512.1
512.2


A233

1H-NMR (400 MHz, CDCl3) δ = 8.43 (s, 1H), 8.29 (d, J = 2.8 Hz, 1H), 8.11 (s, 1H), 7.86 (br d,

584.2
584.1



J = 7.6 Hz, 1H), 7.64 (d, J = 2.8 Hz, 1H), 7.61 (d, J = 8.8 Hz, 2H), 7.51 (s, 1H), 6.96 (d, J = 8.8





Hz, 2H), 4.29 (q, J = 8.4 Hz, 2H), 3.90-3.85 (m, 4H), 3.75-3.71 (m, 4H), 3.26 (br s, 3H)




A234

1H-NMR (400 MHz, CDCl3) δ = 7.99 (d, J = 3.2 Hz, 1H), 7.24 (d, J = 2.8 Hz, 1H), 7.05 (d, J =

523.2
523.1



8.8 Hz, 2H), 6.92 (d, J = 8.8 Hz, 2H), 4.99 (s, 1H), 4.34-4.26 (m, 1H), 4.19 (q, J = 8.4 Hz, 2H),





4.10 (s, 2H), 3.02 (s, 3H), 2.14 (s, 6H), 0.86-0.78 (m, 4H)




A235
NA
522.2
522.1


A237
NA
500.1
500.0


A238

1H-NMR (400 MHz, CDCl3) δ = 9.28 (br s, 1H), 8.74-8.66 (m, 1H), 7.32 (br d, J = 4.8 Hz, 1H),

590.1
590.0



7.16 (d, J = 8.0 Hz, 2H), 7.06 (br d, J = 8.4 Hz, 2H), 6.93 (br s, 1H), 6.72 (br s, 1H), 5.16 (s,





2H), 4.33 (t, J = 6.0 Hz, 2H), 4.19 (q, J = 8.8 Hz, 2H), 3.86 (t, J = 6.4 Hz, 2H), 3.49 (s, 3H).




A239
NA
569.2
569.1


A240
NA
498.2
498.0


A241

1H-NMR (400 MHz, CDCl3) δ = 9.08 (s, 1H), 7.93 (dd, J = 2.4, 8.8 Hz, 1H), 7.85 (d, J = 1.6 Hz,

498.2
498.1



1H), 7.71-7.64 (m, 3H), 7.32 (d, J = 2.8 Hz, 1H), 7.14-7.11 (m, 3H), 4.49-4.44 (m, 1H), 4.04 (t,





J = 5.2 Hz, 4H), 3.06 (t, J = 4.8 Hz, 4H), 1.09-1.02 (m, 2H), 0.75-0.68 (m, 2H)




A242

1H-NMR (400 MHz, CDCl3) δ = 8.63 (s, 1H), 8.12 (d, J = 2.0 Hz, 1H), 7.87 (dd, J = 2.0, 8.4 Hz,

540.2
540.2



1H), 7.78 (d, J = 8.8 Hz, 1H), 7.74 (d, J = 8.8 Hz, 2H), 7.33 (d, J = 3.2 Hz, 1H), 7.16-7.12 (m, 3H),





4.48-4.44 (m, 1H), 3.92-3.87 (m, 2H), 3.83 (br s, 4H), 3.71-3.65 (m, 2H), 2.19 (s, 3H), 1.09-1.01





(m, 2H), 0.75-0.68 (m, 2H).




A243
NA
504.2
504.2


A244

1H-NMR (400 MHz, CDCl3) δ = 8.34 (d, J = 4.8 Hz, 1H), 7.42 (d, J = 2.0 Hz, 1H), 7.31 (d, J =

546.2
546.1



2.0 Hz, 1H), 7.11 (d, J = 8.4 Hz, 2H), 6.89 (d, J = 8.4 Hz, 2H), 6.79 (d, J = 5.2 Hz, 1H), 4.97 (s,





2H), 4.49-4.45 (m, 1H), 3.91-3.83 (m, 4H), 3.75-3.67 (m, 2H), 3.59-3.51 (m, 2H), 2.17 (s, 3H),





1.64 (s, 6H), 1.05-0.98 (m, 2H), 0.76-0.67 (m, 2H).




A245

1H-NMR (400 MHz, CDCl3) δ = 8.38 (d, J = 5.2 Hz, 1H), 7.15-7.11 (m, 2H), 7.05-7.01 (m, 2H),

601.2
601.2



6.93 (d, J = 2.8 Hz, 1H), 6.81 (d, J = 4.8 Hz, 1H), 6.72 (d, J = 3.2 Hz, 1H), 5.02 (s, 2H), 4.39-4.32





(m, 1H), 4.18 (q, J = 8.8 Hz, 2H), 3.92-3.85 (m, 4H), 3.73-3.68 (m, 2H), 3.58-3.52 (m, 2H), 2.17





(s, 3H), 1.03-0.97 (m, 2H), 0.67-0.62 (m, 2H).











Biological Assays


Example 19: Activity of Exemplary Compounds in Cellular Assays

The PSA (6.1 kb)-luciferase reporter contains functional AREs (androgen response elements) to which AR binds in response to androgen to induce luciferase activity. LNCaP cells were transiently transfected with the PSA (6.1 kb)-luciferase reporter for 24 h, and then pretreated with vehicle (DMSO) or indicated concentration of representative compounds for one hour prior to the addition of synthetic androgen, R1881 (1 nM). After 24 h of incubation with R1881, the cells were harvested, and relative luciferase activities were determined (FIG. 1). To determine the IC50, treatments were normalized to vehicle control activity induced by R1881 (Table 2).


Luciferase Assay A: Cells were lysed using Passive Lysis Buffer (Promega) and then collected into V-bottom 96-well tissue culture plates. Lysates were centrifuged at 4° C. for 5 minutes at 3000 rpm. To measure luminescence of LNCaP cell lysates the Firefly Luciferase Assay System (Promega) was employed, according to manufacturer's protocol. Relative luminescence units (RLU) in cell lysates were detected for 10 seconds using Promega GloMax-Multi Detection Luminometer (Promega). Values were normalized to protein content. GraphPad Prism graphing software was used to calculate IC50 values. Luciferase Assay A was run for all compounds in Table 2 unless otherwise noted below.


Luciferase Assay B: Cells stably expressing probasin promoter (ARR2PB) or human prostate-specific antigen (PSA)-firefly luciferase reporter were plated in poly-D-lysine-coated 384-well plates. 48 h after plating, cells were pretreated with vehicle or compounds for 1 h before adding R1881 under serum-free and phenol red-free conditions and then further incubated for 24 h. R1881 was added only for LNCaP cells. Firefly luciferase activities were determined by EnVision plate reader (Perkinemer) using the Steady-Glo Luciferase Reporter Assay System (Promega). Values were normalized to protein content. GraphPad Prism graphing software was used to calculate IC50 values. Luciferase Assay B was run for Compounds A186-A219 and A221-A245 in Table 2.


Statistical analyses were performed using GraphPad Prism (Version 6.01 for Windows; La Jolla, CA, USA). Comparisons between treatment and control groups were compared using Two-Way ANOVA with post-hoc Dunnett's and Tukey's tests. Differences were considered statistically significant at P values less than 0.05.


Table 2 shows IC50 ranges of Compounds from Table A.









TABLE 2







IC50 Ranges of Various Compounds in Androgen-Induced


PSA-Luciferase Assay










Compound ID
PSA-Luc IC50 Range







A1  
**



A2  
**



A3  
**



A4  
**



A5  
**



A6  
***



A7  
*



A8  
*



A9  
*



A10 
**



A11 
**



A12 
**



A13 
*



A14 
**



A15 
**



A16 
**



A17 
**



A18 




A19 




A20 
*



A21 
**



A22 
**



A23 
*



A24 
**



A25 
*



A26 
*



A27 
*



A28 
**



A29 
*



A30 
*



A31 
*



A32 
***



A33 
*



A34 
*



A35 
*



A36 
***



A37 
***



A38 
**



A39 
**



A40 
*



A41 
*



A42 
**



A43 
***



A44 
***



A45 
**



A46 
*



A47 
*



A48 
*



A49 
**



A50 
***



A51 
*



A52 
**



A53 
***



A54 
***



A55 
***



A56 
*



A57 




A58 
*



A59 
**



A60 
**



A61 
**



A62 
**



A63 
**



A64 
**



A65 
*



A66 
*



A67 
**



A68 
***



A69 
**



A70 
*



A71 
**



A72 
**



A73 




A74 
**



A75 
**



A76 
**



A77 
*



A78 
**



A79 
*



A80 
***



A81 
***



A82 
***



A83 
**



A84 
**



A85 
**



A86 
**



A87 
**



A88 
*



A89 




A90 




A91 




A92 




A93 
*



A94 
**



A95 
**



A96 
***



A97 
**



A98 
**



A99 
**



A100
**



A101
**



A102
*



A103
**



A104
*



A105
**



A106
**



A107
*



A108
**



A109
*



A110
**



A111
*



A112
**



A113
*



A114
*



A115
**



A116
*



A117
*



A118
***



A119
*



A120
*



A121
**



A122
*



A123
*



A124
**



A125
**



A126
*



A127




A128
*



A129
*



A130
**



A131
**



A132
*



A133
*



A134
**



A135
**



A136
**



A137
**



A138
**



A139
**



A140
**



A141
**



A142
**



A143
**



A144




A145
**



A146
**



A147
**



A148
**



A149
**



A150
**



A151
*



A152
**



A153
*



A154
**



A155
**



A156




A157
**



A158




A159
*



A160
*



A161
*



A162
**



A163
***



A164
**



A165
*



A166
**



A167
*



A168
*



A169
*



A170
*



A171
*



A172




A173




A174




A175




A176




A177
**



A178
***



A179
**



A180
*



A181
*



A182
**



A183
*



A184
*



A185
*



A186
***



A187
**



A188
**



A189
**



A190
**



A191
**



A192
***



A193
**



A194
*



A195
**



A196
**



A197
**



A198
***



A199
***



A200
*



A201
**



A202
***



A203
***



A204
*



A205
**



A206
**



A207
**



A208
***



A209
*



A210
**



A211
*



A212
**



A213
**



A214
**



A215
**



A216
*



A217
*



A218
**



A219
**



A220




A221
***



A222
***



A223
***



A224
***



A225
**



A226
**



A227
**



A228
*



A229
*



A230
***



A231
*



A232
**



A233
**



A234
**



A235
*



A236
***



A237
**



A238
***



A239
***



A240
*



A241
*



A242
**



A243
**



A244
***



A245
***







Note:



*** represent IC50 < 500 nM, ** represents IC50 in the range of 500-2000 nM, * represents IC50 in the range of >2000 nM






Example 20: Stability Assays

Microsomal Stability Assay: Microsomal stability assay is a widely used in vitro model to characterize the metabolic conversion by phase I enzymes, such as cytochrome P450 (CYP) enzymes. Since metabolism is known to be highly variable in different species, microsomal stability assay is commonly run in multiple species. Metabolic stability of testing compound can be evaluated using human, rat, mouse, or other animal liver or intestine microsomes to predict intrinsic clearance.


The assay was carried out in 96-well microtiter plates at 37° C. Reaction mixtures (25 μL) contained a final concentration of 1 μM test compound, 0.5 mg/mL liver microsomes protein, and 1 mM NADPH and/or 1 mM UDPGA (with alamethicin) in 100 mM potassium phosphate, pH 7.4 buffer with 3 mM MgCl2. The incubation was done with N=1, but duplicate incubation at each time point can be prepared if necessary. At each of the time points (for example, 0, 15, 30, and 60 minutes), 150 μL of quench solution (100% acetonitrile with 0.1% formic acid) with internal standard was transferred to each well. Besides the zero minute controls, mixtures containing the same components except the NADPH can also be prepared as the negative control. Verapamil was included as a positive control to verify assay performance. Plates were sealed, vortexed, and centrifuged at 4° C. for 15 minutes at 4000 rpm. The supernatant is transferred to fresh plates for LC/MS/MS analysis.


Summarized conditions: [Compound]=1 μM, [LM]=0.5 mg/mL, [NADPH]=1 mM and/or [UDPGA]=1 mM, Buffer=100 mM Potassium Phosphate, pH 7.4 with 3 mM MgCl2, Time=0, 15, 30, and 60 min, and Temperature=37° C.


All samples were analyzed on LC/MS/MS using an AB Sciex API 4000 instrument, coupled to a Shimadzu LC-20AD LC Pump system. Analytical samples were separated using a Waters Atlantis T3 dC18 reverse phase HPLC column (20 mm×2.1 mm) at a flow rate of 0.5 mL/min. The mobile phase consisted of 0.1% formic acid in water (solvent A) and 0.1% formic acid in 100% acetonitrile (solvent B).


The extent of metabolism is calculated as the disappearance of the test compound, compared to the 0-min time incubation. Initial rates are calculated for the compound concentration and used to determine half-life (t1/2).


Liver microsome half life of representative compounds are shown in Tables 2. Compound X has the following structures:




embedded image









TABLE 3







Liver Microsome Half-Life








Compound
Liver microsome + NADPH T½ (min)












ID
Human
Mouse
Rat
Dog
Monkey















A1
>120
>120
61
62
>120


A11
58
>120
103
>120
28


A12
82
>120
72
>120
19


A15
>120
>120
>120
>120
>120


A16
>120
>120
>120
>120
>120


A17
>120
>120
>120
>120
>120


A22
>120
>120
>120
>120
>120


A24
>120
>120
>120
78
>120


A28
78
>120
>120
>120
>120


A36
>120
>120
66
>120
49


A37
>120
>120
>120
>120
32


A43
>120
>120
>120
>120
>120


A44
>120
>120
112
>120
117


A72
>120
>120
>120
>120
>120


A75
>120
>120
>120
>120
>120


A78
>120
>120
>120
>120
>120


A80
>120
96
>120
>120
>120


A81
>120
>120
>120
>120
>120


A82
>120
>120
>120
>120
>120


A83
>120
>120
>120
>120
>120


A84
101
>120
>120
>120
100


A94
>120
>120
>120
>120
117


A97
>120
>120
>120
>120
>120


A98
>120
>120
>120
>120
>120


A145
>120
>120
>120
>120
>120


A157
>120
>120
>120
>120
>120


A163
>120
>120
>120
>120
>120


A164
>120
>120
>120
>120
>120


A177
>120
>120
>120
>120
>120


A186
>120
>120
>120
>120
>120


A187
>120
>120
>120
>120
>120


A190
>120
>120
>120
>120
>120


A191
>120
>120
>120
>120
>120


A198
>120
>120
>120
>120
>120


A199
>120
>120
>120
>120
>120


A202
>120
>120
>120
>120
>120


A203
>120
>120
>120
>120
>120


A205
>120
>120
>120
>120
>120


A208
>120
>120
>120
>120
>120


X
>120
NA
NA
NA
NA


Enzalutamide
>120
>120
>120
>120
>120









The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.


While the invention has been described in connection with proposed specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth and as follows in the scope of the appended claims.

Claims
  • 1. A compound having the structure of formula (I-A):
  • 2. The compound of claim 1 having the structure of formula (A):
  • 3. The compound of claim 2, wherein C is not quinazoline ring.
  • 4. (canceled)
  • 5. The compound of claim 2, wherein A is phenyl, pyridyl, pyrimidinyl, cyclohexyl, piperidinyl.
  • 6. The compound of claim 2, wherein B is phenyl.
  • 7. The compound of claim 2, wherein Y is a bond or -O-.
  • 8. The compound of claim 2, wherein W is a bond, —CH2—, —CH2CH2—, or heteroarylene.
  • 9. The compound of claim 2, wherein W is pyrazole, pyridine, pyrimidine, or pyrazine.
  • 10. The compound of claim 2, wherein C is a 4-10 membered monocyclic carbocyclyl or a bridged 5-10 membered bicyclic carbocyclyl, a 4-10 membered monocyclic heterocyclyl, a 5-10 membered monocyclic heteroaryl, or a fused 8-10 membered bicyclic heteroaryl.
  • 11. (canceled)
  • 12. The compound of claim 2, wherein C is a 6,6-fused heteroaryl or heterocycle, 5,6-fused heteroaryl or heterocycle, 6,5-fused heteroaryl or heterocycle, or 5,5-fused heteroaryl or heterocycle.
  • 13. The compound of claim 2, wherein C is
  • 14. The compound of claim 2, wherein at least one R3 is —CH2NHSO2CH3,-CH(CH3)NHSO2CH3,-CH2CH2NHSO2CH3,-CH2N(CH3)SO2CH3,-NHSO2CH3,-N(CH3)SO2CH3,-NHSO2CH2CH3,-NH2, —NHCH3,-SO2NH2, or -SO2CH3; and the other R3 is, if present, halogen, —CN,-CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, oxo, —S(C1-C3 alkyl),-SO2(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —(C1-C3 alkyl)NH2,-NHSO2CH3,-NHSO2CF3,-N(CH3)SO2CH3,-NHSO2CH2CH3,-N(CH3)SO2CH2CH3,-CH2NHSO2CH3,-CH2N(CH3)SO2CH3,-SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl),-N(CH3)CO(C1-C3 alkyl),-NHCOCF3,-N(CH3)COCF3, —NHCOO(C1-C3 alkyl), or -N(CH3)COO(C1-C3 alkyl).
  • 15. The compound of claim 2, wherein: Z is —O—;V is —CH2—, —CH2CH2—, or —(CR8a′R9a′)-;R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered carbocyclyl; andL is hydrogen or halogen.
  • 16. The compound of claim 2, wherein-Z—V-L is-O-CH3,-O-CH2CH2Cl, —O-cyclopropyl, or —O-cyclobutyl.
  • 17. The compound of claim 2, wherein: Z is a bond;V is—(CR8a′R9a′)-;R8a′ and R9a′ taken together form an optionally substituted 3- to 6-membered heterocyclyl containing one, two, or three heteroatoms selected from N, S, or O; andL is hydrogen.
  • 18. The compound of claim 2, wherein B is phenyl and X and Z are connected at para positions of B.
  • 19. The compound of claim 2, wherein A is a 6-membered monocyclic ring selected from aryl, carbocyclyl, heteroaryl, or heterocyclyl; and X and Y are connected at 1 and 4 positions of A.
  • 20. The compound of claim 1 having the structure of formula (B-I):
  • 21.-30. (canceled)
  • 31. The compound of claim 1 having the structure of formula (C-I):
  • 32.-45. (canceled)
  • 46. The compound of claim 1 having the structure of formula (D-I):
  • 47.-86. (canceled)
  • 87. The compound of claim 1, wherein the compound is
  • 88. The compound of claim 1, wherein the compound is
  • 89. The compound of claim 1, wherein the compound is
  • 90. The compound of claim 1, wherein the compound is
  • 91. The compound of claim 1, wherein the compound is
  • 92.-96. (canceled)
  • 97. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier.
  • 98. A method for treating cancer, comprising administering the compound, pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug of the compound of claim 1, to a subject in need thereof.
  • 99. The method of claim 98, wherein the cancer is prostate cancer.
  • 100. The method of claim 99, wherein the prostate cancer is metastatic castration-resistant prostate cancer.
  • 101. The method of claim 99, wherein the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of and priority to U.S. Provisional Application No. 63/333,808 filed Apr. 22, 2022, and U.S. Provisional Application No. 63/377,777 filed Sep. 30, 2022, each disclosure is hereby incorporated by reference in its entirety.

Provisional Applications (2)
Number Date Country
63377777 Sep 2022 US
63333808 Apr 2022 US