US20200123117A1 - Androgen receptor modulators and methods for their use - Google Patents

Androgen receptor modulators and methods for their use Download PDF

Info

Publication number
US20200123117A1
US20200123117A1 US16/657,625 US201916657625A US2020123117A1 US 20200123117 A1 US20200123117 A1 US 20200123117A1 US 201916657625 A US201916657625 A US 201916657625A US 2020123117 A1 US2020123117 A1 US 2020123117A1
Authority
US
United States
Prior art keywords
alkyl
optionally substituted
halogen
alkenyl
alkynyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/657,625
Inventor
Han-Jie Zhou
Peter VIRSIK
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ESSA Pharma Inc
Original Assignee
ESSA Pharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by ESSA Pharma Inc filed Critical ESSA Pharma Inc
Priority to US16/657,625 priority Critical patent/US20200123117A1/en
Assigned to ESSA PHARMA, INC. reassignment ESSA PHARMA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VIRSIK, PETER, ZHOU, HAN-JIE
Priority to US16/852,810 priority patent/US11059795B2/en
Publication of US20200123117A1 publication Critical patent/US20200123117A1/en
Priority to US17/363,534 priority patent/US20220380325A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/46Sulfur atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/4211,3-Oxazoles, e.g. pemoline, trimethadione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/74Two oxygen atoms, e.g. hydantoin with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to other ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/76Two oxygen atoms, e.g. hydantoin with substituted hydrocarbon radicals attached to the third ring carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/80Two oxygen atoms, e.g. hydantoin with hetero atoms or acyl radicals directly attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/84Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/88Nitrogen atoms, e.g. allantoin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/20Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/08Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/36One oxygen atom
    • C07D263/38One oxygen atom attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/48Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/101,3,4-Oxadiazoles; Hydrogenated 1,3,4-oxadiazoles
    • C07D271/1131,3,4-Oxadiazoles; Hydrogenated 1,3,4-oxadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/42Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/56Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/64Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present disclosure generally relates to tricyclic compounds and their use for treatment of various indications.
  • 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.
  • AR androgen receptor
  • 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)).
  • 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).
  • ABT androgen ablation therapy
  • ADT androgen depravation therapy
  • Androgens also play a role in female diseases such as polycystic ovary syndrome as well as cancers.
  • 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.
  • 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.
  • LBD carboxy-terminal ligand-binding domain
  • DBD DNA-binding domain
  • NTD N-terminus domain
  • AF-1 activation function-1
  • 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).
  • PKA cAMP-dependent protein kinase pathway
  • IL-6 interleukin-6
  • 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).
  • Clinically available inhibitors of the AR include nonsteroidal antiandrogens such as apalutamide, darolutamide, bicalutamide (CasodexTM), nilutamide, flutamide, and enzalutamide.
  • nonsteroidal antiandrogens such as apalutamide, darolutamide, bicalutamide (CasodexTM), nilutamide, flutamide, and enzalutamide.
  • steroidal antiandrogens such as cyproterone acetate and spironolactone. Both steroidal and non-steroidal antiandrogens target the LBD of the AR and predominantly fail presumably due to poor affinity and mutations that lead to activation of the AR by these same antiandrogens (Taplin, M. E., Bubley, G. J., Kom Y. J., Small E.
  • AR degraders such as niclosamide (Liu C et al 2014), galeterone (Njar et al 2015; Yu Z at al 2014), and ARV-330/Androgen receptor PROTAC (Neklesa et al 2016 J Clin Oncol 34 suppl 2S; abstr 267); AR DBD inhibitor VPC-14449 (Dalal K et al 2014 J Biol Chem.
  • 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).
  • AF-1 Activation-Function-1
  • 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.
  • 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).
  • 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; and WO 2018/045450, and which are hereby incorporated by reference in their entireties.
  • AR mechanisms of resistance to ADT include: overexpression of AR (Visakorpi, T. et al Nature Genetics 1995, 9, 401-406; Koivisto, P. et al Scandinavian Journal of Clinical and Laboratory Investigation Supplementum 1996, 226, 57-63); gain-of-function mutations in the AR LBD (Culig Z.
  • AR-Vs AR splice variants
  • LBD ligand-binding domain
  • Anti-androgens such as bicalutamide and enzalutamide target AR LBD, but have no effect on truncated constitutively active AR-Vs such as AR-V7 (Li Y. et al Cancer Research 2013, 73, 483-489). Expression of AR-V7 is associated with resistance to current hormone therapies (Li Y. et al Cancer Research 2013, 73, 483-489; Antonarakis E. S. et al The New England Journal of Medicine 2014, 371, 1028-1038).
  • the compounds of the present disclosure are androgen receptor modulators which may be useful in treating various diseases and conditions as disclosed herein.
  • the present disclosure provides compounds comprising the structure of formula (IIIA):
  • the present disclosure provides compounds comprising the structure of formula (IVA):
  • C is 5- to 10-membered heteroaryl or aryl. In some embodiments, C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • C, which is substituted with (R 3 )n3, is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl. In some embodiments, C, which is substituted with (R 3 )n3, is selected from
  • R 3a is C 1 -C 3 alkyl.
  • R 1 and R 2 are each independently C 1 , —CN, —CF 3 , —OH, methyl, methoxy, or —CONH 2 .
  • the present disclosure provides compounds comprising the structure of formula (A-I):
  • At least one R 3 is selected from —CN, C 1 -C 3 alkoxy, —CONH 2 , —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , or —SO 2 CH 3 and the other R 3 , if present, is selected from —CN, —CF 3 , C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —S(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NH 2 , —(C 1 -C 3 alkyl)NH 2 , —NHSO 2 CH 3 , —NHSO 2 CF 3 , —N(CH 3 )SO 2 CH 3 , —NHSO
  • the present disclosure provides compounds comprising the structure of formula (G-II):
  • At least one R 3 is selected from —NHSO 2 CH 3 , —NHSO 2 CH 2 CH 3 , or —SO 2 CH 3 and the other R 3 , if present, is selected from —CN, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, —SO 2 (C 1 -C 3 alkyl), —NH 2 , —(C 1 -C 3 alkyl)NH 2 , —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -C 3 alkyl), —N(CH 3 )COO(C 1 -C 3
  • the present disclosure provides Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (IIIA), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (IVA), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (A-I), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (G-II), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising any one of Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising any one of Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a method for treating cancer, comprising administering any one of the compound of formula (IIIA), (IVA), (A-I), or (G-II), and Compounds A1-A96, A98-A116, A118-A159, A161-A175, or A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, to a subject in need thereof.
  • the cancer is prostate cancer.
  • the prostate cancer is metastatic castration-resistant prostate cancer.
  • the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.
  • the present disclosure provides a method for treating cancer, comprising administering any one of Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • FIG. 1 shows a dose-dependent curve of PSA-luciferase activities in response to representative compounds in transiently transfected LNCaP cells treated with synthetic androgen (R1881).
  • FIG. 2A shows the percent PSA-luciferase activities of representative compounds in LNCaP cells transiently transfected with the PSA (6.1 kb)-luciferase reporter and treated without or with R1881.
  • FIG. 2B shows the percent PSA-luciferase activities of representative compounds in LNCaP cells co-transfected with an expression vector for AR-V7 and the PSA-luciferase reporter and treated without or with R1881.
  • FIG. 3 shows concentration of representative compounds in plasma of male CD-1 mice after a single dose PO (oral) dose.
  • FIG. 4 shows change in tumor volume in male NCG mice bearing LNCaP tumors after oral administration of representative compounds.
  • FIG. 5 shows change in % body weight in male NCG mice bearing LNCaP tumors after oral administration of representative compounds.
  • FIG. 6 shows individual tumor volume change from baseline measured at the end of experiment for oral administration of representative compounds to male NCG mice bearing LNCaP tumors.
  • FIG. 7 shows concentration dependent effects on cell proliferation of LNCaP, PC3, and LNCaP95 cells treated with Compound A13.
  • 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.
  • 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.
  • 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.).
  • a androgen receptor modulator refers to one or more androgen receptor modulators or at least one androgen receptor modulator.
  • the terms “a” (or “an”), “one or more” and “at least one” are used interchangeably herein.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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).
  • HPLC high pressure liquid chromatography
  • 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.
  • a prodrug includes a compound of formula (I), (IA), (IB), (IC), (II), (IIA), (IIIA), (IIB), (III), (IV), (V), (VA), (VI), (A), (A-I), (B)-(D), (E), (E-I)-(E-VII), (F), (G), (G-I), (G-II), (H), and (H-I), 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.
  • terapéuticaally 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.
  • 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.
  • 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
  • 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.
  • laboratory animals e.g., mice, rats, monkeys, dogs, etc.
  • household pets e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits
  • non-domestic animals such as wildlife and the like.
  • substantially refers to the complete or nearly complete extent or degree of an action, characteristic, property, state, structure, item, or result.
  • 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.
  • compositions 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.
  • 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
  • Amino refers to the —NH 2 radical.
  • Halo or “halogen” refers to bromo, chloro, fluoro or iodo radical, including their radioisotopes.
  • 123 I refers to the radioactive isotope of iodine having atomic mass 123.
  • the compounds of Formula I can comprise at least one 123 I moiety. Throughout the present application, where structures depict a 123 I moiety at a certain position it is meant that the I moiety at this position is enriched for 123 I. In other words, the compounds contain more than the natural abundance of 123 I at the indicated position(s). It is not required that the compounds comprise 100% 123 I at the indicated positions, provided 123 I is present in more than the natural abundance.
  • the 123 I isotope is enriched to greater than 50%, greater than 60%, greater than 70%, greater than, 80% or greater than 90%, relative to 127 I.
  • 18 F refers to the radioactive isotope of fluorine having atomic mass 18.
  • F or 19 F refers to the abundant, non-radioactive fluorine isotope having atomic mass 19.
  • the compounds of Formula I can comprise at least one 18 F moiety. Throughout the present application, where structures depict a 18 F moiety at a certain position it is meant that the F moiety at this position is enriched for 18 F. In other words, the compounds contain more than the natural abundance of 18 F at the indicated position(s).
  • the compounds comprise 100% 18 F at the indicated positions, provided 18 F is present in more than the natural abundance.
  • 18 F isotope is enriched to greater than 50%, greater than 60%, greater than 70%, greater than 80% or greater than 90%, relative to 19 F.
  • Niro refers to the —NO 2 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 C 1 -C 12 alkyl, an alkyl comprising up to 10 carbon atoms is a C 1 -C 10 alkyl, an alkyl comprising up to 6 carbon atoms is a C 1 -C 6 alkyl and an alkyl comprising up to 5 carbon atoms is a C 1 -C 5 alkyl.
  • a C 1 -C 5 alkyl includes C 5 alkyls, C 4 alkyls, C 3 alkyls, C 2 alkyls and C 1 alkyl (i.e., methyl).
  • a C 1 -C 6 alkyl includes all moieties described above for C 1 -C 5 alkyls but also includes C 6 alkyls.
  • a C 1 -C 10 alkyl includes all moieties described above for C 1 -C 5 alkyls and C 1 -C 6 alkyls, but also includes C 7 , C 8 , C 9 and C 10 alkyls.
  • a C 1 -C 12 alkyl includes all the foregoing moieties, but also includes C 11 and C 12 alkyls.
  • Non-limiting examples of C 1 -C 12 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.
  • 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.
  • C 1 -C 12 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 C 2 -C 12 alkenyl
  • an alkenyl comprising up to 10 carbon atoms is a C 2 -C 10 alkenyl
  • an alkenyl group comprising up to 6 carbon atoms is a C 2 -C 6 alkenyl
  • an alkenyl comprising up to 5 carbon atoms is a C 2 -C 5 alkenyl.
  • a C 2 -C 5 alkenyl includes C 5 alkenyls, C 4 alkenyls, C 3 alkenyls, and C 2 alkenyls.
  • a C 2 -C 6 alkenyl includes all moieties described above for C 2 -C 5 alkenyls but also includes C 6 alkenyls.
  • a C 2 -C 10 alkenyl includes all moieties described above for C 2 -C 5 alkenyls and C 2 -C 6 alkenyls, but also includes C 7 , C 8 , C 9 and C 10 alkenyls.
  • a C 2 -C 12 alkenyl includes all the foregoing moieties, but also includes C 11 and C 12 alkenyls.
  • Non-limiting examples of C 2 -C 12 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-noneny
  • 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.
  • C 2 -C 12 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 C 2 -C 12 alkynyl
  • an alkynyl comprising up to 10 carbon atoms is a C 2 -C 10 alkynyl
  • an alkynyl group comprising up to 6 carbon atoms is a C 2 -C 6 alkynyl
  • an alkynyl comprising up to 5 carbon atoms is a C 2 -C 5 alkynyl.
  • a C 2 -C 5 alkynyl includes C 5 alkynyls, C 4 alkynyls, C 3 alkynyls, and C 2 alkynyls.
  • a C 2 -C 6 alkynyl includes all moieties described above for C 2 -C 5 alkynyls but also includes C 6 alkynyls.
  • a C 2 -C 10 alkynyl includes all moieties described above for C 2 -C 5 alkynyls and C 2 -C 6 alkynyls, but also includes C 7 , C 8 , C 9 and C 10 alkynyls.
  • a C 2 -C 12 alkynyl includes all the foregoing moieties, but also includes C 11 and C 12 alkynyls.
  • Non-limiting examples of C 2 -C 12 alkenyl include ethynyl, propynyl, butynyl, pentynyl and the like. Unless stated otherwise specifically in the specification, an alkyl 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.
  • C 2 -C 12 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 —OR a where R a 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 —NHR a or —NR a R a where each R a 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)R a moiety, wherein R a is an alkyl, alkenyl or alkynyl radical as defined above.
  • R a 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 in R a , as defined above.
  • C 1 -C 10 acyl refers to alkylcarbonyl group as defined above, where R a is C 1 -C 10 alkyl, C 1 -C 10 alkenyl, or C 1 -C 10 alkynyl radical as defined above. Unless stated otherwise specifically in the specification, an alkyl carbonyl 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.
  • 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.
  • aryl is meant to include aryl radicals that are optionally substituted.
  • Alkyl or “arylalkyl” refers to a radical of the formula —R b —R c where R b is an alkylene group as defined above and R c 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.
  • alkenyl or “arylalkenyl” refers to a radical of the formula —R b —R c where R b is an alkenylene o group as defined above and R c is one or more aryl radicals as defined above. Unless stated otherwise specifically in the specification, an aralkenyl group can be optionally substituted.
  • Alkynyl or “arylalkynyl” refers to a radical of the formula —R b —R c where R b is an alkynylene group as defined above and R c 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 —R b —R d where R b is an alkylene, alkenylene, or alkynylene group as defined above and R d 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 haloalkenyl group can be optionally substituted.
  • Heterocyclyl 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. Heterocyclyl or heterocyclic rings include heteroaryls as defined below.
  • 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.
  • 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-thio
  • Heterocyclylalkyl refers to a radical of the formula —R b —R e where R b 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 —R b —R e where R b 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 —R b —Re where R b 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.
  • 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,
  • 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 —R b —R f where R b is an alkylene chain as defined above and R f 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 —R b —R f where R b is an alkenylene, chain as defined above and R f 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 —R b —R f where R b is an alkynylene chain as defined above and R f 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 —SR a where R a 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.
  • substituted 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 hydroxyl 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.
  • a higher-order bond e.g., a double- or triple-bond
  • nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • substituted includes any of the above groups in which one or more hydrogen atoms are replaced with —NR g R h , —NR g C( ⁇ O)R h , —NR g C( ⁇ O)NR g R h , —NR g C( ⁇ O)OR h , —NR g SO 2 R h , —OC( ⁇ O)NR g R h , —OR g , —SR g , —SOR g , —SO 2 R g , —OSO 2 R g , —SO 2 OR g , ⁇ NSO 2 R g , and —SO 2 NR g R h .
  • “Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with —C( ⁇ O)R g , —C( ⁇ O)OR g , —C( ⁇ O)NR g R h , —CH 2 SO 2 R g , —CH 2 SO 2 NR g R h .
  • R g and R h 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.
  • each of the foregoing substituents can also be optionally substituted with one or more of the above substituents.
  • 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.
  • fused refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention.
  • 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 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.
  • the cancer is prostate cancer or breast cancer.
  • the present disclosure provides compounds comprising the structure of formula (I):
  • the present disclosure provides compounds comprising the structure of formula (II):
  • the present disclosure provides compounds comprising the structure of formula (IIA):
  • the present disclosure provides compounds comprising the structure of formula (IIB):
  • the present disclosure provides compounds comprising the structure of formula (III):
  • the present disclosure provides compounds comprising the structure of formula (IIIA):
  • the present disclosure provides compounds comprising the structure of formula (IV):
  • the present disclosure provides compounds comprising the structure of formula (V):
  • the present disclosure provides compounds comprising the structure of formula (VA):
  • the present disclosure provides compounds comprising the structure of formula (VI):
  • the present disclosure provides compounds comprising the structure of formula (A):
  • the present disclosure provides compounds comprising the structure of formula (B)
  • —V-L is —CH 2 CH 2 C 1 , —CH 2 CH 2 CH 2 C 1 , —CH 2 CH 2 NH 2 , or —CH 2 CH 2 CH 2 NH 2 .
  • —Y—W— is a bond, —OCH 2 —, —OCH 2 CH 2 —, —OCH(CH 3 )—, —NH—, —NHCH 2 —, —NHC( ⁇ O)—, or —C( ⁇ O)NH—.
  • X is a bond, —CH 2 —, —C(CH 3 )H—, —C(CH 3 ) 2 —, or —CH 2 CH 2 —.
  • the present disclosure provides compounds comprising the structure of formula (C):
  • the present disclosure provides compounds comprising the structure of formula (D):
  • the present disclosure provides compounds comprising the structure of formula (E):
  • R 3 is selected from hydrogen, F, Cl, Br, I, —CN, —CF 3 , —OH, methyl, methoxy, —S(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NH 2 , —NHSO 2 CH 3 , —NHSO 2 CF 3 , —N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -C 3 alkyl), or —NHCO(C 1 -C 3 alkyl).
  • the present disclosure provides compounds comprising the structure of formula (F):
  • the present disclosure provides compounds comprising the structure of formula (G):
  • the present disclosure provides compounds comprising the structure of formula (G-II)
  • the present disclosure provides compounds comprising the structure of formula (H):
  • R 2A and R 2B are not both Cl.
  • R 2A and R 2B are Cl, then the other of R 2A and R 2B is not —CN.
  • R 2A and R 2B are not both Cl.
  • a and B are each independently 5- or 6-membered aryl or heteroaryl.
  • a and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene.
  • a and B are each phenyl.
  • A has a meta or para connectivity with X and Y.
  • B has a meta or para connectivity with X and Z.
  • a and B are phenyl and has one of the connectivity as shown:
  • C is aryl or heteroaryl.
  • C is 5- to 10-membered aryl or heteroaryl.
  • C is aryl.
  • C is phenyl or naphthyl.
  • C is aryl.
  • C is phenyl.
  • C is heteroaryl.
  • C monocyclic or bicyclic heteroaryl.
  • C is monocyclic heteroaryl.
  • C is 5- or 10-membered heteroaryl.
  • C is 5- or 6-membered heteroaryl, which is optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • C is 5- or 6-membered heteroaryl containing 1, 2, or 3 heteroatoms selected from O, S, or N, wherein the heteroaryl is optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • C is 5- or 6-membered heteroaryl containing 1 or 2 heteroatoms selected from O, S, or N, wherein the heteroaryl is optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, or pyrimidyl, which are each optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • C, which is substituted with (R 3 )n3, is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl.
  • C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl, which are each substituted with 1, 2, 3, 4, or 5 R 3 .
  • C is selected from
  • R 3a is C 1 -C 3 alkyl.
  • C is selected from
  • R 3a is C 1 -C 3 alkyl.
  • C is
  • C is
  • C is heterocyclyl.
  • C is saturated or partially saturated heterocycle.
  • C is monocyclic or bicyclic.
  • C is 5- to 7-membered heterocyclyl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • C is imidazolidine, imidazolidine-dione, or dihydrooxazole. In one embodiment, C is selected from
  • C is
  • D is —O—, —NH— or —NR 3 —; and U is each independently O, S, or NR 16 .
  • D is —NH— or —NR 3 —.
  • at least one U is O.
  • each U is O.
  • at least one R 3 is —SO 2 CH 3 , —NHSO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , or —NHCOCH 3 .
  • C is aryl. In some embodiments, C is phenyl or naphthyl. In one embodiment of the compounds of formula (I)-(VA) or (A), C is phenyl.
  • C is bicyclic heteroaryl or heterocyclyl. In one embodiment, C is
  • Z is a bond, —(CR 8 R 9 ) m —, —O—, —C( ⁇ O)—, —S—, —S( ⁇ O)—, —SO 2 —, or —NR 7 —.
  • Z is —(CR 8 R 9 ) m —, —O—, —C( ⁇ O)—, —S—, —S( ⁇ O)—, —SO 2 —, or —NR 7 —.
  • Z is —C( ⁇ O)—.
  • Z is a bond, —CH 2 —, —C(CH 3 )H—, —O—, —S—, —NH—, —NCH 3 —, or —N(COCH 3 )—.
  • Z is —CH 2 —, —C(CH 3 )H—, —O—, —S—, —NH—, —NCH 3 —, or —N(COCH 3 )—.
  • Z is a bond, —CH 2 —, —O—, or —NCH 3 —.
  • Z is a bond, —CH 2 —, —O—, or —NH—.
  • Z is —O—.
  • “compounds of formula (I)-(IV) and (A)-(H-I)” refers to compounds of formula (I), (IA), (IB), (IC), (II), (IIA), (IIIA), (IIB), (III), (IV), (IVA), (V), (VA), (VI), (A), (A-I), (B)-(D), (E), (E-I)-(E-VII), (F), (G), (G-I), (G-II), (H), and (H-I).
  • Y is a bond, —(CR 8 R 9 ) m —, —O—, —C( ⁇ O)—, —S—, —S( ⁇ O)—, —SO 2 —, or —NR 7 —.
  • Y is —(CR 8 R 9 ) m —, —O—, —C( ⁇ O)—, —S—, —S( ⁇ O)—, —SO 2 —, or —NR 7 —.
  • Y is —C( ⁇ O)—.
  • Y is a bond, —CH 2 —, —C(CH 3 )H—, —O—, —S—, —NH—, —NCH 3 —, or —N(COCH 3 )—.
  • Y is —CH 2 —, —C(CH 3 )H—, —O—, —S—, —NH—, —NCH 3 —, or —N(COCH 3 )—.
  • Y is a bond, —CH 2 —, —O—, or —NCH 3 —.
  • Y is a bond, —CH 2 —, —O—, or —NH—. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), Y is —O—.
  • V is a bond, —(CR 8a R 9a ) m —, or —C( ⁇ O)—. In some embodiments, V is bond, or —(CR 8a R 9a ) m —.
  • V is —(CR 8a R 9a ) m —, wherein m is 1, 2, or 3.
  • V is —(CR 8a R 9a ) m —, wherein R 8a and R 8b are each independently hydrogen, —OH, halogen, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 3 alkenyl, optionally substituted C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —NR 13 R 14 , optionally substituted —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 COR 16 , optionally substituted —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , or optionally
  • V is —(CR 8a R 9a ) m —, wherein R 8a and R 8b are each independently hydrogen, —OH, halogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 COR 16 , —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , or —(C 1 -C 3 alkyl)-CONR 14 R 15 ; or R 8a and R 8b , on the same carbon atom or on a different carbon atom, taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl.
  • V is —CH 2 —, —CH 2 CH 2 —, —CH(CH 3 )CH 2 —, —CH 2 CH(CH 3 )—, or —CH 2 CH 2 CH 2 —.
  • V is —CH 2 —, —CH 2 CH 2 —, or —CH 2 CH 2 CH 2 —, each optionally substituted with one or more of —OH, halogen, or C 1 -C 3 alkyl.
  • V is —CH 2 —, —CH 2 CH 2 —, —CH 2 CH(OH)CH 2 — or —CH 2 CH 2 CH 2 —.
  • V is —CH 2 —, —CH 2 CH 2 —, or —CH 2 CH 2 CH 2 —.
  • V is —CH 2 — or —CH 2 CH 2 —.
  • V is —CH 2 — and L is halogen, —NH 2 , or —CF 3 ; or V is —CH 2 CH 2 — and L is halogen or —NH 2 .
  • L is hydrogen, halogen, —CF 2 H, —CF 3 , —CN, —O(C 1 -C 3 alkyl), —NR 11 R 12 , or —CONR 11 R 12 .
  • L is hydrogen, halogen, —CF 2 H, —CF 3 , —CN, —O(C 1 -C 3 alkyl), —NH 2 , —NH(C 1 -C 3 alkyl), —N(C 1 -C 3 alkyl) 2 , —CONH 2 , —CONH(C 1 -C 3 alkyl), or —CON(C 1 -C 3 alkyl) 2 .
  • L is hydrogen, halogen, —CF 3 , or —NH 2 .
  • L is halogen, —CCl 3 , —CCl 2 , —CF 3 , or —NH 2 .
  • L is halogen, —CF 3 , or —NH 2 .
  • L is hydrogen or halogen.
  • L is halogen.
  • L is Cl, or Br.
  • L is Cl.
  • W is a bond.
  • W is —(CR 8a R 9a ) m —, —C( ⁇ O)—, —N(R′)CO—, —CONR 7 —, or —NSO 2 R 7 —.
  • W is —(CR 8a R 9a ) m —, wherein m is 1, 2, or 3.
  • W is a bond, —CH 2 —, —C(CH 3 )H—, —C( ⁇ O)—, —N(R 7 )CO—, or —CONR 7 —, wherein R 7 is H or C 1 -C 6 alkyl. In some embodiments, W is a bond, —CH 2 —, —C(CH 3 )H—, —C( ⁇ O)—, —N(R 7 )CO—, or —CONR 7 —, wherein R 7 is H or C 1 -C 3 alkyl.
  • W is a bond, —CH 2 —, —C(CH 3 )H—, —C( ⁇ O)—, —NHCO—, —N(C 1 -C 3 alkyl)CO—, —CONH—, or —CON(C 1 -C 3 alkyl)-.
  • W is a bond, —CH 2 —, or —C(CH 3 )H—.
  • W is a —CH 2 — or —C(CH 3 )H—.
  • —Y—W— is a bond, —OCH 2 —, —OCH 2 CH 2 —, —OCH(CH 3 )—, —NH—, —NHCH 2 —, —NHC( ⁇ O)—, or —C( ⁇ O)NH—.
  • —Y—W— is —OCH 2 —, —OCH 2 CH 2 —, or —OCH(CH 3 )—.
  • —Z—V-L is —Z—CH 2 CH 2 C 1 , —Z—CH 2 CH 2 CH 2 C 1 , —Z—CH 2 CH 2 NH 2 , or —Z—CH 2 CH 2 CH 2 NH 2 , wherein Z is a bond, —O—, —NH—, or —N(COCH 3 )—.
  • —Z—V-L is —OCH 3 .
  • —Z—V-L is —O—CH 2 CH 2 Cl or —O—CH 2 CH 2 CH 2 Cl.
  • —V-L is —CH 2 CH 2 C 1 , —CH 2 CH 2 CH 2 C 1 , —CH 2 CH 2 NH 2 , or —CH 2 CH 2 CH 2 NH 2 . In one embodiment, —V-L is —CH 3 .
  • X is a bond, —(CR 5 R 6 ) t —, —O—, —C( ⁇ O)—, —S—, —S( ⁇ O)—, —SO 2 —, or —NR 7 —.
  • X is a bond, —(CR 5 R 6 ) t —, —O—, —C( ⁇ O)—, —S—, —S( ⁇ O)—, —SO 2 —, or —NR 7 —, wherein R 7 is H or C 1 -C 6 alkyl.
  • X is a bond, —(CR 5 R 6 ) t —, or —NR 7 —. In some embodiments, X is a bond or —(CR 5 R 6 ) t —. In some embodiments, X is a bond, —CH 2 —, —C(CH 3 )H—, —C(CH 3 ) 2 —, —CH 2 CH 2 —, —NH—, or —N(C 1 -C 6 alkyl)-.
  • X is a bond, —CH 2 —, —C(CH 3 )H—, —C(CH 3 ) 2 —, —CH 2 CH 2 —, —NH—, —N(CH 3 )—, —N(CH 2 CH 3 )—, —N(iPr)—, or —N(tBu)-.
  • X is a bond, —CH 2 —, —C(CH 3 )H—, —C(CH 3 ) 2 —, or —CH 2 CH 2 —.
  • X is —CH 2 —, —C(CH 3 )H—, or —C(CH 3 ) 2 -. In one embodiment, X is —C(CH 3 ) 2 -.
  • R 1 and R 2 are each independently halogen, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , —(C 1 -C 3 alkyl)-CONR 14 R 15 , —SO 2 NR 14 R 15 , —(C 1 -C 3 alkyl)
  • R 1 and R 2 are each independently hydrogen, halogen, —CN, —CF 3 , —OH, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted —(C 1 -C 6 alkyl)-(C 1 -C 6 alkoxy), optionally substituted —(C 1 -C 6 alkyl)-OH, —NR 13 R 14 , optionally substituted —(C 1 -C 6 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , optionally substituted —(C 1 -C 6 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , optionally substituted —(C 1 -C 6 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , optionally substituted —(C 1 -C 6 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , optionally substituted
  • R 1 and R 2 are each independently hydrogen, halogen, —CN, —CF 3 , —OH, optionally substituted C 1 -C 3 alkyl, C 1 -C 3 alkoxy, optionally substituted —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), optionally substituted —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , optionally substituted —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , optionally substituted —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , optionally substituted —(C 1 -C 3 alkyl)-CONR 14 R 15 , —SO 2 NR 14 R 15 , optionally substituted —(C
  • R 1 and R 2 are each independently hydrogen, halogen, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , —(C 1 -C 3 alkyl)-CONR 14 R 15 , —SO 2 NR 14 R 15 , —(C 1 -C 3 alkyl)-CONR 14 R 15 , —SO 2 NR 14 R 15 , —(
  • R 1 and R 2 are each independently hydrogen, halogen, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, or —CONR 14 R 15 .
  • R 1 and R 2 are each independently hydrogen, halogen, —CN, —CF 3 , —OH, methyl, methoxy, or —CONH 2 .
  • R 1 and R 2 are each independently hydrogen, C 1 , —CN, —CF 3 , —OH, methyl, methoxy, or —CONH 2 .
  • R 1 and R 2 are each independently hydrogen, halogen, —CN, —CF 3 , —OH, or methyl. In one embodiment, R 1 and R 2 are each independently C 1 , —CN, —CF 3 , —OH, methyl, methoxy, or —CONH 2 . In one embodiment of the compounds of formula (I)-(VI), R 1 and R 2 are each independently halogen, —CN, —CF 3 , —OH, or methyl.
  • R 1 and R 2 are each halogen, methyl, —CF 3 , or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(F), R 1 and R 2 are each halogen or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), at least one of R 1 and R 2 is Cl or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), at least two of R 1 and R 2 are each independently Cl or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), R 1 and R 2 are each Cl or —CN.
  • R 1 and R 2 are each independently optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In one embodiment, R 1 and R 2 are each independently 3- to 7-membered carbocyclyl, 3- to 7-membered heterocyclyl, phenyl, or 5- to 6-membered heteroaryl.
  • R 1 have one of the connectivity as shown below with respect to X and Y:
  • R 2 have one of the connectivity as shown below with respect to X and Z:
  • 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.
  • 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.
  • 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.
  • R 3 is selected from hydrogen, halogen, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, —SR 16 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted —(C 1 -C 6 alkyl)-(C 1 -C 6 alkoxy), optionally substituted —(C 1 -C 6 alkyl)-OH, —NR 13 R 14 , optionally substituted —(C 1 -C 6 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , optionally substituted —(C 1 -C 6 alkyl)NR 14 SO 2 R 16 , —NR 14
  • R 3 is selected from hydrogen, halogen, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, —SR 16 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted —(C 1 -C 6 alkyl)-(C 1 -C 6 alkoxy), optionally substituted —(C 1 -C 6 alkyl)-OH, —NR 13 R 14 , optionally substituted —(C 1 -C 6 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , optionally substituted —(C 1 -C 6 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , optionally substituted —(C 1 -C 6 alkyl)-NR 14
  • R 3 is hydrogen, halogen, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, —SR 16 , optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 3 alkenyl, optionally substituted C 2 -C 3 alkynyl, optionally substituted C 1 -C 3 alkoxy, optionally substituted —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), optionally substituted —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , optionally substituted —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , optionally substituted —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , optionally substituted —(C 1 -C 3 alkyl)-NR 14 COR 16 , optional
  • R 3 is selected from —NR 14 SO 2 R 16 , optionally substituted —(C 1 -C 6 alkyl)NR 14 SO 2 R 16 , or optionally substituted —SO 2 R 16 ; wherein R 16 is hydrogen, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 3 alkenyl, optionally substituted C 2 -C 3 alkynyl, C 3 -C 6 cycloalkyl, or phenyl.
  • R 3 is selected from —NR 14 SO 2 R 16 , —(C 1 -C 6 alkyl)NR 14 SO 2 R 16 , or —SO 2 R 16 ; wherein R 16 is hydrogen, C 1 -C 3 alkyl, —(C 1 -C 3 alkyl)-NH 2 , C 3 -C 6 cycloalkyl, or phenyl.
  • R 3 is selected from hydrogen, halogen, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, —S(C 1 -C 3 alkyl), C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , —(C 1 -C 6 alkyl)-NR 14 COR 16 , —(C 1 -C 6 alkyl)-NR 14 COR 16 , —(C 1
  • R 3 is selected from hydrogen, F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, —S(C 1 -C 3 alkyl), —SO(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , —CH 2 NHSO 2 CH 3 , —CH 2 N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(
  • R 3 is selected from hydrogen, F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, —S(C 1 -C 3 alkyl), —SO(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —CH 2 N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -C 3 alkyl), or —N(CH 3 )CO(C 1 -C 3 alkyl),
  • R 3 is selected from hydrogen, F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, methyl, —SCH 3 , —SO 2 CH 3 , —NHSO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —CH 2 NHSO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , or —NHCOCH 3 .
  • R 3 is selected from hydrogen, F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, methyl, —SCH 3 , —SO 2 CH 3 , —NHSO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , or —NHCOCH 3 .
  • R 3 is selected from F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, —S(C 1 -C 3 alkyl), —SO(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —CH 2 N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -C 3 alkyl), or —N(CH 3 )CO(C 1 -C 3 alkyl), or
  • R 3 is selected from F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, —S(C 1 -C 3 alkyl), —SO(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , —CH 2 NHSO 2 CH 3 , —CH 2 N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -
  • R 3 is selected from F, Cl, Br, I, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, methyl, —SCH 3 , —SO 2 CH 3 , —NHSO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , or —NHCOCH 3 .
  • R 3 is —SO 2 CH 3 , —NHSO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , or —NHCOCH 3 .
  • R 3 is oxo, ⁇ S, ⁇ NR 16 , C 1 -C 3 alkyl, —SO 2 (C 1 -C 3 alkyl), or —NHSO 2 (C 1 -C 3 alkyl). In one embodiment, at least one of R 3 is oxo, ⁇ S, or ⁇ NR 16 . In one embodiment, at least one of R 3 is oxo, ⁇ S, or ⁇ NR 16 , wherein R 16 is H or C 1 -C 3 alkyl.
  • R 3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —S(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NH 2 , —NHSO 2 CH 3 , —NHSO 2 CF 3 , —N(CH 3 )SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , —CH 2 NHSO 2 CH 3 , —CH 2 N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl)
  • R 3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —S(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NH 2 , —NHSO 2 CH 3 , —NHSO 2 CF 3 , —N(CH 3 )SO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —CH 2 N(CH 3 )SO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -
  • R 3 on a sp 3 carbon is each selected from hydrogen, halogen, oxo, ⁇ S, ⁇ NR 16 , —CN, —CF 3 , —OH, —S(C 1 -C 3 alkyl), C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , —(C 1 -C 6 alkyl
  • R 3 on a sp 2 carbon is each selected from hydrogen, halogen, —CN, —CF 3 , —OH, —S(C 1 -C 3 alkyl), C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , —(C 1 -C 6 alkyl)-NR 14 COR 16 , —CONR
  • R 3 on a nitrogen atom is each selected from C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —(C 1 -C 3 alkyl)-NR 13 R 14 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —(C 1 -C 6 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , —(C 1 -C 3 alkyl)-CONR 14 R 15 , —(C 1 -C 3 alkyl)-SO 2 NR 14 R 15 , or —(C 1 -C 6 alkyl)
  • At least one R 3 is selected from —CN, C 1 -C 3 alkoxy, —CONH 2 , —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , or —SO 2 CH 3 and the other R 3 , if present, is selected from —CN, —CF 3 , C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —S(C 1 -C 3 alkyl), —SO 2 (C 1 -C 3 alkyl), —NH 2 , —(C 1 -C 3 alkyl)NH 2 , —NHSO 2 CH 3 , —NHSO 2 CF 3 , —N(CH
  • At least one R 3 is selected from —NHSO 2 CH 3 , —NHSO 2 CH 2 CH 3 , or —SO 2 CH 3 and the other R 3 , if present, is selected from —CN, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, —SO 2 (C 1 -C 3 alkyl), —NH 2 , —(C 1 -C 3 alkyl)NH 2 , —NHSO 2 CH 3 , —N(CH 3 )SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , —N(CH 3 )SO 2 CH 2 CH 3 , —SO 2 NH 2 , —CONH 2 , —CON(C 1 -C 3 alkyl) 2 , —CONH(C 1 -C 3 alkyl), —NHCO(C 1 -C 3 alkyl), —N(CH 3 )COO(C 1 -C 3 alkyl), —NHCO(C 1 -
  • R 3 is not hydrogen
  • At least one R 3 is —SO 2 CH 3 , —NHSO 2 CH 3 , —NCH 3 SO 2 CH 3 , —NHSO 2 CH 2 CH 3 , or —N(CH 3 )SO 2 CH 2 CH 3 . In one embodiment of the compounds of formula (I)-(VI) and (A)-(G-II), at least one R 3 is —SO 2 CH 3 , —NHSO 2 CH 3 , or —NCH 3 SO 2 CH 3 .
  • R 3 is heterocyclyl
  • R 3 is heterocyclyl selected from
  • R 3 is —NR 14 SO 2 R 16 , wherein R 14 and R 16 together form a 5 or 6 membered ring including the nitrogen and sulfur atoms.
  • R 3 is —NR 14 SO 2 R 16 , wherein R 16 is optionally substituted C 1 -C 6 alkyl.
  • R 3 is —NR 14 SO 2 R 16 , wherein R 16 is C 1 -C 6 alkyl optionally substituted with one or more groups selected from halogen, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, —NH 2 , —NH(C 1 -C 3 alkyl), —N(C 1 -C 3 alkyl) 2 , —SCH 3 .
  • R 3 is —NR 14 SO 2 R 16 , wherein R 16 is C 1 -C 3 alkyl substituted with —NH 2 .
  • R 5 and R 6 are each independently hydrogen, halogen, —OH, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, or C 1 -C 3 alkoxy; or R 5 and R 6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl.
  • R 5 and R 6 are each independently hydrogen, halogen, —OH, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl.
  • R 5 and R 6 are hydrogen, halogen, —OH, or C 1 -C 3 alkyl. In one embodiment, R 5 and R 6 are each independently hydrogen, F, —OH, or C 1 -C 3 alkyl. In one embodiment, R 5 and R 6 are each independently, hydrogen, F, —OH, or methyl. In one embodiment, R 5 and R 6 are each H. In one embodiment, R 5 and R 6 are each methyl. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), R 5 and R 6 are each H or methyl.
  • R 7 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 7 is hydrogen, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 3 alkenyl, optionally substituted C 2 -C 3 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 7 is hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl.
  • R 7 is hydrogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl. In some embodiments, R 7 is hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl. In some embodiments, R 7 is hydrogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, or C 2 -C 3 alkynyl. In some embodiments, R 7 is hydrogen or C 1 -C 6 alkyl. In some embodiments, R 7 is hydrogen or C 1 -C 4 alkyl. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), R 7 is hydrogen or C 1 -C 3 alkyl.
  • R 8a and R 9a are each independently hydrogen, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted C 1 -C 6 alkoxy, —NR 13 R 14 , optionally substituted —(C 1 -C 6 alkyl)-NR 13 R 14 , —NR 14 COR 16 , optionally substituted —(C 1 -C 6 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , optionally substituted —(C 1 -C 6 alkyl)-CONR 14 R 15 , optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or R 8a and R 9a taken together form an optionally substituted carbocycl
  • R 8a and R 8b are each independently hydrogen, —OH, halogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 COR 16 , —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , or —(C 1 -C 3 alkyl)-CONR 14 R 15 ; or R 8a and R 8b taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl.
  • R 8a and R 9a are each independently hydrogen, halogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 COR 16 , —(C 1 -C 3 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , or —(C 1 -C 3 alkyl)-CONR 14 R 15 .
  • R 8a and R 9a are not —OH. In one embodiment, R 8a and R 9a are not —OH.
  • R 7 and R 8a taken together form an optionally substituted heterocyclyl. In one embodiment, R 7 and R 8a taken together form an optionally substituted 3- to 7-membered heterocycle.
  • R 8 and R 9 are each independently hydrogen, halogen, or C 1 -C 3 alkyl.
  • R 10 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 10 is hydrogen, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 3 alkenyl, optionally substituted C 2 -C 3 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 10 is hydrogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl.
  • R 10 is hydrogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl.
  • R 10 is hydrogen or C 1 -C 3 alkyl.
  • R 11 and R 12 are each independently hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 11 and R 12 are each independently hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, or optionally substituted C 2 -C 6 alkynyl. In some embodiments.
  • R 11 and R 12 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl. In some embodiments. R 11 and R 12 are each independently hydrogen or C 1 -C 3 alkyl.
  • R 11 and R 12 taken together form an optionally substituted heterocyclyl. In one embodiment, R 11 and R 12 taken together form an optionally substituted 3- to 7-membered heterocyclyl. In other embodiments, R 11 and R 12 taken together form 3- to 7-membered heterocyclyl.
  • R 13 and R 14 are each independently hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 13 and R 14 are each independently hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, or optionally substituted C 2 -C 6 alkynyl.
  • R 13 and R 14 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl. In some embodiments R 13 and R 14 are each independently hydrogen or C 1 -C 3 alkyl.
  • R 15 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In one embodiment, R 15 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, or optionally substituted C 2 -C 6 alkynyl.
  • R 15 is hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl. In some embodiments, R 15 is hydrogen or C 1 -C 3 alkyl.
  • R 14 and R 15 taken together form an optionally substituted heterocyclyl. In one embodiment, R 14 and R 15 taken together form an optionally substituted 3- to 7-membered heterocyclyl. In other embodiments, R 14 and R 15 taken together form 3- to 7-membered heterocyclyl.
  • R 16 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R 16 is hydrogen, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 3 alkenyl, optionally substituted C 2 -C 3 alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments, R 16 is hydrogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl.
  • R 16 is hydrogen, C 1 -C 3 alkyl, C 2 -C 3 alkenyl, or C 2 -C 3 alkynyl. In some embodiments, R 16 is hydrogen or C 1 -C 3 alkyl.
  • m is 1 or 2.
  • t is 1 or 2. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), t is 1.
  • optional substituent is selected from halogen, —CN, —CF 3 , —OH, —S(C 1 -C 3 alkyl), C 1 -C 3 alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, C 1 -C 3 alkoxy, —(C 1 -C 3 alkyl)-(C 1 -C 3 alkoxy), —(C 1 -C 3 alkyl)-OH, —NR 13 R 14 , —(C 1 -C 3 alkyl)-NR 13 R 14 , —NR 14 SO 2 R 16 , —(C 1 -C 3 alkyl)NR 14 SO 2 R 16 , —NR 14 COR 16 , —(C 1 -C 6 alkyl)-NR 14 COR 16 , —CONR 14 R 15 , —(C 1 -C 3 alkyl), —(C 1 -C 3 alkyl), —CONR 14 R 15
  • the optional substituent is selected from halogen, —CN, —CF 3 , —OH, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, —NH 2 , —SCH 3 , —SO 2 CH 3 , —NHSO 2 CH 3 , —CH 2 NHSO 2 CH 3 , —SO 2 NH 2 , —CONH 2 , or —NHCOCH 3 .
  • a and B are each monocyclic ring.
  • B is phenyl, pyridyl, or pyrimidyl.
  • Z and V are not both a bond.
  • C is a 4- to 10-membered ring.
  • X is a bond, —CH 2 —, —C(CH 3 )H—, —C(CH 3 ) 2 —, or —CH 2 CH 2 —. In one embodiment, X is —CH 2 —, —C(CH 3 )H—, or —C(CH 3 ) 2 —. In some embodiments, X is —C(CH 3 ) 2 —.
  • X is —NR 7 —.
  • X is —NH—, —N(CH 3 )—, —N(CH 2 CH 3 )—, —N(iPr)—, or —N(tBu)-.
  • Y is —O—. In one embodiment of the compounds of formula (D)-(H-I), Z is —O—. In one embodiment of the compounds of formula (D)-(H-I), Y and Z are both —O—.
  • —V-L is CH 2 CH 2 C 1 , —CH 2 CH 2 CH 2 C 1 , or —CH 3 . In some embodiments, —V-L is CH 2 CH 2 Cl or —CH 2 CH 2 CH 2 Cl.
  • n1 is 0.
  • n2 is 0, 1, or 2. In some embodiments, n2 is 2. In some embodiments, n2 is 2 and R 2 are each ortho to Z. In other embodiments, n2 is 2 and R 2 are each ortho to Z, wherein R 2 is halogen or —CN.
  • the compound in one embodiment of the compound of formula (I)-(VI) and (A)-(H-I), can be a stereoisomer.
  • the carbon attached to R 5 and R 6 can be in an S configuration or an R configuration.
  • a hydrogen atom can be replaced with a deuterium atom.
  • the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I) is selected from Table A below, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A3, A5, A7, A13, A17, A18, A22, A23, A24, A25, A28, A30, A31, A32, A34, A35, A38, A40, A41, A42, A45, A49, A52, A53, A54, A56, A57, A58, A62, A63, A64, A65, A68, A73, A74, A75, or A76, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A1, A2, A4, A6, A8, A9, A10, A11, A12, A14, A15, A16, A19, A20, A21, A26, A27, A29, A33, A36, A37, A39, A43, A44, A46, A47, A48, A50, A51, A55, A59, A60, A61, A66, A67, A69, A70, A71, A72, A77, A78, A79, A80, A81, A82, A83, A84, A85, A86, A87, A88, A89, A90, A91, A92, A93, A94, A95, A96, or A97, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A98-A186, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A187-A211, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A1-A211, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A212-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A1-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound in one embodiment of the compound of formula (I)-(IV), (VI), (B) or (C), the compound is selected from Table B below, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(IV), (VI), (B) or (C), the compound is selected from Compounds B1, B2, B3, or B6 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds B4, B5, B7, B8, B9, B10, or B11 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the compound is selected from Compounds B1-B11 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound of any one of formula (I), (IA), (IB), (IC), (II), (IIA), (IIIA), (IIB), (III), (IV), (IVA), (V), (VA), (VI), (A), (A-I), (B)-(D), (E), (E-I)-(E-VII), (F), (G), (G-I), (G-II), (H), and (H-I) (“formula (I)-(VI) and (A)-(H-I)”) or compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A3, A5, A7, A13, A17, A18, A22, A23, A24, A25, A28, A30, A31, A32, A34, A35, A38, A40, A41, A42, A45, A49, A52, A53, A54, A56, A57, A58, A62, A63, A64, A65, A68, A73, A74, A75, A76, B1, B2, B3, or B6 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1, A2, A4, A6, A8, A9, A10, A11, A12, A14, A15, A16, A19, A20, A21, A26, A27, A29, A33, A36, A37, A39, A43, A44, A46, A47, A48, A50, A51, A55, A59, A60, A61, A66, A67, A69, A70, A71, A72, A77, A78, A79, A80, A81, A82, A83, A84, A85, A86, A87, A88, A89, A90, A91, A92, A93, A94, A95, A96, A97, B4, B5, B7, B8, B9, B10, or B11, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prod
  • the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A98-A186, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A187-A211, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A212-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1-A211 or B1-B11, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound selected from Compounds A1-A234 or B1-B11, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or a pharmaceutically acceptable excipient and a compound of any one of formula (D)-(H-I), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • the present compounds find use in any number of methods.
  • the compounds are useful in methods for modulating androgen receptor (AR).
  • the present disclosure provides the use of any one of the foregoing compounds of formula (I)-(VI) and (A)-(H-I), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, for modulating androgen receptor (AR) activity.
  • 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.
  • the modulating AR is binding to AR. In other embodiments, the modulating AR is inhibiting AR.
  • 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).
  • 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.
  • the indication is prostate cancer.
  • 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.
  • a method of treating a condition associated with cell proliferation in a patient in need thereof comprising administering a compounds of formula (I)-(VI) and (A)-(H-I), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, to a subject in need thereof.
  • the present invention provides a method of treating cancer or tumors.
  • the present invention provides a method of treating prostate cancer or breast cancer.
  • a method of reducing, inhibiting, or ameliorating proliferation comprising administering a therapeutically effective amount of a compound of formula (I)-(VI) and (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof is provided.
  • the reducing, inhibiting, or ameliorating in the method disclosed herein is in vivo.
  • the reducing, inhibiting, or ameliorating is in vitro.
  • the cells in the method disclosed herein are a cancer cells.
  • the cancer cells are a prostate cancer cells.
  • 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.
  • 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.
  • the prostate cancer cells are cells of a metastatic castration-resistant prostate cancer.
  • the prostate cancer cells are an androgen-dependent prostate cancer cells or an androgen-independent prostate cancer cells.
  • the cancer cells are breast cancer cells.
  • the condition or disease associated with cell proliferation is cancer.
  • 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.
  • the condition or disease is prostate cancer.
  • 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.
  • 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.
  • the prostate cancer is a metastatic castration-resistant prostate cancer.
  • the prostate cancer is an androgen-dependent prostate cancer cells or an androgen-independent prostate cancer.
  • the condition or disease is breast cancer.
  • a method for reducing or preventing tumor growth comprising contacting tumor cells with a therapeutically effective amount of a compound of (I)-(VI) and (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof is provided.
  • 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.
  • the reducing or preventing in the method disclosed herein is in vivo. In another embodiment, the treating is in vitro.
  • the tumor cell in the method disclosed herein is selected from prostate cancer, breast cancer, ovarian cancer, endometrial cancer, or salivary gland carcinoma.
  • the tumor cells are prostate cancer tumor cells.
  • 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.
  • 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.
  • the prostate cancer is a metastatic castration-resistant prostate cancer.
  • the prostate cancer is androgen-dependent prostate cancer or androgen-independent prostate cancer.
  • the tumor cells are is breast cancer tumor cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds provide metabolic stability.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human liver microsomes (HLMs) of less than about 500 ⁇ L/min/mg protein, less than about 450 ⁇ L/min/mg protein, less than about 400 ⁇ L/min/mg protein, less than about 350 ⁇ L/min/mg protein, less than about 300 ⁇ L/min/mg protein, less than about 250 ⁇ L/min/mg protein, less than about 225 ⁇ L/min/mg protein, or less than about 200 ⁇ L/min/mg protein.
  • HLMs human liver microsomes
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than about 200 ⁇ L/min/mg protein, less than about 190 ⁇ L/min/mg protein, less than about 180 ⁇ L/min/mg protein, less than about 170 ⁇ L/min/mg protein, less than about 160 ⁇ L/min/mg protein, less than about 150 ⁇ L/min/mg protein, less than about 140 ⁇ L/min/mg protein, less than about 130 ⁇ L/min/mg protein, less than about 120 ⁇ L/min/mg protein, less than about 110 ⁇ L/min/mg protein, less than about 100 ⁇ L/min/mg
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than about 50 ⁇ L/min/mg protein, less than about 48 ⁇ L/min/mg protein, less than about 45 ⁇ L/min/mg protein, less than about 40 ⁇ L/min/mg protein, less than about 35 ⁇ L/min/mg protein, less than about 30 ⁇ L/min/mg protein, less than about 25 ⁇ L/min/mg protein, less than about 20 ⁇ L/min/mg protein, or less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than about 15 ⁇ L/min/mg protein, less than about 14 ⁇ L/min/mg protein, less than about 13 ⁇ L/min/mg protein, less than about 12 ⁇ L/min/mg protein, less than about 11 ⁇ L/min/mg protein, or less than about 10 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs in the range of about 0.1 ⁇ L/min/mg protein to about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than 48 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs in the range of about 12 ⁇ L/min/mg protein to about 48 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs in the range of about 0.1 ⁇ L/min/mg protein to about 48 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse liver microsomes (MLMs) of less than about 500 ⁇ L/min/mg protein, less than about 450 ⁇ L/min/mg protein, less than about 400 ⁇ L/min/mg protein, less than about 350 ⁇ L/min/mg protein, less than about 300 ⁇ L/min/mg protein, less than about 250 ⁇ L/min/mg protein, less than about 225 ⁇ L/min/mg protein, or less than about 200 ⁇ L/min/mg protein.
  • MLMs mouse liver microsomes
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MLMs of less than about 200 ⁇ L/min/mg protein, less than about 190 ⁇ L/min/mg protein, less than about 180 ⁇ L/min/mg protein, less than about 170 ⁇ L/min/mg protein, less than about 160 ⁇ L/min/mg protein, less than about 150 ⁇ L/min/mg protein, less than about 140 ⁇ L/min/mg protein, less than about 130 ⁇ L/min/mg protein, less than about 120 ⁇ L/min/mg protein, less than about 110 ⁇ L/min/mg protein, less than about 100 ⁇ L/min/mg
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MLMs of less than about 50 ⁇ L/min/mg protein, less than about 48 ⁇ L/min/mg protein, less than about 45 ⁇ L/min/mg protein, less than about 40 ⁇ L/min/mg protein, less than about 35 ⁇ L/min/mg protein, less than about 30 ⁇ L/min/mg protein, less than about 25 ⁇ L/min/mg protein, less than about 20 ⁇ L/min/mg protein, or less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MLMs of less than about 15 ⁇ L/min/mg protein, less than about 14 ⁇ L/min/mg protein, less than about 13 ⁇ L/min/mg protein, less than about 12 ⁇ L/min/mg protein, less than about 11 ⁇ L/min/mg protein, or less than about 10 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs in the range of about 0.1 ⁇ L/min/mg protein to about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 48 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs in the range of about 12 ⁇ L/min/mg protein to about 48 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs in the range of about 0.1 ⁇ L/min/mg protein to about 48 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human hepatocytes (HHs) of less than about 500 ⁇ L/min/million cells, less than about 450 ⁇ L/min/million cells, less than about 400 ⁇ L/min/million cells, less than about 350 ⁇ L/min/million cells, less than about 300 ⁇ L/min/million cells, less than about 250 ⁇ L/min/million cells, less than about 225 ⁇ L/min/million cells, or less than about 200 ⁇ L/min/million cells.
  • HHs human hepatocytes
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 200 ⁇ L/min/million cells, less than about 190 ⁇ L/min/million cells, less than about 180 ⁇ L/min/million cells, less than about 170 ⁇ L/min/million cells, less than about 160 ⁇ L/min/million cells, less than about 150 ⁇ L/min/million cells, less than about 140 ⁇ L/min/million cells, less than about 130 ⁇ L/min/million cells, less than about 120 ⁇ L/min/million cells, less than about 110 ⁇ L/min/million cells, less than about 100 ⁇ L/min/million cells, less than about 90 ⁇ L/min
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 50 ⁇ L/min/million cells, less than about 48 ⁇ L/min/million cells, less than about 45 ⁇ L/min/million cells, less than about 40 ⁇ L/min/million cells, less than about 35 ⁇ L/min/million cells, less than about 30 ⁇ L/min/million cells, less than about 25 ⁇ L/min/million cells, less than about 20 ⁇ L/min/million cells, or less than about 15 ⁇ L/min/million cells.
  • the compounds of the present disclosure have an in vitro metabolic clearance in HHs of less than about 15 ⁇ L/min/million cells, less than about 14 ⁇ L/min/million cells, less than about 13 ⁇ L/min/million cells, less than about 12 ⁇ L/min/million cells, less than about 11 ⁇ L/min/million cells, less than about 10 ⁇ L/min/million cells, less than about 9 ⁇ L/min/million cells, less than about 8 ⁇ L/min/million cells, less than about 8 ⁇ L/min/million cells, less than about 6 ⁇ L/min/million cells, or less than about 5 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs in the range of about 0.1 ⁇ L/min/million cells to about 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 18 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs in the range of about 4 ⁇ L/min/million cells to about 18 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs in the range of about 0.1 ⁇ L/min/million cells to about 18 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 10 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 5 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 10 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 5 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human hepatocytes of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes (MHs) of less than about 500 ⁇ L/min/million cells, less than about 450 ⁇ L/min/million cells, less than about 400 ⁇ L/min/million cells, less than about 350 ⁇ L/min/million cells, less than about 300 ⁇ L/min/million cells, less than about 250 ⁇ L/min/million cells, less than about 225 ⁇ L/min/million cells, or less than about 200 ⁇ L/min/million cells.
  • MHs mouse hepatocytes
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 200 ⁇ L/min/million cells, less than about 190 ⁇ L/min/million cells, less than about 180 ⁇ L/min/million cells, less than about 170 ⁇ L/min/million cells, less than about 160 ⁇ L/min/million cells, less than about 150 ⁇ L/min/million cells, less than about 140 ⁇ L/min/million cells, less than about 130 ⁇ L/min/million cells, less than about 120 ⁇ L/min/million cells, less than about 110 ⁇ L/min/million cells, less than about 100 ⁇ L/min/million cells, less than about 90 ⁇ L/min
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 50 ⁇ L/min/million cells, less than about 48 ⁇ L/min/million cells, less than about 45 ⁇ L/min/million cells, less than about 40 ⁇ L/min/million cells, less than about 35 ⁇ L/min/million cells, less than about 30 ⁇ L/min/million cells, less than about 25 ⁇ L/min/million cells, less than about 20 ⁇ L/min/million cells, or less than about 15 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 15 ⁇ L/min/million cells, less than about 14 ⁇ L/min/million cells, less than about 13 ⁇ L/min/million cells, less than about 12 ⁇ L/min/million cells, less than about 11 ⁇ L/min/million cells, less than about 10 ⁇ L/min/million cells, less than about 9 ⁇ L/min/million cells, less than about 8 ⁇ L/min/million cells, less than about 8 ⁇ L/min/million cells, less than about 6 ⁇ L/min/million cells, or less than about 5 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of about 0.1 ⁇ L/min/million cells to about 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 18 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of about 4 ⁇ L/min/million cells to about 18 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of about 0.1 ⁇ L/min/million cells to about 18 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 17 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 7 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 17 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 7 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compounds with a combination of in vitro metabolic clearance in HLM, MLM, HHs, and MHs as described herein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 17 ⁇ L/min/million cells and a an in vitro metabolic clearance in human hepatocytes of less than about 10 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 7 ⁇ L/min/million cells, and an in vitro metabolic clearance in human hepatocytes of less than about 5 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 17 ⁇ L/min/million cells and a an in vitro metabolic clearance in human hepatocytes of less than about 10 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 7 ⁇ L/min/million cells, and an in vitro metabolic clearance in human hepatocytes of less than about 5 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein and an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 17 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 7 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds s have an HLM and MLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM or MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human or mouse hepatocytes of less than about 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM or MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human or mouse hepatocytes of less than about 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM or MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than about 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than about 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 ⁇ L/min/mg protein and an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 ⁇ L/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 17 ⁇ L/min/million cells.
  • the compounds of the present are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 ⁇ L/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 7 ⁇ L/min/million cells.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 11.55 ⁇ L/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 120 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 110 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 90 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 80 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 70 minutes.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human microsomes greater than about 120 min.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse microsomes greater than about 120 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 120 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 110 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 90 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 80 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 70 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 60 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 50 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 40 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 30 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 20 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 10 minutes.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 360 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 350 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 325 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 310 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 300 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 290 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 250 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 200 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 150 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 80 minutes.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 360 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 350 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 325 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 300 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 250 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 225 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 200 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 150 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 50 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 30 minutes.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 360 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 350 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 325 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 300 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 250 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 225 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 200 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 150 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 50 minutes. In one embodiment, the compounds have an in vitro half-life in rat hepatocytes greater than about 30 minutes. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 80 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 90 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 110 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 115 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 120 minutes.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the mammal microsome is human or mouse microsome.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 50 min to about 200 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 80 min to about 180 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 100 min to about 160 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 110 min to about 150 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 110 min to about 140 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 110 min to about 130 min.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 115 min to about 130 min.
  • the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, having an in vitro half-life stability in mammal microsome in the range of about 115 min to about 130 min.
  • the mammal microsome is human or mouse microsome.
  • the present disclosure provides compounds which demonstrate blocking androgen-induced PSA-luciferase activity (PSA-luciferase assay).
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 4500 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 4000 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 3500 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 3000 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 2500 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 2000 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1500 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 950 nm.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 900 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 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.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 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.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 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.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 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.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 500 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds having in vitro IC 50 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.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 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.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 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.
  • the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 200 nM.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 2000 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 1500 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 1000 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 900 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 850 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 800 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 750 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 700 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 650 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 600 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 550 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 IC 50 in a PSA-luciferase assay of less than about 500 nM.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the mammal microsome is human or mouse microsome.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM and a HLM or MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM and a HLM and MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM and an in vitro metabolic clearance in human and mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM; a HLM or MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 800 nM; a HLM or MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 700 nM; a HLM or MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 1000 nM; a HLM or MLM in vitro metabolic clearance of less than 12 ⁇ L/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 4 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 4 ⁇ L/min/million cells and in vitro IC 50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro metabolic clearance in HHs of less than 4 ⁇ L/min/million cells and in vitro IC 50 in a PSA-luciferase assay of less than about 675 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in HHs of less than about 350 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in HHs of greater than about 320 minutes and in vitro IC 50 in a PSA-luciferase assay of less than about 700 nM.
  • the compounds have an in vitro half-life in HHs of greater than about 350 minutes and in vitro IC 50 in a PSA-luciferase assay of less than about 700 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 15 ⁇ L/min/mg protein.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 15 ⁇ L/min/mg protein and in vitro IC 50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro metabolic clearance in MLMs of less than 15 ⁇ L/min/mg protein and in vitro IC 50 in a PSA-luciferase assay of less than about 675 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 10 ⁇ L/min/million cells. In one embodiment, the compounds have an in vitro metabolic clearance in MHs of less than 8 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 10 ⁇ L/min/million cells and in vitro IC 50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro metabolic clearance in MHs of less than 8 ⁇ L/min/million cells and in vitro IC 50 in a PSA-luciferase assay of less than about 675 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in MLMs of greater than about 100 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in MHs of greater than about 100 minutes. In one embodiment, the compounds have an in vitro half-life in MHs of greater than about 180 minutes.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of 3 ⁇ L/min/million cells to about 20 ⁇ L/min/million cells. In one embodiment, the compounds have an in vitro metabolic clearance in MHs in the range of 3.2 ⁇ L/min/million cells to about 18 ⁇ L/min/million cells. In one embodiment, the compounds have an in vitro metabolic clearance in MHs in the range of 3.4 ⁇ L/min/million cells to about 17 ⁇ L/min/million cells.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than 650 nM. In one embodiment, the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 640 nM. In one embodiment, the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 635 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay in the range of 200 nM to about 700 nM. In one embodiment, the compounds have an in vitro IC 50 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 IC 50 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 IC 50 in a PSA-luciferase assay in the range of about 350 nM to about 675 nM.
  • the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than 525 nM. In one embodiment, the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 500 nM. In one embodiment, the compounds have an in vitro IC 50 in a PSA-luciferase assay of less than about 450 nM.
  • a pharmaceutical composition for modulating androgen receptor (AR) in a subject.
  • a pharmaceutical composition comprises one or more compounds of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof.
  • a pharmaceutical composition comprises a therapeutically effective amounts of one or more compounds of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof.
  • a pharmaceutical composition as described herein, comprises one or more compounds selected from Table A, or a pharmaceutically acceptable salt or solvate thereof. In one embodiment, a pharmaceutical composition as described herein comprise one or more compounds selected from Table B, or a pharmaceutically acceptable salt or solvate thereof.
  • a pharmaceutical composition comprises one or more compounds selected from Compounds A3, A5, A7, A13, A17, A18, A22, A23, A24, A25, A28, A30, A31, A32, A34, A35, A38, A40, A41, A42, A45, A49, A52, A53, A54, A56, A57, A58, A62, A63, A64, A65, A68, A73, A74, A75, A76, B1, B2, B3, or B6, or a pharmaceutically acceptable salt or solvate thereof.
  • a pharmaceutical composition comprises one or more compounds selected from Compounds A1, A2, A4, A6, A8, A9, A10, A11, A12, A14, A15, A16, A19, A20, A21, A26, A27, A29, A33, A36, A37, A39, A43, A44, A46, A47, A48, A50, A51, A55, A59, A60, A61, A66, A67, A69, A70, A71, A72, A77, A78, A79, A80, A81, A82, A83, A84, A85, A86, A87, A88, A89, A90, A91, A92, A93, A94, A95, A96, A97, B4, B5, B7, B8, B9, B10, or B11, or a pharmaceutically acceptable salt or solvate thereof.
  • a pharmaceutical composition comprises one or more compounds selected from Compounds A98-A186. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A187-A211. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A212-A234. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234.
  • a pharmaceutical composition as described herein, comprises one or more compounds selected from Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • a pharmaceutical composition as described herein, comprises one or more compounds selected from Compounds A1-A211 or B1-B11.
  • a pharmaceutical composition as described herein, comprises one or more compounds selected from Compounds A1-A234 or B1-B11.
  • a pharmaceutical composition as described herein, comprising one or more compounds of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof, further comprises one or more additional therapeutically active agents.
  • one or more additional therapeutically active agents are selected from therapeutics useful for treating cancer, neurological disease, a disorder characterized by abnormal accumulation of ⁇ -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.
  • 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
  • PARP
  • a pharmaceutical composition comprising one or more compounds of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient or adjuvant.
  • the pharmaceutically acceptable excipients and adjuvants are added to the composition or formulation for a variety of purposes.
  • a pharmaceutical composition comprising one or more compounds of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof, further comprises a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier includes a pharmaceutically acceptable excipient, binder, and/or diluent.
  • 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.
  • the pharmaceutical compositions of the present disclosure may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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, 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).
  • the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • 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.
  • 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.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof, as disclosed herein, combined with a pharmaceutically acceptable carrier.
  • 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).
  • 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 propellent.
  • Solid carriers suitable for use in the present application include, but are not limited to, inert substances such as lactose, starch, glucose, methyl-cellulose, 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.
  • the carrier can be a finely divided solid which is in admixture with the finely divided active compound.
  • 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/or combination, and may make a pharmaceutical dosage form containing the composition and/or combination easier for the patient and care giver to handle. Diluents for solid compositions and/or combinations 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(r)), potassium chloride, powdered cellulose, sodium chloride, sorbitol, and talc.
  • microcrystalline cellulose e.g., AVICEL
  • microfine cellulose lacto
  • a pharmaceutical composition of the present invention is a solid (e.g., a powder, tablet, a capsule, granulates, and/or aggregates).
  • 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 and/or combinations 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.
  • carbomer e.g., carbopol
  • 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 and/or combination.
  • 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.
  • a disintegrant include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g., AC-DI-SOL and PRIMELLOSE), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.
  • Glidants can be added to improve the flowability of a non-compacted solid composition and/or combination and to improve the accuracy of dosing.
  • Excipients that may function as glidants include colloidal silicon dioxide, magnesium trisilicate, powdered cellulose, starch, talc, and tribasic calcium phosphate.
  • a dosage form such as a tablet
  • 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 and/or combination 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 and/or combination 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.
  • a pharmaceutical composition of the present invention is a liquid (e.g., a suspension, elixir and/or solution).
  • 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)-(VI) and/or (A)-(H-I), 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.
  • a liquid carrier such as water, vegetable oil, alcohol, polyethylene glycol, propylene glycol, or glycerin.
  • 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.
  • polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like.
  • 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 and/or combination an active ingredient or other excipient that is not soluble in the liquid carrier.
  • Emulsifying agents that may be useful in liquid compositions and/or combinations 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.
  • a viscosity enhancing agent 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.
  • a buffer such as guconic acid, lactic acid, citric acid or acetic acid, sodium guconate, sodium lactate, sodium citrate, or sodium acetate.
  • a pharmaceutical composition is prepared for administration by injection (e.g., intravenous, subcutaneous, intramuscular, etc.).
  • 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.
  • other ingredients are included (e.g., ingredients that aid in solubility or serve as preservatives).
  • injectable suspensions are prepared using appropriate liquid carriers, suspending agents and the like.
  • 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.
  • 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.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • 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.
  • the formulations can also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection.
  • 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.
  • the compound can be dispensed in a formulation with an infusion bottle containing sterile pharmaceutical grade water, saline or dextrose/water.
  • 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.
  • 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.
  • a pharmaceutical composition of the present invention comprises a delivery system.
  • 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.
  • certain organic solvents such as dimethylsulfoxide are used.
  • a pharmaceutical composition of the present invention comprises a co-solvent system.
  • co-solvent systems comprise, for example, benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • 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.
  • 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.
  • a pharmaceutical composition of the present invention comprises a sustained-release system.
  • a sustained-release system is a semi-permeable matrix of solid hydrophobic polymers.
  • 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.
  • the route of administration is systemic, e.g., oral or by injection.
  • 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.
  • the route of administration is local, e.g., topical, intra-tumor and peri-tumor.
  • the compound is administered orally.
  • a pharmaceutical composition of the present disclosure is prepared for oral administration.
  • 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, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • such a mixture is optionally ground and auxiliaries are optionally added.
  • pharmaceutical compositions are formed to obtain tablets or dragee cores.
  • disintegrating agents e.g., cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate are added.
  • dragee cores are provided with coatings.
  • 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.
  • 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.
  • pharmaceutical compositions for oral administration are soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • 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.
  • stabilizers may be added.
  • compositions are prepared for buccal administration. Certain of such pharmaceutical compositions are tablets or lozenges formulated in conventional manner.
  • a pharmaceutical composition is prepared for transmucosal administration.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • 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.
  • the dosage unit may be determined with a valve that delivers a metered amount.
  • 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.
  • the compound of the present disclosure are administered by the intravenous route.
  • the parenteral administration may be provided in a bolus or by infusion.
  • 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.
  • a pharmaceutical composition is prepared for topical administration.
  • Certain of such pharmaceutical compositions comprise bland moisturizing bases, such as ointments or creams.
  • ointments or creams include, but are not limited to, petrolatum, petrolatum plus volatile silicones, and lanolin and water in oil emulsions.
  • suitable cream bases include, but are not limited to, cold cream and hydrophilic ointment.
  • 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.
  • one or more compounds of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof are formulated as a prodrug.
  • a prodrug upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically more active form.
  • prodrugs are useful because they are easier to administer than the corresponding active form.
  • a prodrug may be more bioavailable (e.g., through oral administration) than is the corresponding active form.
  • a prodrug may have improved solubility compared to the corresponding active form.
  • prodrugs are less water soluble than the corresponding active form.
  • a prodrug is an ester.
  • the ester is metabolically hydrolyzed to carboxylic acid upon administration.
  • the carboxylic acid containing compound is the corresponding active form.
  • a prodrug comprises a short peptide (polyaminoacid) bound to an acid group.
  • the peptide is cleaved upon administration to form the corresponding active form.
  • 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.
  • the amount of the compound of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof, or compounds disclosed in Tables A and B, 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.
  • 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, 4 th edition, John Wiley & Sons, publisher, New York, 1992 which are incorporated herein by reference in their entirety.
  • 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.
  • 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.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pyrrole Compounds (AREA)
  • Pyridine Compounds (AREA)

Abstract

The present invention relates to compounds of formula (I)-(VI) and/or (A)-(H-I), 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

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 62/747,209, filed Oct. 18, 2018, U.S. Provisional Application No. 62/803,516, filed Feb. 10, 2019, and U.S. Provisional Application No. 62/857,516, filed Jun. 5, 2019, the disclosures of which are herein incorporated by reference in their entirety for all purposes.
  • 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).
  • Clinically available inhibitors of the AR include nonsteroidal antiandrogens such as apalutamide, darolutamide, bicalutamide (Casodex™), nilutamide, flutamide, and enzalutamide. There is also a class of steroidal antiandrogens, such as cyproterone acetate and spironolactone. Both steroidal and non-steroidal antiandrogens target the LBD of the AR and predominantly fail presumably due to poor affinity and mutations that lead to activation of the AR by these same antiandrogens (Taplin, M. E., Bubley, G. J., Kom Y. J., Small E. J., Uptonm M., Rajeshkumarm B., Balkm S. P., Cancer Res., 59, 2511-2515 (1999)), and constitutively active AR splice variants. Antiandrogens have no effect on the constitutively active AR splice variants that lack the ligand-binding domain (LBD) and are associated with castration-recurrent prostate cancer (Dehm S M, Schmidt L J, Heemers H V, Vessella R L, Tindall D J., Cancer Res 68, 5469-77, 2008; Guo Z, Yang X, Sun F, Jiang R, Linn D E, Chen H, Chen H, Kong X, Melamed J, Tepper C G, Kung H J, Brodie A M, Edwards J, Qiu Y., Cancer Res. 69, 2305-13, 2009; Hu et al 2009 Cancer Res. 69, 16-22; Sun et al 2010 J Clin Invest. 2010 120, 2715-30) and resistant to abiraterone and enzalutamide (Antonarakis et al., N Engl J Med. 2014, 371, 1028-38; Scher et al JAMA Oncol. 2016 doi: 10.1001). Conventional therapy has concentrated on androgen-dependent activation of the AR through its C-terminal domain.
  • Other relevant AR antagonists previously reported (see, WO 2010/000066, WO 2011/082487; WO 2011/082488; WO 2012/145330; WO 2015/031984; WO 2016/058080; and WO 2016/058082) that bind to full-length AR and/or truncated AR splice variants that have been recently developed or are currently being developed include: AR degraders such as niclosamide (Liu C et al 2014), galeterone (Njar et al 2015; Yu Z at al 2014), and ARV-330/Androgen receptor PROTAC (Neklesa et al 2016 J Clin Oncol 34 suppl 2S; abstr 267); AR DBD inhibitor VPC-14449 (Dalal K et al 2014 J Biol Chem. 289(38):26417-29; Li H et al 2014 J Med Chem. 57(15):6458-67); antiandrogens apalutamide (Clegg N J et al 2012), ODM-201 (Moilanen A M et al 2015), ODM-204 (Kallio et al J Clin Oncol 2016 vol. 34 no. 2_suppl 230), TAS3681 (Minamiguchi et al 2015 J Clin Oncol 33, suppl 7; abstr 266); and AR NTD inhibitors 3E10-AR441bsAb (Goicochea N L et al 2015), and sintokamide (Sadar et al 2008; Banuelos et al 2016).
  • 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; and WO 2018/045450, and which are hereby incorporated by reference in their entireties.
  • Transcriptionally active androgen receptor plays a major role in CRPC in spite of reduced blood levels of androgen (Karantanos, T. et al Oncogene 2013, 32, 5501-5511; Harris, W. P. et al Nature Clinical Practice Urology, 2009, 6, 76-85). AR mechanisms of resistance to ADT include: overexpression of AR (Visakorpi, T. et al Nature Genetics 1995, 9, 401-406; Koivisto, P. et al Scandinavian Journal of Clinical and Laboratory Investigation Supplementum 1996, 226, 57-63); gain-of-function mutations in the AR LBD (Culig Z. et al Molecular Endocrinology 1993, 7, 1541-1550); intratumoral androgen synthesis (Cai, C. et al Cancer Research 2011, 71, 6503-6513); altered expression and function of AR coactivators (Ueda, T. et al The Journal of Biological Chemistry 2002, 277, 38087-38094; Xu J. et al Nature Reviews Cancer 2009, 9, 615-630); aberrant post-translational modifications of AR (Gioeli D. et al Molecular and Cellular Endocrinology 2012, 352, 70-78; van der Steen T. et al International Journal of Molecular Sciences 2013, 14, 14833-14859); and expression of AR splice variants (AR-Vs) which lack the ligand-binding domain (LBD) (Karantanos, T. et al Oncogene 2013, 32, 5501-5511; Andersen R. J. et al Cancer Cell 2010, 17, 535-546; Myung J. K. et al The Journal of Clinical Investigation 2013, 123, 2948-2960; Sun S. et al The Journal of Clinical Investigation 2010, 120, 2715-2730). Anti-androgens such as bicalutamide and enzalutamide target AR LBD, but have no effect on truncated constitutively active AR-Vs such as AR-V7 (Li Y. et al Cancer Research 2013, 73, 483-489). Expression of AR-V7 is associated with resistance to current hormone therapies (Li Y. et al Cancer Research 2013, 73, 483-489; Antonarakis E. S. et al The New England Journal of Medicine 2014, 371, 1028-1038).
  • 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 (IIIA):
  • Figure US20200123117A1-20200423-C00001
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, 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)—, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 R8b 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 a 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;
      • 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 a 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 comprising the structure of formula (IVA):
  • Figure US20200123117A1-20200423-C00002
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y and Z are each independently a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2— and L is halogen, —NH2, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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 compound of formula (IVA), C is 5- to 10-membered heteroaryl or aryl. In some embodiments, C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member. In some embodiments, C, which is substituted with (R3)n3, is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl. In some embodiments, C, which is substituted with (R3)n3, is selected from
  • Figure US20200123117A1-20200423-C00003
    Figure US20200123117A1-20200423-C00004
  • wherein R3a is C1-C3 alkyl.
  • In one embodiment of the compound of formula (IVA), R1 and R2 are each independently C1, —CN, —CF3, —OH, methyl, methoxy, or —CONH2.
  • In one embodiment of the compound of formula (IVA),
      • A and B are phenyl;
      • X is —(CR5R6)t—;
      • Y and Z are each —O—;
      • V is —CH2— or —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, or optionally substituted C1-C6 alkyl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl; and
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.
  • In one embodiment of the compound of formula (IVA),
      • A and B are phenyl;
      • X is —(CR5R6)t—;
      • W is —CH2— or —C(CH3)H—;
      • Y and Z are each —O—;
      • V is —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, or —CN;
      • R5 and R6 are each independently hydrogen, or C1-C3 alkyl; and
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (A-I):
  • Figure US20200123117A1-20200423-C00005
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a phenyl or a 5- to 7-membered monocyclic heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2— and L is halogen, —NH2, or —CF3; or
      • V is —CH2CH2— and L is halogen or —NH2;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from —CN, C1-C3 alkoxy, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • 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 compound of formula (IVA),
  • In one embodiment of the compound of formula (A-I), at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl).
  • In one embodiment of the compound of formula (A-I),
      • X is a bond or —(CR5R6)t;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • Y is —O—;
      • Z is —O—;
      • V is —CH2— or —CH2CH2—; and
      • L is halogen
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (G-II):
  • Figure US20200123117A1-20200423-C00006
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00007
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or methyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1 or 2; and
      • t is 1.
  • In one embodiment of the compound of formula (G-II), at least one R3 is selected from —NHSO2CH3, —NHSO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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, the present disclosure provides Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment, the present disclosure provides Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • In one embodiment, the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (IIIA), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier. In one embodiment, the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (IVA), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier. In one embodiment, the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (A-I), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier. In one embodiment, the present disclosure provides a pharmaceutical composition comprising any one of the compound of formula (G-II), or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • In one embodiment, the present disclosure provides a pharmaceutical composition comprising any one of Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier. In one embodiment, the present disclosure provides a pharmaceutical composition comprising any one of Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, and a pharmaceutically acceptable carrier.
  • In one embodiment, the present disclosure provides a method for treating cancer, comprising administering any one of the compound of formula (IIIA), (IVA), (A-I), or (G-II), and Compounds A1-A96, A98-A116, A118-A159, A161-A175, or A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, to a subject in need thereof. In one embodiment, the cancer is prostate cancer. In one embodiment, the prostate cancer is metastatic castration-resistant prostate cancer. In other embodiments, the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.
  • In one embodiment, the present disclosure provides a method for treating cancer, comprising administering any one of Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows a dose-dependent curve of PSA-luciferase activities in response to representative compounds in transiently transfected LNCaP cells treated with synthetic androgen (R1881).
  • FIG. 2A shows the percent PSA-luciferase activities of representative compounds in LNCaP cells transiently transfected with the PSA (6.1 kb)-luciferase reporter and treated without or with R1881. FIG. 2B shows the percent PSA-luciferase activities of representative compounds in LNCaP cells co-transfected with an expression vector for AR-V7 and the PSA-luciferase reporter and treated without or with R1881.
  • FIG. 3 shows concentration of representative compounds in plasma of male CD-1 mice after a single dose PO (oral) dose.
  • FIG. 4 shows change in tumor volume in male NCG mice bearing LNCaP tumors after oral administration of representative compounds.
  • FIG. 5 shows change in % body weight in male NCG mice bearing LNCaP tumors after oral administration of representative compounds.
  • FIG. 6 shows individual tumor volume change from baseline measured at the end of experiment for oral administration of representative compounds to male NCG mice bearing LNCaP tumors.
  • FIG. 7 shows concentration dependent effects on cell proliferation of LNCaP, PC3, and LNCaP95 cells treated with Compound A13.
  • 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), (IA), (IB), (IC), (II), (IIA), (IIIA), (IIB), (III), (IV), (V), (VA), (VI), (A), (A-I), (B)-(D), (E), (E-I)-(E-VII), (F), (G), (G-I), (G-II), (H), and (H-I), 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, 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 alkenyl include ethynyl, propynyl, butynyl, pentynyl and the like. Unless stated otherwise specifically in the specification, an alkyl 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 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 alkyl carbonyl 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 haloalkenyl 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. Heterocyclyl 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
  • Figure US20200123117A1-20200423-C00008
  • (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,
  • Figure US20200123117A1-20200423-C00009
  • 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
  • Figure US20200123117A1-20200423-C00010
  • 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.
  • “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):
  • Figure US20200123117A1-20200423-C00011
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, —NR7—, —N(R′)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—, —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, —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, 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, 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 R8b 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 (R1 and R12) or (R14 and R15) taken together form an optionally substituted heterocyclyl;
      • each m is independently 0, 1 or 2;
      • 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 the present disclosure provides compounds comprising the structure of formula (IA):
  • Figure US20200123117A1-20200423-C00012
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-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—, —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, —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, 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, —NR14COOR16, —NR14COR16, —NR14CONR14R15, 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;
      • 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 R8b 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, C1-C6 haloalkyl, C2-C6 haloalkenyl, C2-C6 haloalkynyl, optionally substituted carbocyclyl, optionally substituted —CO(C1-C6 alkyl), —CO(optionally substituted heterocyclyl), 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 —COO(C1-C6 alkyl), 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 or 2;
      • 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 the present disclosure provides compounds comprising the structure of formula (IB):
  • Figure US20200123117A1-20200423-C00013
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, —NR7—, —N(R7)CO—, —CON(R7)—, or —NSO2R7—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —(CR8aR9a)m—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10; —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, deuterium, 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, —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, 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, —NR14COOR16, —NR14COR16, —NR14CONR14R15, 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;
      • 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, 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 R8b 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 C1-C6 alkyl-NH2, C1-C6 haloalkyl, C2-C6 haloalkenyl, C2-C6 haloalkynyl, optionally substituted carbocyclyl, optionally substituted —CO(C1-C6 alkyl), —CO(optionally substituted heterocyclyl), 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 —COO(C1-C6 alkyl), optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or (R11 and R12) or (R14 and R15) or (R14 and R16) taken together form an optionally substituted heterocyclyl;
      • each m is independently 0, 1 or 2;
      • 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 the present disclosure provides compounds comprising the structure of formula (IC)
  • Figure US20200123117A1-20200423-C00014
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, —NR7—, —N(R7)CO—, —CON(R7)—, or —NSO2R7—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —(CR8aR9a)m—, —C(═O)—, N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CCl2R10, —CCl3, —CN, —OR10; —NR11R12 or —CONR11R12;
      • R1 and R2 are each independently hydrogen, deuterium, 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, —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, 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, —NR14COOR16, —NR14COR16, —NR14CONR14R15, 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;
      • 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, —CONR13R14, —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, —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 R8b 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 C1-C6 alkyl-NH2, C1-C6 haloalkyl, C2-C6 haloalkenyl, C2-C6 haloalkynyl, optionally substituted carbocyclyl, optionally substituted —CO(C1-C6 alkyl), —CO(optionally substituted heterocyclyl), 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 —COO(C1-C6 alkyl), optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or (R11 and R12) or (R14 and R15) or (R14 and R16) taken together form an optionally substituted heterocyclyl;
      • each m is independently 0, 1 or 2;
      • 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, the present disclosure provides compounds comprising the structure of formula (II):
  • Figure US20200123117A1-20200423-C00015
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —NHCO—, —N(C1-C3 alkyl)CO—, or —CONH—, or —CON(C1-C3 alkyl)-;
      • V is a bond, —(CR8aR9a)m—, —C(═O)—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • L is hydrogen, halogen, —CF2R1, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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;
      • 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 R8b taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R11, R12, 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 a 3- to 6-membered heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 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, the present disclosure provides compounds comprising the structure of formula (IIA):
  • Figure US20200123117A1-20200423-C00016
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —CH2—, —C(CH3)H—, —NHCO—, —N(C1-C3 alkyl)CO—, or —CONH—, or —CON(C1-C3 alkyl)-;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10; —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, deuterium, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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)-SO2NR4R5, optionally substituted —SO2R16 or optionally substituted —(C1-C6 alkyl)-SO2R16;
      • R3 is selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R11, R12, 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 a 3- to 6-membered heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl or C1-C6 alkyl-NH2; or R14 and R16 taken together form a 3- to 6-membered heterocyclyl;
      • 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, the present disclosure provides compounds comprising the structure of formula (IIB):
  • Figure US20200123117A1-20200423-C00017
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is a bond, —(CR8aR9a)m—, —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, deuterium, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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;
      • 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 R8b taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R11, R12, 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 a 3- to 6-membered heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl or C1-C6 alkyl-NH2; or R14 and R16 taken together form a 3- to 6-membered heterocyclyl;
      • 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 comprising the structure of formula (III):
  • Figure US20200123117A1-20200423-C00018
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —(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—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 R8b 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or 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, the present disclosure provides compounds comprising the structure of formula (IIIA):
  • Figure US20200123117A1-20200423-C00019
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, 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)—, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 R8b 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 a 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 comprising the structure of formula (IV):
  • Figure US20200123117A1-20200423-C00020
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y and Z are each independently a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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 (V):
  • Figure US20200123117A1-20200423-C00021
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 5- to 10-membered heteroaryl or aryl;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, halogen, oxo, ═S, —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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is H or C1-C6 alkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl; or R14 and R15 taken together form a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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 (VA):
  • Figure US20200123117A1-20200423-C00022
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 5- to 10-membered heteroaryl or aryl;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —OH, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, halogen, oxo, ═S, —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, —NR14CONR14R15, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, —NH2, 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 H, C1-C6 alkyl, —CO(C1-C6 alkyl);
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, or —COO(C1-C6 alkyl); or R14 and R15 taken together form a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C1-C3 haloalkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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 (VI):
  • Figure US20200123117A1-20200423-C00023
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 5- to 10-membered heterocyclyl;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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):
  • Figure US20200123117A1-20200423-C00024
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a phenyl or a 5- to 7-membered monocyclic heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • 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 (B)
  • Figure US20200123117A1-20200423-C00025
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a 5- to 7-membered saturated or partially saturated monocyclic heterocycle comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • R16 is hydrogen or C1-C3 alkyl;
      • 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), (IC), (II), (III), (IIIA), (IV), (V), (VI), (A), or (B), —V-L is —CH2CH2C1, —CH2CH2CH2C1, —CH2CH2NH2, or —CH2CH2CH2NH2.
  • In one embodiment of the compounds of formula (I), (IC), (II), (III), (IIIA), (IV), (V), (VI), (A), or (B), —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), (IC), (II), (III), (IIIA), (IV), (V), (VI), (A), or (B), X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—.
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (C):
  • Figure US20200123117A1-20200423-C00026
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • X is a bond, (CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • D is —NH or —NR3;
      • U is each independently O, S, or NR16;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, halogen, —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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, 2, or 3; and
      • t is 0, 1 or 2.
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (D):
  • Figure US20200123117A1-20200423-C00027
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a 5- or 6-membered heteroaryl comprising 1 or 2 heteroatoms selected from O, S, or N as a ring member;
      • X is —(CR5R6)t— or —NR7—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —NH2, or —CF3;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • 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 (E):
  • Figure US20200123117A1-20200423-C00028
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00029
      • X is —(CR5R6)t— or —NR7—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is hydrogen or halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, or 2; and
      • t is 1 or 2.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-I):
  • Figure US20200123117A1-20200423-C00030
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00031
      • X is —(CR5R6)t— or —NR7—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, —CH2CH2CH2—, or —CH2CHClCH2—;
      • L is hydrogen, —OH, or halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, or 2; and
      • t is 1 or 2.
  • In one embodiment of the compounds of formula (A)-(C) or (E-1), R3 is selected from hydrogen, 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, the present disclosure provides compounds comprising the structure of formula (F):
  • Figure US20200123117A1-20200423-C00032
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00033
      • X is —(CR5R6)t— or —NR7—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is hydrogen or halogen;
      • R1 and R2 are each independently halogen or —CN;
      • R3 is selected from —NHSO2CH3, —N(CH3)SO2CH3, or —SO2CH3;
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, or 2; and
      • t is 1 or 2
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (G):
  • Figure US20200123117A1-20200423-C00034
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00035
      • X is —(CR5R6)t— or —NR7—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2— and L is hydrogen;
      • or alternatively, V is —CH2CH2— or —CH2CH2CH2—, and L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • R3 is selected from —NHSO2CH3, —N(CH3)SO2CH3, or —SO2CH3;
      • R5 and R6 are each independently hydrogen or methyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1, or 2; and
      • t is 1.
  • In one embodiment the present disclosure provides compounds comprising the structure of (G-I)
  • Figure US20200123117A1-20200423-C00036
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00037
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2CH2— or —CH2CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • R3 is selected from —NHSO2CH3, —N(CH3)SO2CH3, or —SO2CH3;
      • R5 and R6 are each independently hydrogen or methyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1 or 2; and
      • t is 1.
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (G-II)
  • Figure US20200123117A1-20200423-C00038
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00039
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or methyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1 or 2; and
      • t is 1.
  • In one embodiment, the present disclosure provides compounds comprising the structure of formula (H):
  • Figure US20200123117A1-20200423-C00040
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C—I is
  • Figure US20200123117A1-20200423-C00041
      • X is —(CR5R6)t— or —NR7—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2—, —CH2CH2— or —CH2CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • R5 and R6 are each independently hydrogen or methyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • t is 1.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (H-I):
  • Figure US20200123117A1-20200423-C00042
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C—I is
  • Figure US20200123117A1-20200423-C00043
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2—, —CH2CH2— or —CH2CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • R5 and R6 are each independently hydrogen or methyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • t is 1.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-II):
  • Figure US20200123117A1-20200423-C00044
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00045
      • X is —(CR5R6)—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2;
      • wherein when C—R3 is
  • Figure US20200123117A1-20200423-C00046
  • R2A and R2B are not both Cl.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-III):
  • Figure US20200123117A1-20200423-C00047
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00048
      • X is —(CR5R6)—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2;
      • wherein when C—R3 is
  • Figure US20200123117A1-20200423-C00049
  • and one of R2A and R2B is Cl, then the other of R2A and R2B is not —CN.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-IV):
  • Figure US20200123117A1-20200423-C00050
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00051
      • X is —(CR5R6)—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-V):
  • Figure US20200123117A1-20200423-C00052
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00053
      • X is —(CR5R6)—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2;
      • wherein when C—R3 is
  • Figure US20200123117A1-20200423-C00054
  • R2A and R2B are not both Cl.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-VI):
  • Figure US20200123117A1-20200423-C00055
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00056
      • X is —(CR5R6)—,
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2;
  • Figure US20200123117A1-20200423-C00057
      • wherein when C—R3 is N and one of R2A and R2B is Cl, then the other of R2A and R2B is not —CN.
  • In one embodiment the present disclosure provides compounds comprising the structure of formula (E-VII):
  • Figure US20200123117A1-20200423-C00058
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
  • C is
  • Figure US20200123117A1-20200423-C00059
      • X is —(CR5R6)—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2.
  • In one embodiment of the compounds of formula (I), (IA), (IB), (IC), (II), (IIA), (IIB), (III), (IIIA), (IV), (V), (VA), or (VI) (denoted as formula “(I)-(VI)”), A and B are each independently 5- or 6-membered aryl or heteroaryl. In one embodiment, A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene. In one embodiment, A and B are each phenyl.
  • In another embodiment, A has a meta or para connectivity with X and Y. In some embodiments, B has a meta or para connectivity with X and Z.
  • In one embodiment of the compounds of formula (I)-(VI), A and B are phenyl and has one of the connectivity as shown:
  • Figure US20200123117A1-20200423-C00060
  • In one embodiment of the compounds of formula (I)-(VA) and (A) (e.g., formula (I), (IA), (IB), (IC), (II), (IIA), (IIB), (III), (IIIA), (IV), (V) (VA), and (A)), C is aryl or heteroaryl. In some embodiments, C is 5- to 10-membered aryl or heteroaryl. In other embodiments, C is aryl. In some embodiments, C is phenyl or naphthyl. In other embodiments, C is aryl. In some embodiments, C is phenyl.
  • In one embodiment of the compounds of formula (I)-(VA) and (A), C is heteroaryl. In one embodiment, C monocyclic or bicyclic heteroaryl. In another embodiment, C is monocyclic heteroaryl. In some embodiments, C is 5- or 10-membered heteroaryl. In some embodiments, C is 5- or 6-membered heteroaryl, which is optionally substituted with 1, 2, 3, 4, or 5 R3. In some embodiments, C is 5- or 6-membered heteroaryl containing 1, 2, or 3 heteroatoms selected from O, S, or N, wherein the heteroaryl is optionally substituted with 1, 2, 3, 4, or 5 R3. In some embodiments, C is 5- or 6-membered heteroaryl containing 1 or 2 heteroatoms selected from O, S, or N, wherein the heteroaryl is optionally substituted with 1, 2, 3, 4, or 5 R3.
  • In one embodiment of the compounds of formula (I)-(VA), (A), or (D), C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, or pyrimidyl, which are each optionally substituted with 1, 2, 3, 4, or 5 R3. In one embodiment, C, which is substituted with (R3)n3, is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl. In one embodiment, C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl, which are each substituted with 1, 2, 3, 4, or 5 R3.
  • In one embodiment of the compounds of formula (I)-(VA), (A), or (D), C is selected from
  • Figure US20200123117A1-20200423-C00061
    Figure US20200123117A1-20200423-C00062
  • wherein R3a is C1-C3 alkyl. In one embodiment of the compounds of formula (I)-(VA), (A) or (D), C is selected from
  • Figure US20200123117A1-20200423-C00063
    Figure US20200123117A1-20200423-C00064
    Figure US20200123117A1-20200423-C00065
  • wherein R3a is C1-C3 alkyl.
  • In one embodiment of the compounds of formula (I)-(VA), (A), or (D), C is
  • Figure US20200123117A1-20200423-C00066
  • In one embodiment, C is
  • Figure US20200123117A1-20200423-C00067
  • or in its tautomeric form
  • Figure US20200123117A1-20200423-C00068
  • In one embodiment, C is
  • Figure US20200123117A1-20200423-C00069
  • or in its tautomeric form
  • Figure US20200123117A1-20200423-C00070
  • In one embodiment of the compounds of formula (I)-(IV), C is heterocyclyl. In one embodiment, C is saturated or partially saturated heterocycle. In some embodiments, C is monocyclic or bicyclic. In some embodiments, C is 5- to 7-membered heterocyclyl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • In one embodiment of the compounds of formula (I)-(VA), (B) and (C), C is imidazolidine, imidazolidine-dione, or dihydrooxazole. In one embodiment, C is selected from
  • Figure US20200123117A1-20200423-C00071
  • In one embodiment of the compounds of formula (I)-(VA), (B) and (C), C is
  • Figure US20200123117A1-20200423-C00072
  • D is —O—, —NH— or —NR3—; and U is each independently O, S, or NR16. In one embodiment, D is —NH— or —NR3—. In some embodiments, at least one U is O. In other embodiments, each U is O. In some embodiments, at least one R3 is —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3.
  • In one embodiment of the compounds of formula (I)-(VA), C is aryl. In some embodiments, C is phenyl or naphthyl. In one embodiment of the compounds of formula (I)-(VA) or (A), C is phenyl.
  • In on embodiment compounds of formula (I)-(VI), C is bicyclic heteroaryl or heterocyclyl. In one embodiment, C is
  • Figure US20200123117A1-20200423-C00073
  • In one embodiment of the compounds of formula (I), Z is a bond, —(CR8R9)m—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—. In one embodiment, Z is —(CR8R9)m—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—. In some embodiments, Z is —C(═O)—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—. In one embodiment, Z is —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—. In some embodiments, Z is a bond, —CH2—, —O—, or —NCH3—. In some embodiments, Z is a bond, —CH2—, —O—, or —NH—. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), Z is —O—. As used herein, “compounds of formula (I)-(IV) and (A)-(H-I)” refers to compounds of formula (I), (IA), (IB), (IC), (II), (IIA), (IIIA), (IIB), (III), (IV), (IVA), (V), (VA), (VI), (A), (A-I), (B)-(D), (E), (E-I)-(E-VII), (F), (G), (G-I), (G-II), (H), and (H-I).
  • In one embodiment of the compounds of formula (I), Y is a bond, —(CR8R9)m—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—. In one embodiment, Y is —(CR8R9)m—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—. In some embodiments, Y is —C(═O)—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—. In one embodiment, Y is —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—. In some embodiments, Y is a bond, —CH2—, —O—, or —NCH3—. In some embodiments, Y is a bond, —CH2—, —O—, or —NH—. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), Y is —O—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), V is a bond, —(CR8aR9a)m—, or —C(═O)—. In some embodiments, V is bond, or —(CR8aR9a)m—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), V is —(CR8aR9a)m—, wherein m is 1, 2, or 3. In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), V is —(CR8aR9a)m—, wherein R8a and R8b are each independently hydrogen, —OH, halogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C1-C3 alkoxy, —NR13R14, optionally substituted —(C1-C3 alkyl)-NR13R14, —NR14COR16, optionally substituted —(C1-C3 alkyl)-NR14COR16, —CONR14R15, or optionally substituted —(C1-C3 alkyl)-CONR14R15; or R8a and R8b taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl. In one embodiment, V is —(CR8aR9a)m—, wherein R8a and R8b 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 R8b, on the same carbon atom or on a different carbon atom, taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl.
  • In one embodiment of the compounds of formula (I)-(IIB) and (VA), V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), V is —CH2—, —CH2CH2—, or —CH2CH2CH2—, each optionally substituted with one or more of —OH, halogen, or C1-C3 alkyl. In other embodiments, V is —CH2—, —CH2CH2—, —CH2CH(OH)CH2— or —CH2CH2CH2—. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), V is —CH2—, —CH2CH2—, or —CH2CH2CH2—. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), V is —CH2— or —CH2CH2—.
  • In some embodiments of the compounds of formula (I)-(VI) and (A)-(D), V is —CH2— and L is halogen, —NH2, or —CF3; or V is —CH2CH2— and L is halogen or —NH2.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), L is hydrogen, halogen, —CF2H, —CF3, —CN, —O(C1-C3 alkyl), —NR11R12, or —CONR11R12. In one embodiment, L is hydrogen, halogen, —CF2H, —CF3, —CN, —O(C1-C3 alkyl), —NH2, —NH(C1-C3 alkyl), —N(C1-C3 alkyl)2, —CONH2, —CONH(C1-C3 alkyl), or —CON(C1-C3 alkyl)2. In some embodiments, L is hydrogen, halogen, —CF3, or —NH2.
  • In some embodiments of the compounds of formula (IC) and (IIIA), L is halogen, —CCl3, —CCl2, —CF3, or —NH2. In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), L is halogen, —CF3, or —NH2. In one embodiment, L is hydrogen or halogen. In one embodiment, L is halogen. In other embodiments, L is Cl, or Br. In one embodiment, L is Cl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), W is a bond. In one embodiment, W is —(CR8aR9a)m—, —C(═O)—, —N(R′)CO—, —CONR7—, or —NSO2R7—. In one embodiment, W is —(CR8aR9a)m—, wherein m is 1, 2, or 3. In some embodiments, W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—, wherein R7 is H or C1-C6 alkyl. In some embodiments, W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—, wherein R7 is H or C1-C3 alkyl. In one embodiment, W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —NHCO—, —N(C1-C3 alkyl)CO—, —CONH—, or —CON(C1-C3 alkyl)-. In one embodiment of the compounds of formula (I)-(VI) and (A)-(E-VI), W is a bond, —CH2—, or —C(CH3)H—. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), W is a —CH2— or —C(CH3)H—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), —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)-(VI) and (A)-(H-I), —Y—W— is —OCH2—, —OCH2CH2—, or —OCH(CH3)—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C),
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—; and
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C),
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—; and
      • L is halogen, —NH2, or —CF3.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), —Z—V-L is —Z—CH2CH2C1, —Z—CH2CH2CH2C1, —Z—CH2CH2NH2, or —Z—CH2CH2CH2NH2, wherein Z is a bond, —O—, —NH—, or —N(COCH3)—. In one embodiment, —Z—V-L is —OCH3.
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), —Z—V-L is —O—CH2CH2Cl or —O—CH2CH2CH2Cl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), —V-L is —CH2CH2C1, —CH2CH2CH2C1, —CH2CH2NH2, or —CH2CH2CH2NH2. In one embodiment, —V-L is —CH3.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—. In one embodiment, X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, or —NR7—, wherein R7 is H or C1-C6 alkyl. In some embodiments, X is a bond, —(CR5R6)t—, or —NR7—. In some embodiments, X is a bond or —(CR5R6)t—. In some embodiments, X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, —CH2CH2—, —NH—, or —N(C1-C6 alkyl)-. In some embodiments, X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, —CH2CH2—, —NH—, —N(CH3)—, —N(CH2CH3)—, —N(iPr)—, or —N(tBu)-. In some embodiments, X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—.
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), X is —CH2—, —C(CH3)H—, or —C(CH3)2-. In one embodiment, X is —C(CH3)2-.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), R1 and R2 are each independently halogen, —CN, —CF3, —OH, C1-C3 alkyl, 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-C3 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2R16, —(C1-C3 alkyl)-SO2R16, 3- to 7-membered carbocyclyl, 3- to 7-membered heterocyclyl, phenyl, or 5- to 6-membered heteroaryl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), 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, —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, or optionally substituted —(C1-C6 alkyl)-SO2R16. In one embodiment, R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, optionally substituted C1-C3 alkyl, C1-C3 alkoxy, optionally substituted —(C1-C3 alkyl)-(C1-C3 alkoxy), optionally substituted —(C1-C3 alkyl)-OH, —NR13R14, —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, optionally substituted —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C3 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C3 alkyl)-SO2NR14R15, —SO2R16, or optionally substituted —(C1-C3 alkyl)-SO2R16. In one embodiment, R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, C1-C3 alkyl, 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-C3 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2R16, or —(C1-C3 alkyl)-SO2R16.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, C1-C3 alkyl, or —CONR14R15. In some embodiments, R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, methyl, methoxy, or —CONH2. In one embodiment, R1 and R2 are each independently hydrogen, C1, —CN, —CF3, —OH, methyl, methoxy, or —CONH2. In one embodiment, R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, or methyl. In one embodiment, R1 and R2 are each independently C1, —CN, —CF3, —OH, methyl, methoxy, or —CONH2. In one embodiment of the compounds of formula (I)-(VI), R1 and R2 are each independently halogen, —CN, —CF3, —OH, or methyl.
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(E-I), R1 and R2 are each halogen, methyl, —CF3, or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(F), R1 and R2 are each halogen or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), at least one of R1 and R2 is Cl or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), at least two of R1 and R2 are each independently Cl or —CN. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), R1 and R2 are each Cl or —CN.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), R1 and R2 are each independently optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In one embodiment, R1 and R2 are each independently 3- to 7-membered carbocyclyl, 3- to 7-membered heterocyclyl, phenyl, or 5- to 6-membered heteroaryl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R1 have one of the connectivity as shown below with respect to X and Y:
  • Figure US20200123117A1-20200423-C00074
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R2 have one of the connectivity as shown below with respect to X and Z:
  • Figure US20200123117A1-20200423-C00075
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), 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)-(VI), (A), (A-I), (B), and (C), 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 (I)-(VI), (A), (A-I), (B), and (C), 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)-(VI), (A), (A-I), (B), and (C), R3 is selected from 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, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments, R3 is selected from 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, or optionally substituted —(C1-C6 alkyl)-SO2R16. In another embodiment, R3 is hydrogen, halogen, oxo, ═S, ═NR16, —CN, —CF3, —OH, —SR16, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, optionally substituted C1-C3 alkoxy, optionally substituted —(C1-C3 alkyl)-(C1-C3 alkoxy), optionally substituted —(C1-C3 alkyl)-OH, —NR13R14, optionally substituted —(C1-C3 alkyl)-NR13R14, —NR14SO2R16, optionally substituted —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, optionally substituted —(C1-C3 alkyl)-NR14COR16, —CONR14R15, optionally substituted —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, optionally substituted —(C1-C3 alkyl)-SO2NR14R15, optionally substituted —SO2R16, or optionally substituted —(C1-C3 alkyl)-SO2R16.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R3 is selected from —NR14SO2R16, optionally substituted —(C1-C6 alkyl)NR14SO2R16, or optionally substituted —SO2R16; wherein R16 is hydrogen, optionally substituted C1-C3 alkyl, optionally substituted C2-C3 alkenyl, optionally substituted C2-C3 alkynyl, C3-C6 cycloalkyl, or phenyl. In one embodiment, R3 is selected from —NR14SO2R16, —(C1-C6 alkyl)NR14SO2R16, or —SO2R16; wherein R16 is hydrogen, C1-C3 alkyl, —(C1-C3 alkyl)-NH2, C3-C6 cycloalkyl, or phenyl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R3 is selected from hydrogen, halogen, oxo, ═S, ═NR16, —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 some embodiments, R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —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), or —N(CH3)CO(C1-C3 alkyl). In some embodiments, R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl). In one embodiment, R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, methyl, —SCH3, —SO2CH3, —NHSO2CH3, —NHSO2CH2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3. In one embodiment, R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, methyl, —SCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3. In some embodiments, R3 is selected from F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl). In some embodiments, R3 is selected from F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —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), or —N(CH3)CO(C1-C3 alkyl). In one embodiment, R3 is selected from F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, methyl, —SCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3. In another embodiment, R3 is —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3. In one embodiment, R3 is oxo, ═S, ═NR16, C1-C3 alkyl, —SO2(C1-C3 alkyl), or —NHSO2(C1-C3 alkyl). In one embodiment, at least one of R3 is oxo, ═S, or ═NR16. In one embodiment, at least one of R3 is oxo, ═S, or ═NR16, wherein R16 is H or C1-C3 alkyl.
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(E-VII), R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(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), or —N(CH3)COO(C1-C3 alkyl). In one embodiment of the compounds of formula (I)-(VI) and (A)-(E-VII), R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl).
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R3 on a sp3 carbon is each selected from hydrogen, halogen, oxo, ═S, ═NR16, —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). When R3 on a sp3 carbon is oxo, ═S, or ═NR16, the carbon becomes sp2.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R3 on a sp2 carbon is each selected from hydrogen, 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 one embodiment of the compounds of formula (I)-(VI), (A), (A-I), (B), and (C), R3 on a nitrogen atom is each selected from C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —(C1-C3 alkyl)-(C1-C3 alkoxy), —(C1-C3 alkyl)-OH, —(C1-C3 alkyl)-NR13R14, —(C1-C3 alkyl)NR14SO2R16, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —(C1-C3 alkyl)-SO2NR14R15, or —(C1-C6 alkyl)-SO2(C1-C3 alkyl).
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(G-II), at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl). In one embodiment, at least one R3 is selected from —NHSO2CH3, —NHSO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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)-(VI) and (A)-(E-II), R3 is not hydrogen.
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(G-II), at least one R3 is —SO2CH3, —NHSO2CH3, —NCH3SO2CH3, —NHSO2CH2CH3, or —N(CH3)SO2CH2CH3. In one embodiment of the compounds of formula (I)-(VI) and (A)-(G-II), at least one R3 is —SO2CH3, —NHSO2CH3, or —NCH3SO2CH3.
  • In one embodiment the compounds of formula (I), (IA), (IB), or (IC), R3 is heterocyclyl.
  • In one embodiment, R3 is heterocyclyl selected from
  • Figure US20200123117A1-20200423-C00076
  • In one embodiment of the compounds of formula (IB), (IC), (IIA), or (IIB), 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), (IA), (IB), or (IC), 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)-(VI), (A), (B), and (C), 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)-(VI) and (A)-(H-I), R5 and R6 are each H or methyl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), 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)-(VI) and (A)-(H-I), R7 is hydrogen or C1-C3 alkyl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), 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 R8b 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 R8b taken together 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 (IB), (IC), (III), or (IIIA), R8a and R9a are not —OH. In one embodiment, R8a and R9a are not —OH.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), 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)-(VI), (A), (B), and (C), R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl.
  • In one embodiment of the compounds of formula (I)-(IIB), 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. In some embodiments, R10 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, R10 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl. In some embodiments, R10 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl. In some embodiments, R10 is hydrogen or C1-C3 alkyl.
  • In one embodiment of the compounds of formula (I)-(IIB), 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. In one embodiment, R11 and R12 are each independently hydrogen, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, or optionally substituted C2-C6 alkynyl. In some embodiments. R11 and R12 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl. In some embodiments. R11 and R12 are each independently hydrogen or C1-C3 alkyl.
  • In one embodiment of the compounds of formula (I)-(IIB), R11 and R12 taken together form an optionally substituted heterocyclyl. In one embodiment, R11 and R12 taken together form an optionally substituted 3- to 7-membered heterocyclyl. In other embodiments, R11 and R12 taken together form 3- to 7-membered heterocyclyl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), 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)-(VI), (A), (B), and (C), 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)-(VI), (A), (B), and (C), 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 other embodiments, R14 and R15 taken together form 3- to 7-membered heterocyclyl.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), 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)-(IIIA), m is 1 or 2.
  • In one embodiment of the compounds of formula (I)-(VI) and (A)-(F), t is 1 or 2. In one embodiment of the compounds of formula (I)-(VI) and (A)-(H-I), t is 1.
  • In one embodiment of the compounds of formula (I)-(VI), (A), (B), and (C), 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 selected from halogen, —CN, —CF3, —OH, C1-C3 alkyl, C1-C3 alkoxy, —NH2, —SCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3.
  • In one embodiment of the compounds of formula (I)-(VI), A and B are each monocyclic ring.
  • In one embodiment of the compounds of formula (I)-(VI), B is phenyl, pyridyl, or pyrimidyl.
  • In one embodiment of the compounds of formula (I)-(IIIA), Z and V are not both a bond.
  • In one embodiment of the compounds of formula (I)-(VI), (A)-(C), Y and W are not both a bond.
  • In one embodiment of the compounds of formula (I)-(VI), C is a 4- to 10-membered ring.
  • In one embodiment of the compounds of formula (D)-(H-I), 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 of the compounds of formula (D)-(H), X is —NR7—. In one embodiment, X is —NH—, —N(CH3)—, —N(CH2CH3)—, —N(iPr)—, or —N(tBu)-.
  • In one embodiment of the compounds of formula (D)-(H-I), Y is —O—. In one embodiment of the compounds of formula (D)-(H-I), Z is —O—. In one embodiment of the compounds of formula (D)-(H-I), Y and Z are both —O—.
  • In one embodiment of the compounds of formula (D)-(H-I), —V-L is CH2CH2C1, —CH2CH2CH2C1, or —CH3. In some embodiments, —V-L is CH2CH2Cl or —CH2CH2CH2Cl.
  • In one embodiment of the compounds of formula (D)-(H-I), n1 is 0.
  • In one embodiment of the compounds of formula (D)-(H-I), n2 is 0, 1, or 2. In some embodiments, n2 is 2. In some embodiments, n2 is 2 and R2 are each ortho to Z. In other embodiments, n2 is 2 and R2 are each ortho to Z, wherein R2 is halogen or —CN.
  • In one embodiment of the compound of formula (I)-(VI) and (A)-(H-I), the compound can be a stereoisomer. For example, if X is —(CR5R6)— and R5 and R6 are different, the carbon attached to R5 and R6 can be in an S configuration or an R configuration.
  • In some embodiments of the compounds of formula (I)-(VI) and (A)-(H-I), a hydrogen atom can be replaced with a deuterium atom.
  • In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Table A below, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), or (D)-(H-I), the compound is selected from Compounds A3, A5, A7, A13, A17, A18, A22, A23, A24, A25, A28, A30, A31, A32, A34, A35, A38, A40, A41, A42, A45, A49, A52, A53, A54, A56, A57, A58, A62, A63, A64, A65, A68, A73, A74, A75, or A76, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A1, A2, A4, A6, A8, A9, A10, A11, A12, A14, A15, A16, A19, A20, A21, A26, A27, A29, A33, A36, A37, A39, A43, A44, A46, A47, A48, A50, A51, A55, A59, A60, A61, A66, A67, A69, A70, A71, A72, A77, A78, A79, A80, A81, A82, A83, A84, A85, A86, A87, A88, A89, A90, A91, A92, A93, A94, A95, A96, or A97, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A98-A186, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A187-A211, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A1-A211, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A212-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A1-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • In one embodiment of the compound of formula (I)-(VA), (A), (A-I), or (D)-(H-I), the compound is selected from A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • TABLE A
    Compounds
    Compound
    ID Structure
    A1 
    Figure US20200123117A1-20200423-C00077
    A2 
    Figure US20200123117A1-20200423-C00078
    A3 
    Figure US20200123117A1-20200423-C00079
    A4 
    Figure US20200123117A1-20200423-C00080
    A5 
    Figure US20200123117A1-20200423-C00081
    A6 
    Figure US20200123117A1-20200423-C00082
    A7 
    Figure US20200123117A1-20200423-C00083
    A8 
    Figure US20200123117A1-20200423-C00084
    A9 
    Figure US20200123117A1-20200423-C00085
    A10 
    Figure US20200123117A1-20200423-C00086
    A11 
    Figure US20200123117A1-20200423-C00087
    A12 
    Figure US20200123117A1-20200423-C00088
    A13 
    Figure US20200123117A1-20200423-C00089
    A14 
    Figure US20200123117A1-20200423-C00090
    A15 
    Figure US20200123117A1-20200423-C00091
    A16 
    Figure US20200123117A1-20200423-C00092
    A17 
    Figure US20200123117A1-20200423-C00093
    A18 
    Figure US20200123117A1-20200423-C00094
    A19 
    Figure US20200123117A1-20200423-C00095
    A20 
    Figure US20200123117A1-20200423-C00096
    A21 
    Figure US20200123117A1-20200423-C00097
    A22 
    Figure US20200123117A1-20200423-C00098
    A23 
    Figure US20200123117A1-20200423-C00099
    A24 
    Figure US20200123117A1-20200423-C00100
    A25 
    Figure US20200123117A1-20200423-C00101
    A26 
    Figure US20200123117A1-20200423-C00102
    A27 
    Figure US20200123117A1-20200423-C00103
    A28 
    Figure US20200123117A1-20200423-C00104
    A29 
    Figure US20200123117A1-20200423-C00105
    A30 
    Figure US20200123117A1-20200423-C00106
    A31 
    Figure US20200123117A1-20200423-C00107
    A32 
    Figure US20200123117A1-20200423-C00108
    A33 
    Figure US20200123117A1-20200423-C00109
    A34 
    Figure US20200123117A1-20200423-C00110
    A35 
    Figure US20200123117A1-20200423-C00111
    A36 
    Figure US20200123117A1-20200423-C00112
    A37 
    Figure US20200123117A1-20200423-C00113
    A38 
    Figure US20200123117A1-20200423-C00114
    A39 
    Figure US20200123117A1-20200423-C00115
    A40 
    Figure US20200123117A1-20200423-C00116
    A41 
    Figure US20200123117A1-20200423-C00117
    A42 
    Figure US20200123117A1-20200423-C00118
    A43 
    Figure US20200123117A1-20200423-C00119
    A44 
    Figure US20200123117A1-20200423-C00120
    A45 
    Figure US20200123117A1-20200423-C00121
    A46 
    Figure US20200123117A1-20200423-C00122
    A47 
    Figure US20200123117A1-20200423-C00123
    A48 
    Figure US20200123117A1-20200423-C00124
    A49 
    Figure US20200123117A1-20200423-C00125
    A50 
    Figure US20200123117A1-20200423-C00126
    A51 
    Figure US20200123117A1-20200423-C00127
    A52 
    Figure US20200123117A1-20200423-C00128
    A53 
    Figure US20200123117A1-20200423-C00129
    A54 
    Figure US20200123117A1-20200423-C00130
    A55 
    Figure US20200123117A1-20200423-C00131
    A56 
    Figure US20200123117A1-20200423-C00132
    A57 
    Figure US20200123117A1-20200423-C00133
    A58 
    Figure US20200123117A1-20200423-C00134
    A59 
    Figure US20200123117A1-20200423-C00135
    A60 
    Figure US20200123117A1-20200423-C00136
    A61 
    Figure US20200123117A1-20200423-C00137
    A62 
    Figure US20200123117A1-20200423-C00138
    A63 
    Figure US20200123117A1-20200423-C00139
    A64 
    Figure US20200123117A1-20200423-C00140
    A65 
    Figure US20200123117A1-20200423-C00141
    A66 
    Figure US20200123117A1-20200423-C00142
    A67 
    Figure US20200123117A1-20200423-C00143
    A68 
    Figure US20200123117A1-20200423-C00144
    A69 
    Figure US20200123117A1-20200423-C00145
    A70 
    Figure US20200123117A1-20200423-C00146
    A71 
    Figure US20200123117A1-20200423-C00147
    A72 
    Figure US20200123117A1-20200423-C00148
    A73 
    Figure US20200123117A1-20200423-C00149
    A74 
    Figure US20200123117A1-20200423-C00150
    A75 
    Figure US20200123117A1-20200423-C00151
    A76 
    Figure US20200123117A1-20200423-C00152
    A77 
    Figure US20200123117A1-20200423-C00153
    A78 
    Figure US20200123117A1-20200423-C00154
    A79 
    Figure US20200123117A1-20200423-C00155
    A80 
    Figure US20200123117A1-20200423-C00156
    A81 
    Figure US20200123117A1-20200423-C00157
    A82 
    Figure US20200123117A1-20200423-C00158
    A83 
    Figure US20200123117A1-20200423-C00159
    A84 
    Figure US20200123117A1-20200423-C00160
    A85 
    Figure US20200123117A1-20200423-C00161
    A86 
    Figure US20200123117A1-20200423-C00162
    A87 
    Figure US20200123117A1-20200423-C00163
    A88 
    Figure US20200123117A1-20200423-C00164
    A89 
    Figure US20200123117A1-20200423-C00165
    A90 
    Figure US20200123117A1-20200423-C00166
    A91 
    Figure US20200123117A1-20200423-C00167
    A92 
    Figure US20200123117A1-20200423-C00168
    A93 
    Figure US20200123117A1-20200423-C00169
    A94 
    Figure US20200123117A1-20200423-C00170
    A95 
    Figure US20200123117A1-20200423-C00171
    A96 
    Figure US20200123117A1-20200423-C00172
    A97 
    Figure US20200123117A1-20200423-C00173
    A98 
    Figure US20200123117A1-20200423-C00174
    A99 
    Figure US20200123117A1-20200423-C00175
    A100
    Figure US20200123117A1-20200423-C00176
    A101
    Figure US20200123117A1-20200423-C00177
    A102
    Figure US20200123117A1-20200423-C00178
    A103
    Figure US20200123117A1-20200423-C00179
    A104
    Figure US20200123117A1-20200423-C00180
    A105
    Figure US20200123117A1-20200423-C00181
    A106
    Figure US20200123117A1-20200423-C00182
    A107
    Figure US20200123117A1-20200423-C00183
    A108
    Figure US20200123117A1-20200423-C00184
    A109
    Figure US20200123117A1-20200423-C00185
    A110
    Figure US20200123117A1-20200423-C00186
    A111
    Figure US20200123117A1-20200423-C00187
    A112
    Figure US20200123117A1-20200423-C00188
    A113
    Figure US20200123117A1-20200423-C00189
    A114
    Figure US20200123117A1-20200423-C00190
    A115
    Figure US20200123117A1-20200423-C00191
    A116
    Figure US20200123117A1-20200423-C00192
    A117
    Figure US20200123117A1-20200423-C00193
    A118
    Figure US20200123117A1-20200423-C00194
    A119
    Figure US20200123117A1-20200423-C00195
    A120
    Figure US20200123117A1-20200423-C00196
    A121
    Figure US20200123117A1-20200423-C00197
    A122
    Figure US20200123117A1-20200423-C00198
    A123
    Figure US20200123117A1-20200423-C00199
    A124
    Figure US20200123117A1-20200423-C00200
    A125
    Figure US20200123117A1-20200423-C00201
    A126
    Figure US20200123117A1-20200423-C00202
    A127
    Figure US20200123117A1-20200423-C00203
    A128
    Figure US20200123117A1-20200423-C00204
    A129
    Figure US20200123117A1-20200423-C00205
    A130
    Figure US20200123117A1-20200423-C00206
    A131
    Figure US20200123117A1-20200423-C00207
    A132
    Figure US20200123117A1-20200423-C00208
    A133
    Figure US20200123117A1-20200423-C00209
    A134
    Figure US20200123117A1-20200423-C00210
    A135
    Figure US20200123117A1-20200423-C00211
    A136
    Figure US20200123117A1-20200423-C00212
    A137
    Figure US20200123117A1-20200423-C00213
    A138
    Figure US20200123117A1-20200423-C00214
    A139
    Figure US20200123117A1-20200423-C00215
    A140
    Figure US20200123117A1-20200423-C00216
    A141
    Figure US20200123117A1-20200423-C00217
    A142
    Figure US20200123117A1-20200423-C00218
    A143
    Figure US20200123117A1-20200423-C00219
    A144
    Figure US20200123117A1-20200423-C00220
    A145
    Figure US20200123117A1-20200423-C00221
    A146
    Figure US20200123117A1-20200423-C00222
    A147
    Figure US20200123117A1-20200423-C00223
    A148
    Figure US20200123117A1-20200423-C00224
    A149
    Figure US20200123117A1-20200423-C00225
    A150
    Figure US20200123117A1-20200423-C00226
    A151
    Figure US20200123117A1-20200423-C00227
    A152
    Figure US20200123117A1-20200423-C00228
    A153
    Figure US20200123117A1-20200423-C00229
    A154
    Figure US20200123117A1-20200423-C00230
    A155
    Figure US20200123117A1-20200423-C00231
    A156
    Figure US20200123117A1-20200423-C00232
    A157
    Figure US20200123117A1-20200423-C00233
    A158
    Figure US20200123117A1-20200423-C00234
    A159
    Figure US20200123117A1-20200423-C00235
    A160
    Figure US20200123117A1-20200423-C00236
    A161
    Figure US20200123117A1-20200423-C00237
    A162
    Figure US20200123117A1-20200423-C00238
    A163
    Figure US20200123117A1-20200423-C00239
    A164
    Figure US20200123117A1-20200423-C00240
    A165
    Figure US20200123117A1-20200423-C00241
    A166
    Figure US20200123117A1-20200423-C00242
    A167
    Figure US20200123117A1-20200423-C00243
    A168
    Figure US20200123117A1-20200423-C00244
    A169
    Figure US20200123117A1-20200423-C00245
    A170
    Figure US20200123117A1-20200423-C00246
    A171
    Figure US20200123117A1-20200423-C00247
    A172
    Figure US20200123117A1-20200423-C00248
    A173
    Figure US20200123117A1-20200423-C00249
    A174
    Figure US20200123117A1-20200423-C00250
    A175
    Figure US20200123117A1-20200423-C00251
    A176
    Figure US20200123117A1-20200423-C00252
    A177
    Figure US20200123117A1-20200423-C00253
    A178
    Figure US20200123117A1-20200423-C00254
    A179
    Figure US20200123117A1-20200423-C00255
    A180
    Figure US20200123117A1-20200423-C00256
    A181
    Figure US20200123117A1-20200423-C00257
    A182
    Figure US20200123117A1-20200423-C00258
    A183
    Figure US20200123117A1-20200423-C00259
    A184
    Figure US20200123117A1-20200423-C00260
    A185
    Figure US20200123117A1-20200423-C00261
    A186
    Figure US20200123117A1-20200423-C00262
    A187
    Figure US20200123117A1-20200423-C00263
    A188
    Figure US20200123117A1-20200423-C00264
    A189
    Figure US20200123117A1-20200423-C00265
    A190
    Figure US20200123117A1-20200423-C00266
    A191
    Figure US20200123117A1-20200423-C00267
    A192
    Figure US20200123117A1-20200423-C00268
    A193
    Figure US20200123117A1-20200423-C00269
    A194
    Figure US20200123117A1-20200423-C00270
    A195
    Figure US20200123117A1-20200423-C00271
    A196
    Figure US20200123117A1-20200423-C00272
    A197
    Figure US20200123117A1-20200423-C00273
    A198
    Figure US20200123117A1-20200423-C00274
    A199
    Figure US20200123117A1-20200423-C00275
    A200
    Figure US20200123117A1-20200423-C00276
    A201
    Figure US20200123117A1-20200423-C00277
    A202
    Figure US20200123117A1-20200423-C00278
    A203
    Figure US20200123117A1-20200423-C00279
    A204
    Figure US20200123117A1-20200423-C00280
    A205
    Figure US20200123117A1-20200423-C00281
    A206
    Figure US20200123117A1-20200423-C00282
    A207
    Figure US20200123117A1-20200423-C00283
    A208
    Figure US20200123117A1-20200423-C00284
    A209
    Figure US20200123117A1-20200423-C00285
    A210
    Figure US20200123117A1-20200423-C00286
    A211
    Figure US20200123117A1-20200423-C00287
    A212
    Figure US20200123117A1-20200423-C00288
    A213
    Figure US20200123117A1-20200423-C00289
    A214
    Figure US20200123117A1-20200423-C00290
    A215
    Figure US20200123117A1-20200423-C00291
    A216
    Figure US20200123117A1-20200423-C00292
    A217
    Figure US20200123117A1-20200423-C00293
    A218
    Figure US20200123117A1-20200423-C00294
    A219
    Figure US20200123117A1-20200423-C00295
    A220
    Figure US20200123117A1-20200423-C00296
    A221
    Figure US20200123117A1-20200423-C00297
    A222
    Figure US20200123117A1-20200423-C00298
    A223
    Figure US20200123117A1-20200423-C00299
    A224
    Figure US20200123117A1-20200423-C00300
    A225
    Figure US20200123117A1-20200423-C00301
    A226
    Figure US20200123117A1-20200423-C00302
    A227
    Figure US20200123117A1-20200423-C00303
    A228
    Figure US20200123117A1-20200423-C00304
    A229
    Figure US20200123117A1-20200423-C00305
    A230
    Figure US20200123117A1-20200423-C00306
    A231
    Figure US20200123117A1-20200423-C00307
    A232
    Figure US20200123117A1-20200423-C00308
    A233
    Figure US20200123117A1-20200423-C00309
    A234
    Figure US20200123117A1-20200423-C00310
  • In one embodiment of the compound of formula (I)-(IV), (VI), (B) or (C), the compound is selected from Table B below, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(IV), (VI), (B) or (C), the compound is selected from Compounds B1, B2, B3, or B6 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(IV), (VI), (B) or (C), the compound is selected from Compounds B4, B5, B7, B8, B9, B10, or B11 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In one embodiment of the compound of formula (I)-(IV), (VI), (B) or (C), the compound is selected from Compounds B1-B11 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • TABLE B
    Compounds
    Compound
    ID Structure
    B1
    Figure US20200123117A1-20200423-C00311
    B2
    Figure US20200123117A1-20200423-C00312
    B3
    Figure US20200123117A1-20200423-C00313
    B4
    Figure US20200123117A1-20200423-C00314
    B5
    Figure US20200123117A1-20200423-C00315
    B6
    Figure US20200123117A1-20200423-C00316
    B7
    Figure US20200123117A1-20200423-C00317
    B8
    Figure US20200123117A1-20200423-C00318
    B9
    Figure US20200123117A1-20200423-C00319
    B10
    Figure US20200123117A1-20200423-C00320
    B11
    Figure US20200123117A1-20200423-C00321
  • 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), (IA), (IB), (IC), (II), (IIA), (IIIA), (IIB), (III), (IV), (IVA), (V), (VA), (VI), (A), (A-I), (B)-(D), (E), (E-I)-(E-VII), (F), (G), (G-I), (G-II), (H), and (H-I) (“formula (I)-(VI) and (A)-(H-I)”) or compounds of Tables A and B, 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 A3, A5, A7, A13, A17, A18, A22, A23, A24, A25, A28, A30, A31, A32, A34, A35, A38, A40, A41, A42, A45, A49, A52, A53, A54, A56, A57, A58, A62, A63, A64, A65, A68, A73, A74, A75, A76, B1, B2, B3, or B6 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, A2, A4, A6, A8, A9, A10, A11, A12, A14, A15, A16, A19, A20, A21, A26, A27, A29, A33, A36, A37, A39, A43, A44, A46, A47, A48, A50, A51, A55, A59, A60, A61, A66, A67, A69, A70, A71, A72, A77, A78, A79, A80, A81, A82, A83, A84, A85, A86, A87, A88, A89, A90, A91, A92, A93, A94, A95, A96, A97, B4, B5, B7, B8, B9, B10, or B11, 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 A98-A186, 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 A187-A211, 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 A212-A234, 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-A96, A98-A116, A118-A159, A161-A175, and A177-A234, 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 A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, 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-A211 or B1-B11, 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-A234 or B1-B11, 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 of any one of formula (D)-(H-I), 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)-(VI) and (A)-(H-I), 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)-(VI) and (A)-(H-I), 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)-(VI) and (A)-(H-I), 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)-(VI) and (A)-(H-I), 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 accordance with another embodiment, there is provided a use of the compounds of formula (I)-(VI) and (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof for preparation of a medicament for modulating androgen receptor (AR). In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds provide metabolic stability. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human liver microsomes (HLMs) of less than about 500 μL/min/mg protein, less than about 450 μL/min/mg protein, less than about 400 μL/min/mg protein, less than about 350 μL/min/mg protein, less than about 300 μL/min/mg protein, less than about 250 μL/min/mg protein, less than about 225 μL/min/mg protein, or less than about 200 μL/min/mg protein. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than about 200 μL/min/mg protein, less than about 190 μL/min/mg protein, less than about 180 μL/min/mg protein, less than about 170 μL/min/mg protein, less than about 160 μL/min/mg protein, less than about 150 μL/min/mg protein, less than about 140 μL/min/mg protein, less than about 130 μL/min/mg protein, less than about 120 μL/min/mg protein, less than about 110 μL/min/mg protein, less than about 100 μL/min/mg protein, less than about 90 μL/min/mg protein, less than about 80 μL/min/mg protein, less than about 70 μL/min/mg protein, less than about 60 μL/min/mg protein, or less than about 50 μL/min/mg protein. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than about 50 μL/min/mg protein, less than about 48 μL/min/mg protein, less than about 45 μL/min/mg protein, less than about 40 μL/min/mg protein, less than about 35 μL/min/mg protein, less than about 30 μL/min/mg protein, less than about 25 μL/min/mg protein, less than about 20 μL/min/mg protein, or less than about 15 μL/min/mg protein. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than about 15 μL/min/mg protein, less than about 14 μL/min/mg protein, less than about 13 μL/min/mg protein, less than about 12 μL/min/mg protein, less than about 11 μL/min/mg protein, or less than about 10 μL/min/mg protein.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than 12 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs in the range of about 0.1 μL/min/mg protein to about 12 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs of less than 48 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs in the range of about 12 μL/min/mg protein to about 48 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HLMs in the range of about 0.1 μL/min/mg protein to about 48 μL/min/mg protein.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 20 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 15 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 20 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 15 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein.
  • In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse liver microsomes (MLMs) of less than about 500 μL/min/mg protein, less than about 450 μL/min/mg protein, less than about 400 μL/min/mg protein, less than about 350 μL/min/mg protein, less than about 300 μL/min/mg protein, less than about 250 μL/min/mg protein, less than about 225 μL/min/mg protein, or less than about 200 μL/min/mg protein. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MLMs of less than about 200 μL/min/mg protein, less than about 190 μL/min/mg protein, less than about 180 μL/min/mg protein, less than about 170 μL/min/mg protein, less than about 160 μL/min/mg protein, less than about 150 μL/min/mg protein, less than about 140 μL/min/mg protein, less than about 130 μL/min/mg protein, less than about 120 μL/min/mg protein, less than about 110 μL/min/mg protein, less than about 100 μL/min/mg protein, less than about 90 μL/min/mg protein, less than about 80 μL/min/mg protein, less than about 70 μL/min/mg protein, less than about 60 μL/min/mg protein, or less than about 50 μL/min/mg protein. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MLMs of less than about 50 μL/min/mg protein, less than about 48 μL/min/mg protein, less than about 45 μL/min/mg protein, less than about 40 μL/min/mg protein, less than about 35 μL/min/mg protein, less than about 30 μL/min/mg protein, less than about 25 μL/min/mg protein, less than about 20 μL/min/mg protein, or less than about 15 μL/min/mg protein. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MLMs of less than about 15 μL/min/mg protein, less than about 14 μL/min/mg protein, less than about 13 μL/min/mg protein, less than about 12 μL/min/mg protein, less than about 11 μL/min/mg protein, or less than about 10 μL/min/mg protein.
  • In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 12 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs in the range of about 0.1 μL/min/mg protein to about 12 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 48 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs in the range of about 12 μL/min/mg protein to about 48 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs in the range of about 0.1 μL/min/mg protein to about 48 μL/min/mg protein.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 20 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 15 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 20 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 15 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an MLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein.
  • In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human hepatocytes (HHs) of less than about 500 μL/min/million cells, less than about 450 μL/min/million cells, less than about 400 μL/min/million cells, less than about 350 μL/min/million cells, less than about 300 μL/min/million cells, less than about 250 μL/min/million cells, less than about 225 μL/min/million cells, or less than about 200 μL/min/million cells. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 200 μL/min/million cells, less than about 190 μL/min/million cells, less than about 180 μL/min/million cells, less than about 170 μL/min/million cells, less than about 160 μL/min/million cells, less than about 150 μL/min/million cells, less than about 140 μL/min/million cells, less than about 130 μL/min/million cells, less than about 120 μL/min/million cells, less than about 110 μL/min/million cells, less than about 100 μL/min/million cells, less than about 90 μL/min/million cells, less than about 80 μL/min/million cells, less than about 70 μL/min/million cells, less than about 60 μL/min/million cells, or less than about 50 μL/min/million cells. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 50 μL/min/million cells, less than about 48 μL/min/million cells, less than about 45 μL/min/million cells, less than about 40 μL/min/million cells, less than about 35 μL/min/million cells, less than about 30 μL/min/million cells, less than about 25 μL/min/million cells, less than about 20 μL/min/million cells, or less than about 15 μL/min/million cells. In some embodiments, the compounds of the present disclosure have an in vitro metabolic clearance in HHs of less than about 15 μL/min/million cells, less than about 14 μL/min/million cells, less than about 13 μL/min/million cells, less than about 12 μL/min/million cells, less than about 11 μL/min/million cells, less than about 10 μL/min/million cells, less than about 9 μL/min/million cells, less than about 8 μL/min/million cells, less than about 8 μL/min/million cells, less than about 6 μL/min/million cells, or less than about 5 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 4 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs in the range of about 0.1 μL/min/million cells to about 4 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 18 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs in the range of about 4 μL/min/million cells to about 18 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs in the range of about 0.1 μL/min/million cells to about 18 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 10 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 5 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 3.85 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 10 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in HHs of less than about 5 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human hepatocytes of less than about 3.85 μL/min/million cells.
  • In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes (MHs) of less than about 500 μL/min/million cells, less than about 450 μL/min/million cells, less than about 400 μL/min/million cells, less than about 350 μL/min/million cells, less than about 300 μL/min/million cells, less than about 250 μL/min/million cells, less than about 225 μL/min/million cells, or less than about 200 μL/min/million cells. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 200 μL/min/million cells, less than about 190 μL/min/million cells, less than about 180 μL/min/million cells, less than about 170 μL/min/million cells, less than about 160 μL/min/million cells, less than about 150 μL/min/million cells, less than about 140 μL/min/million cells, less than about 130 μL/min/million cells, less than about 120 μL/min/million cells, less than about 110 μL/min/million cells, less than about 100 μL/min/million cells, less than about 90 μL/min/million cells, less than about 80 μL/min/million cells, less than about 70 μL/min/million cells, less than about 60 μL/min/million cells, or less than about 50 μL/min/million cells. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 50 μL/min/million cells, less than about 48 μL/min/million cells, less than about 45 μL/min/million cells, less than about 40 μL/min/million cells, less than about 35 μL/min/million cells, less than about 30 μL/min/million cells, less than about 25 μL/min/million cells, less than about 20 μL/min/million cells, or less than about 15 μL/min/million cells. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 15 μL/min/million cells, less than about 14 μL/min/million cells, less than about 13 μL/min/million cells, less than about 12 μL/min/million cells, less than about 11 μL/min/million cells, less than about 10 μL/min/million cells, less than about 9 μL/min/million cells, less than about 8 μL/min/million cells, less than about 8 μL/min/million cells, less than about 6 μL/min/million cells, or less than about 5 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 4 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of about 0.1 μL/min/million cells to about 4 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 18 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of about 4 μL/min/million cells to about 18 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of about 0.1 μL/min/million cells to about 18 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 17 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 7 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 3.85 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 17 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 7 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A or B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in MHs of less than about 3.85 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are any compounds with a combination of in vitro metabolic clearance in HLM, MLM, HHs, and MHs as described herein.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein.
  • In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 17 μL/min/million cells and a an in vitro metabolic clearance in human hepatocytes of less than about 10 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 7 μL/min/million cells, and an in vitro metabolic clearance in human hepatocytes of less than about 5 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 17 μL/min/million cells and a an in vitro metabolic clearance in human hepatocytes of less than about 10 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in mouse hepatocytes of less than about 7 μL/min/million cells, and an in vitro metabolic clearance in human hepatocytes of less than about 5 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 μL/min/mg protein and an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 μL/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 17 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 7 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds s have an HLM and MLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM or MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein and b) an in vitro metabolic clearance in human or mouse hepatocytes of less than about 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM or MLM in vitro metabolic clearance of less than 12 μL/min/mg protein and b) an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein and b) an in vitro metabolic clearance in human or mouse hepatocytes of less than about 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM or MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than about 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than about 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than 12 μL/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than 4 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an a) HLM and MLM in vitro metabolic clearance of less than 12 μL/min/mg protein and b) an in vitro metabolic clearance in human and mouse hepatocytes of less than 4 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 20 μL/min/mg protein and an in vitro metabolic clearance in human and mouse hepatocytes of less than about 20 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 15 μL/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 17 μL/min/million cells. In another embodiment, the compounds of the present are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 12 μL/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 7 μL/min/million cells. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an HLM and MLM in vitro metabolic clearance of less than about 11.55 μL/min/mg protein, an in vitro metabolic clearance in human and mouse hepatocytes of less than about 3.85 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 120 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 110 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 100 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 90 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 80 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human, mouse, rat, monkey or dog microsome greater than about 70 minutes.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human microsomes greater than about 120 min.
  • In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse microsomes greater than about 120 min.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 120 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 110 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 100 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 90 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 80 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 70 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 60 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 50 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 40 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 30 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 20 minutes. In some embodiments, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat microsome greater than about 10 minutes. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 360 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 350 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 325 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 310 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 300 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 290 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 250 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 200 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 150 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 100 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in human hepatocytes greater than about 80 minutes. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 360 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 350 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 325 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 300 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 250 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 225 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 200 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 150 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 100 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 50 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in mouse hepatocytes greater than about 30 minutes. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 360 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 350 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 325 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 300 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 250 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 225 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 200 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 150 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 100 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life in rat hepatocytes greater than about 50 minutes. In one embodiment, the compounds have an in vitro half-life in rat hepatocytes greater than about 30 minutes. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 80 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 90 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 100 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 110 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 115 minutes. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome of greater than 120 minutes. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof. In one embodiment, the mammal microsome is human or mouse microsome.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 50 min to about 200 min. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 80 min to about 180 min. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 100 min to about 160 min. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 110 min to about 150 min. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 110 min to about 140 min. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 110 min to about 130 min. In one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein the compounds have an in vitro half-life stability in mammal microsome in the range of about 115 min to about 130 min. In another embodiment, the compounds of the present disclosure are any compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, having an in vitro half-life stability in mammal microsome in the range of about 115 min to about 130 min. In one embodiment, the mammal microsome is human or mouse microsome.
  • 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 Tables A and B 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 one embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 500 nM. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof. In one embodiment, the mammal microsome is human or mouse microsome.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 and a HLM or MLM in vitro metabolic clearance of less than 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 and a HLM and MLM in vitro metabolic clearance of less than 12 μL/min/mg protein. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 and an in vitro metabolic clearance in human and mouse hepatocytes of less than 4 μL/min/million cells.
  • In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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; a HLM or MLM in vitro metabolic clearance of less than 12 μL/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 800 nM; a HLM or MLM in vitro metabolic clearance of less than 12 μL/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 μL/min/million cells. In another embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, or the compounds of Tables A and B, 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 700 nM; a HLM or MLM in vitro metabolic clearance of less than 12 μL/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 μL/min/million cells. In one embodiment, the compound is selected from any of the compound of Tables A and B or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof.
  • In a specific embodiment, the compounds of the present disclosure are any compound of Tables A and B, 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; a HLM or MLM in vitro metabolic clearance of less than 12 μL/min/mg protein; and an in vitro metabolic clearance in human or mouse hepatocytes of less than 4 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 4 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in HHs of less than 4 μL/min/million cells and in vitro IC50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro metabolic clearance in HHs of less than 4 μL/min/million cells and in vitro IC50 in a PSA-luciferase assay of less than about 675 nM. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in HHs of less than about 350 minutes. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in HHs of greater than about 320 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro half-life in HHs of greater than about 350 minutes and in vitro IC50 in a PSA-luciferase assay of less than about 700 nM.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 15 μL/min/mg protein. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MLMs of less than 15 μL/min/mg protein and in vitro IC50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro metabolic clearance in MLMs of less than 15 μL/min/mg protein and in vitro IC50 in a PSA-luciferase assay of less than about 675 nM. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 10 μL/min/million cells. In one embodiment, the compounds have an in vitro metabolic clearance in MHs of less than 8 μL/min/million cells. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs of less than 10 μL/min/million cells and in vitro IC50 in a PSA-luciferase assay of less than about 700 nM. In one embodiment, the compounds have an in vitro metabolic clearance in MHs of less than 8 μL/min/million cells and in vitro IC50 in a PSA-luciferase assay of less than about 675 nM. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in MLMs of greater than about 100 minutes. In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro half-life in MHs of greater than about 100 minutes. In one embodiment, the compounds have an in vitro half-life in MHs of greater than about 180 minutes.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, and wherein the compounds have an in vitro metabolic clearance in MHs in the range of 3 μL/min/million cells to about 20 μL/min/million cells. In one embodiment, the compounds have an in vitro metabolic clearance in MHs in the range of 3.2 μL/min/million cells to about 18 μL/min/million cells. In one embodiment, the compounds have an in vitro metabolic clearance in MHs in the range of 3.4 μL/min/million cells to about 17 μL/min/million cells.
  • In a specific embodiment, the compounds of the present disclosure are compounds having the structure of formula (I)-(VI) and/or (A)-(H-I), 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)-(VI) and/or (A)-(H-I), 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)-(VI) and/or (A)-(H-I), 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)-(VI) and/or (A)-(H-I), 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)-(VI) and/or (A)-(H-I), 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 one embodiment, a pharmaceutical composition as described herein comprise one or more compounds selected from Table B, 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 A3, A5, A7, A13, A17, A18, A22, A23, A24, A25, A28, A30, A31, A32, A34, A35, A38, A40, A41, A42, A45, A49, A52, A53, A54, A56, A57, A58, A62, A63, A64, A65, A68, A73, A74, A75, A76, B1, B2, B3, or B6, 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, A2, A4, A6, A8, A9, A10, A11, A12, A14, A15, A16, A19, A20, A21, A26, A27, A29, A33, A36, A37, A39, A43, A44, A46, A47, A48, A50, A51, A55, A59, A60, A61, A66, A67, A69, A70, A71, A72, A77, A78, A79, A80, A81, A82, A83, A84, A85, A86, A87, A88, A89, A90, A91, A92, A93, A94, A95, A96, A97, B4, B5, B7, B8, B9, B10, or B11, 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 A98-A186. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A187-A211. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A212-A234. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A1-A96, A98-A116, A118-A159, A161-A175, and A177-A234. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A13, A57, A74, A93, A109, A112, A122, A126, A131, A134, A136, A137, A164, A168, A169, A170, A171, A172, A184, A185, A195, and/or A204, or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A1-A211 or B1-B11. In a specific embodiment, a pharmaceutical composition, as described herein, comprises one or more compounds selected from Compounds A1-A234 or B1-B11.
  • In one embodiment, a pharmaceutical composition, as described herein, comprising one or more compounds of formula (I)-(VI) and/or (A)-(H-I), 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, SKF105,111; 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)-(VI) and/or (A)-(H-I), 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)-(VI) and/or (A)-(H-I), 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, 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)-(VI) and/or (A)-(H-I), 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 propellent.
  • Solid carriers suitable for use in the present application include, but are not limited to, inert substances such as lactose, starch, glucose, methyl-cellulose, 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/or combination, and may make a pharmaceutical dosage form containing the composition and/or combination easier for the patient and care giver to handle. Diluents for solid compositions and/or combinations 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(r)), 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 and/or combinations 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 and/or combination. 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/or combination and to improve the accuracy of dosing. Excipients that may function as glidants include colloidal silicon dioxide, magnesium trisilicate, 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 and/or combination 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 and/or combination 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)-(VI) and/or (A)-(H-I), 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 and/or combination an active ingredient or other excipient that is not soluble in the liquid carrier. Emulsifying agents that may be useful in liquid compositions and/or combinations 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, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl 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 certain of such embodiments 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)-(VI) and/or (A)-(H-I), 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)-(VI) and/or (A)-(H-I), or a pharmaceutically acceptable salt or solvate thereof, or compounds disclosed in Tables A and B, 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 5-[[4-[1-[3,5-dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-methylsulfonyl-oxazole (A3)
  • Figure US20200123117A1-20200423-C00322
  • (4-Methylsulfonyloxazol-5-yl) 4-methylbenzenesulfonate (2)
  • To a solution of 4-methylsulfonyloxazol-5-ol (1) (0.10 g, 0.56 mmol), TEA (0.11 g, 1.1 mmol) and DMAP (14 mg, 0.11 mol) in DCM (3 mL) was added Tosyl chloride (0.10 g, 0.52 mmol) dropwise at 0° C. Then the solution was stirred at the same temperature for 1 hour. TLC showed the reaction was completed. The mixture was poured into H2O (5 mL), extracted with DCM (3 mL×3). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (4-methylsulfonyloxazol-5-yl) 4-methylbenzenesulfonate (2) (0.11 g, yield: 56.6%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.98 (s, 1H), 7.93 (d, J=8.40 Hz, 2H), 7.42 (d, J=8.80 Hz, 2H), 4.95 (s, 2H), 3.22 (s, 3H).
  • Methyl 3,5-dichloro-4-(3-chloropropoxy)benzoate (5)
  • To a mixture of methyl 3, 5-dichloro-4-hydroxybenzoate (3) (100.0 g, 0.45 mol) and Cs2CO3 (293.2 g, 0.90 mol) in DMF (1.5 L) was added 1-chloro-3-iodopropane (4) (102.2 g, 0.50 mol), and the mixture was stirred at 80° C. for 16 hours. TLC showed the reaction was completed. The mixture was cooled down, quenched with water (3.5 L) and extracted with MTBE (1.0 L×2). The combined organic layers were washed with brine (1 L), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give methyl 3,5-dichloro-4-(3-chloropropoxy)benzoate (70.0 g, yield: 52.0%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 2.29 (q, J=6.03 Hz, 2H), 3.85 (t, J=6.36 Hz, 2H), 3.91 (s, 3H), 4.22 (t, J=5.70 Hz, 2H), 7.92-8.00 (m, 2H).
  • 2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-ol (6)
  • To a solution of 3,5-dichloro-4-(3-chloropropoxy)benzoate (5) (70.0 g, 0.24 mol) in THF (500 mL) was added MeMgBr (400 mL, 1.20 mol) at 0° C. under N2 atmosphere. The mixture was stirred at 20° C. for 16 hours under N2 atmosphere. TLC showed the reaction was completed. The mixture was poured into H2O (500 mL), extracted with EtOAc (200 mL×5), and the combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give 2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-ol (44.0 g, yield: 62.9%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 1.56 (s, 6H), 2.29 (q, J=6.06 Hz, 2H), 3.87 (t, J=6.39 Hz, 2H), 4.13-4.19 (m, 2H), 7.41 (s, 2H).
  • 4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenol (7)
  • To a solution of 2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-ol (6) (20.0 g, 67.2 mmol) and phenol (7.0 g, 73.9 mmol) in CCl4 (400 mL) was added BF3.Et2O (16.6 mL, 134.2 mmol) dropwise at 0° C. under N2 atmosphere. The mixture was stirred at 0° C. for 30 min. TLC showed the reaction was completed. The resulting mixture was poured into H2O (300 mL), extracted with DCM (300 mL×3). The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenol (18.0 g, yield: 71.7%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.12 (s, 2H), 7.10-7.05 (m, 2H), 6.79-6.75 (m, 2H), 4.65 (s, 1H), 4.15 (t, J=5.7 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 2.28 (q, J=6.1 Hz, 2H), 1.62 (s, 6H).
  • 5-[[4-[1-[3,5-Dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-methylsulfonyl-oxazole (A3)
  • To a suspension of 4-[1-[3,5-dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenol (7) (0.135 g, 0.36 mmol) and Cs2CO3 (0.197 g, 0.6 mmol) in DMF (3 mL) was added (4-methylsulfonyloxazol-5-yl)methyl 4-methylbenzenesulfonate (2) (0.1 g, 0.3 mmol) at 25° C. The mixture was stirred at 60° C. for 6 hours. LCMS showed the reaction was completed. The resulting mixture was poured into H2O (8 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC (HCl) to give 5-[[4-[1-[3,5-dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-methylsulfonyl-oxazole (A1) (37.9 mg, yield: 23.6%) as yellow oil. HPLC purity (220 nm): 96.25%. 1H NMR (400 MHz, CHCl3-d) δ 7.99 (s, 1H), 7.16-7.10 (m, 4H), 6.94 (d, J=8.82 Hz, 2H), 5.42 (s, 2H), 4.15 (t, J=5.73 Hz, 2H), 3.86 (t, J=6.50 Hz, 2H) 3.18 (s, 3H), 2.28 (quin, J=6.17 Hz, 2H), 1.62 (s, 6H). LCMS (M+23) m/z: calcd 533. found 556.
  • Example 2: Synthesis of 4-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy) methyl)-1-(methylsulfonyl)-1H-imidazole (A5)
  • Figure US20200123117A1-20200423-C00323
  • (1-Trityl-1H-imidazol-4-yl)methanol (2)
  • To a solution of (1H-imidazol-4-yl)methanol (0.5 g, 5.3 mmol) and Et3N (1.1 g, 10.2 mmol) in DCM (20 mL) was added TrCl (1.5 g, 5.3 mmol) and the mixture was stirred at 50° C. for 4 hours. TLC showed the reaction was completed. The reaction was cooled down, quenched with water (40 mL), extracted with DCM (5 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give (1-trityl-1H-imidazol-4-yl)methanol (2) (1.0 g, yield: 76.8%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ=7.37-7.44 (m, 9H), 7.29 (d, J=1.34 Hz, 2H), 7.10 (dd, J=8.01, 1.41 Hz, 6H), 4.87 (t, J=5.56 Hz, 1H), 4.33 (d, J=5.50 Hz, 2H).
  • 4-(Chloromethyl)-1-trityl-1H-imidazole (3)
  • To a solution of (1-tritylimidazol-4-yl)methanol (2) (200 mg, 0.6 mmol) in DCM (3 mL) was added SOCl2 (0.2 mL, 2.9 mmol) dropwise at 0° C. and the mixture was stirred at 25° C. for 10 min. TLC showed the reaction was completed. The reaction was concentrated under reduced pressure to give 4-(chloromethyl)-1-trityl-1H-imidazole (3) (150 mg, yield: 71.1%) as yellow oil.
  • 4-((4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1-trityl-1H-imidazole (5)
  • A mixture of 4-(chloromethyl)-1-trityl-1H-imidazole (3) (150 mg, 0.4 mmol), Cs2CO3 (272 mg, 0.8 mmol) and 4-[1-[3,5-dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenol (4) (156 mg, 0.4 mmol) in MeCN (5 mL) was stirred at 80° C. for 4 hours. TLC showed the reaction was completed. The resulting mixture was cooled down, poured into H2O (10 mL), extracted with EtOAc (4 mL×2), and the combined organic layers were washed with brine (4 mL×5), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give 4-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1-trityl-1H-imidazole (5) (150 mg, yield: 71.1%) as white solid. 1H NMR (400 MHz, CHCl3-d) δ=7.45 (d, J=1.2 Hz, 1H), 7.38-7.30 (m, 9H), 7.19-7.06 (m, 10H), 6.95-6.86 (m, 3H), 4.99 (s, 2H), 4.14-4.10 (m, 2H), 3.86 (t, J=6.5 Hz, 2H), 2.28 (quin, J=6.1 Hz, 2H), 1.62 (s, 6H).
  • 4-((4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1H-imidazole (6)
  • A solution of 4-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1-trityl-1H-imidazole (5) (150 mg, 0.2 mmol) in HCl/EtOAc (4 M, 2 mL) was stirred at 25° C. for 2 hours. LCMS showed the reaction was completed. The reaction was filtered and the filtrate concentrated under reduced pressure to give 4-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1H-imidazole (6) (95 mg, yield: 97.2%) as colorless oil. LCMS (M+1) m/z: calcd 452.1. found 452.9.
  • 4-((4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1-(methylsulfonyl)-1H-imidazole (A5)
  • To a mixture of 4-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1H-imidazole (6) (80 mg, 0.2 mmol) and TEA (0.1 mL, 0.5 mmol) in DCM (2 mL) was added methanesulfonyl chloride (41 mg, 0.4 mmol) dropwise at 0° C., and the mixture was stirred at 25° C. for 2 hours. TLC showed the reaction was completed. The mixture was diluted with water (20 mL), extracted with DCM (5 mL×3), and the combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (NH4HCO3) to give 4-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-1-(methyl sulfonyl)-imidazole (A5) (16 mg, yield: 16.6%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ=7.99 (d, J=1.3 Hz, 1H), 7.40 (s, 1H), 7.15-7.12 (m, 4H), 6.95-6.90 (m, 2H), 5.05 (s, 2H), 4.15 (t, J=5.7 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.30 (s, 3H), 2.31-2.26 (m, 2H), 1.63 (s, 6H). LCMS (220 nm): 95.2%. LCMS (M+1) m/z: calcd 530.1. found 531.0.
  • Example 3: Synthesis of 2-((4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-5-(methylsulfonyl)-1, 3, 4-oxadiazole (A7)
  • Figure US20200123117A1-20200423-C00324
  • Methyl 2-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)acetate (3)
  • To a solution of 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenol (1) (1.00 g, 2.6 mmol) and methyl 2-bromoacetate (2) (0.49 g, 3.2 mmol) in DMF (10 mL) was added Cs2CO3 (1.30 g, 4.0 mmol) at 25° C. The mixture was stirred at 50° C. for 4 hours. LCMS showed the reaction was completed. The mixture was quenched with water (20 mL) and extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (20 mL×5), dried over Na2SO4, filtered and concentrated under reduced pressure to give the methyl 2-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)acetate (3) (1.00 g, yield: 70%) as colorless oil. LCMS (M+23) m z: calcd 444.07. found 467.1.
  • 2-(4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy) acetohydrazide (4)
  • To a solution of methyl 2-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl) phenoxy) acetate (3) (1.32 g, 2.9 mmol) in MeOH (10 mL) was added NH2NH2.H2O (0.59 g, 11.8 mmol) at 20° C. The mixture was stirred at 90° C. for 16 hours. TLC showed of the reaction was completed. The mixture was quenched with water (10 mL) and extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 2-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy) acetohydrazide (4) (1.0 g, yield: 75.8%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ ppm: 7.73 (br, 1H), 7.11-7.14 (d, J=8.9 Hz, 2H), 7.11 (s, 2H), 6.84 (d, J=8.9 Hz, 2H), 4.58 (s, 2H), 4.14-4.16 (t, J=5.2 Hz, 2H), 3.94 (br, 1H), 3.86 (t, J=6.4 Hz, 2H), 2.28 (m, 2H), 1.62 (s, 6H).
  • 3-(1-(4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)ethyl)-5-methylthio)-4H-pyrazole (5)
  • To a solution of 2-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy) acetohydrazide (4) (500 mg, 1.12 mmol) in MeOH (2 mL) was added KOH (75 mg, 1.33 mmol) and CS2 (76 mg, 2.8 mmol) at 20° C. The mixture was stirred at 65° C. for 2 hours. Then CH3I (2 mL) was added to the reaction mixture and stirred at 65° C. for 2 hours. LCMS showed the reaction was completed. The mixture was quenched with water (20 mL) and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-(1-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)ethyl)-5-(methylthio)-4H-pyrazole (5) (300 mg, yield: 54.5%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ ppm 7.14 (d, J=8.9 Hz, 2H), 7.11 (s, 2H), 6.94 (d, J=8.9 Hz, 2H), 5.22 (s, 1H), 4.15 (t, J=5.8 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 2.74 (s, 3H), 2.28 (t, J=5.99 Hz, 2H), 1.62 (s, 6H).
  • 2-((4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-5-(methylsulfonyl)-1,3,4-oxadiazole (A7)
  • To a solution of 3-(1-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)ethyl)-5-(methylthio)-4H-pyrazole (5) (220 mg, 0.49 mmol) in DCM (5 mL) was added m-CPBA (85% purity, 226 mg, 4.03 mmol) at 0° C. The reaction was stirred at 20° C. for 4 hours. LCMS showed the reaction was completed. The mixture was quenched with saturated aqueous Na2S2O3 (5 mL) and saturated aqueous NaHCO3 (5 mL), then extracted with DCM (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (TFA) to give 2-((4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-5-(methylsulfonyl)-1,3,4-oxadiazole (A7) (74 mg, yield: 31.6%) as colorless oil. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.14 (d, J=8.9 Hz, 2H), 7.11 (s, 2H), 6.95 (d, J=8.8 Hz, 2H), 5.35 (s, 2H), 4.15 (t, J=5.73 Hz, 2H), 3.86 (t, J=6.39 Hz, 2H), 3.50 (s, 3H), 2.28 (t, J=6.06 Hz, 2H), 1.63 (s, 6H). LCMS (220 nm): 97%. LCMS M+H+) m/z: calcd 532.04, found 533.1.
  • Example 4: Synthesis of 5-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)-4-(methylsulfonyl)oxazole (A13)
  • Figure US20200123117A1-20200423-C00325
    Figure US20200123117A1-20200423-C00326
  • Ethyl 4-bromooxazole-5-carboxylate (2)
  • To a solution of ethyl oxazole-5-carboxylate (1) (100.0 g, 0.71 mol) in THF (500 mL) and DMPU (500 mL) was added LiHMDS (1.0 M, 921 mL, 921 mmol) dropwise at −78° C. under N2 atmosphere, and the mixture was stirred for 1.5 hours. Then Br2 (54.5 mL, 1.06 mol) was added dropwise at the same temperature and the resulting mixture was stirred for 0.5 hours. TLC showed the starting material disappeared. The resulting solution was poured into a room temperature saturated aqueous NH4Cl (1000 mL), extracted with EtOAc (500 mL×3). The combined organic layers were washed with brine (500 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give ethyl 4-bromooxazole-5-carboxylate (25.0 g, purity: 90%, yield: 11%) as yellow oil. 1H NMR (400 MHz, CHCl3-d): δ 7.94 (s, 1H), 4.43 (q, J=7.3 Hz, 2H), 1.42 (t, J=7.2 Hz, 3H)
  • Ethyl 4-(methylthio)oxazole-5-carboxylate (3)
  • A mixture of ethyl 4-bromooxazole-5-carboxylate (2) (28.0 g, 127 mmol) and NaSMe (10.8 g, 153 mmol) in EtOH (400 mL) was stirred at 80° C. for 2 hours. LCMS showed the reaction was completed. The resulting solution was cooled down and concentrated under reduced pressure. The residue was diluted with water (300 mL) and extracted with EtOAc (300 mL×3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure The residue was purified by MPLC to give ethyl 4-methylsulfanyloxazole-5-carboxylate (13.0 g, yield: 80%, yield: 43.7%) as yellow oil. 1H NMR (400 MHz, CHCl3-d): δ 7.91 (s, 1H), 4.40 (q, J=7.2 Hz, 2H), 2.62 (s, 3H), 1.41 (t, J=7.2 Hz, 3H).
  • Ethyl 4-(methylsulfonyl)oxazole-5-carboxylate (4)
  • A solution of ethyl 4-methylsulfanyloxazole-5-carboxylate (3) (2.9 g, 15.5 mmol) and m-CPBA (10.0 g, 46.5 mmol) in DCM (10 mL) was stirred at 25° C. for 16 hours. The reaction was completed detected by TLC. The reaction was quenched by sat. aqueous Na2S2O3 (100 mL) and sat. aqueous NaHCO3 (100 mL). The mixture was extracted with DCM (60 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give ethyl 4-methylsulfonyloxazole-5-carboxylate (1.20 g, yield: 35.3%) as colorless oil. 1H NMR (400 MHz, CHCl3-d): δ 8.04 (s, 1H), 4.50 (q, J=7.1 Hz, 2H), 3.38 (s, 3H), 1.44 (t, J=7.2 Hz, 3H).
  • (4-(Methylsulfonyl)oxazol-5-yl)methanol (5)
  • To a solution of ethyl 4-methylsulfonyloxazole-5-carboxylate (4) (1.2 g, 5.47 mmol) in THF (10 mL) was added LiBH4 (179 mg, 8.21 mmol) at 0° C. The mixture was stirred at 0° C. for 1 hour. TLC showed the reaction completed. The mixture was quenched with aqueous HCl solution (3M, 2 mL) at 0° C. and extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give (4-methylsulfonyloxazol-5-yl)methanol (900 mg, yield: 92.8%) as colorless oil. 1H NMR (400 MHz, CHCl3-d): δ=7.92 (s, 1H), 4.93 (s, 2H), 3.24 (s, 3H).
  • 5-(Chloromethyl)-4-(methylsulfonyl)oxazole (6)
  • To a solution of ethyl 4-methylsulfonyloxazole-5-carboxylate (5) (700 mg, 3.95 mmol) in DCM (10 mL) and DMF (0.5 mL) was added SOCl2 (1.44 mL, 19.8 mmol) at 0° C. The mixture was stirred at 25° C. for 2 hours. TLC showed the reaction completed. The mixture was quenched with H2O (15 mL), extracted with DCM (10 mL×3). The combined organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give 5-(chloromethyl)-4-methylsulfonyl-oxazole (700 mg, yield: 90.6%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=7.99 (s, 1H), 4.94 (s, 2H), 3.22 (s, 3H).
  • Methyl 3,5-dichloro-4-(2-chloroethoxy)benzoate (9)
  • A mixture of methyl 3,5-dichloro-4-hydroxy-benzoate (7) (10.0 g, 45.2 mmol), Cs2CO3 (36.9 g, 113 mmol) and 1-bromo-2-chloro-ethane (26.0 g, 181 mmol) in DMF (60.0 mL) was stirred at 70° C. for 1 hour. TLC showed the reaction completed. The resulting solution was cooled down and diluted with water (150 mL), extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give methyl 3,5-dichloro-4-(2-chloroethoxy)benzoate (10.0 g, yield: 78.0%) as a yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=8.01-7.97 (m, 2H), 4.35 (t, J=6.2 Hz, 2H), 3.93 (s, 3H), 3.89 (t, J=6.2 Hz, 2H).
  • 2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-ol (10)
  • To a solution of MeMgBr (3 M, 65.6 mL, 197 mmol) in THF (30 mL) was added methyl 3,5-dichloro-4-(2-chloroethoxy)benzoate (9) (9.3 g, 32.8 mmol) in THF (50 mL) dropwise at 0° C. The reaction was stirred at 0° C. for 1 hour and TLC showed the reaction completed. The mixture was added to sat. aqueous NH4Cl solution (300 mL) and extracted with EtOAc (200 mL×3). The combined organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated to give 2-[3,5-dichloro-4-(2-chloroethoxy)phenyl]propan-2-ol (9.0 g, yield: 96.8%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=7.42 (s, 2H), 4.27 (t, J=6.3 Hz, 2H), 3.87 (t, J=6.3 Hz, 2H), 1.55 (s, 6H).
  • 4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenol (11)
  • To a solution of 2-[3,5-dichloro-4-(2-chloroethoxy)phenyl]propan-2-ol (9.0 g, 31.7 mmol) (10) and phenol (3.58 g, 38.1 mmol) in CCl4 (100 mL) was added BF3-Et2O (7.83 mL, 63.5 mmol) dropwise at 0° C. under N2 atmosphere. The mixture was stirred at 0° C. for 1 h. LCMS TLC showed the reaction almost completed. The mixture was added to water (150 mL) and extracted with DCM (100 mL×3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated. The crude product was purified by reverse phase MPLC (neutral) to give 4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenol (7.0 g, yield: 61.3%) as yellow oil. 1H NMR (400 MHz, CHLOROFORM-d) δ=7.13 (s, 2H), 7.10-7.04 (m, 2H), 6.79-6.74 (m, 2H), 4.81 (br s, 1H), 4.26 (t, J=6.3 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.62 (s, 6H).
  • 5-((4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)-4-(methylsulfonyl)oxazole (A13)
  • A solution of 5-(chloromethyl)-4-methylsulfonyl-oxazole (6) (500 mg, 2.56 mmol), 4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenol (11) (919 mg, 2.56 mmol) and Cs2CO3 (1.67 g, 5.11 mmol) in DMF (20 mL) was stirred at 25° C. for 2 hours. Then the resulting solution was stirred at 40° C. for 0.5 hr. The reaction was completed detected by TLC. The reaction was quenched with water (50 mL), and the mixture was extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (30 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by MPLC to give 5-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-methylsulfonyl-oxazole (528 mg, yield: 39.8%) as white solid. 1H NMR (400 MHz, CHLOROFORM-d) δ=7.92 (s, 1H), 7.09-7.02 (m, 4H), 6.89-6.83 (m, 2H), 5.34 (s, 2H), 4.18 (t, J=6.4 Hz, 2H), 3.79 (t, J=6.4 Hz, 2H), 3.11 (s, 3H), 1.54 (s, 6H). MS(M+H+) m/z: clcd. 517.0. found 518.1, 540.0.
  • Example 5: Synthesis of N-((3-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl) isoxazol-5-yl)methyl)methanesulfonamide (A22)
  • Figure US20200123117A1-20200423-C00327
  • 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl trifluoromethane-sulfonate (2)
  • To a solution of 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenol (1) (4.0 g, 10.7 mmol) and Et3N (4.5 mL, 54.7 mmol) in DCM (40 mL) was added trifluoromethanesulfonic anhydride (5.4 g, 19.3 mmol) dropwise at 0° C. under N2 atmosphere. The mixture was stirred at the same temperature for 1.5 hrs. TLC showed the reaction was completed. The mixture was poured into H2O (60 mL), extracted with DCM (20 mL×3). The combined organic layers were washed with brine (20 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl trifluoromethanesulfonate (4.0 g, yield: 73.4%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.29-7.27 (m, 2H), 7.22-7.19 (m, 2H), 7.11 (s, 2H), 4.17 (t, J=6.3 Hz, 2H), 3.87 (t, J=6.3 Hz, 2H), 3.87 (t, J=6.2 Hz, 2H), 1.66 (s, 6H).
  • 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)benzaldehyde (3)
  • To a solution of 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl trifluoromethanesulfonate (6) (4.0 g, 7.9 mmol), Et3SiH (1.9 mL, 11.9 mmol) and Et3N (2.2 mL, 15.8 mmol) in DMF (50 mL) was added Pd(dppf)Cl2 (290 mg, 0.4 mmol) at 20° C. The reaction was stirred at 60° C. for 16 hours under CO atmosphere at 50 psi. TLC showed the reaction was completed. The resulting mixture was cooled down, poured into H2O (100 mL), extracted with EtOAc (40 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)benzaldehyde (2.6 g, yield: 85.2%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 10.02 (s, 1H), 7.83 (d, J=8.8 Hz, 2H), 7.41 (d, J=8.8 Hz, 2H), 7.13 (s, 2H), 4.18 (t, J=5.8 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 2.31 (m, 2H), 1.70 (s, 6H).
  • (E)-4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)benzaldehyde oxime (4)
  • To a solution of 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)benzaldehyde (0.5 g, 1.3 mmol) (3) in DCM (5 mL) was added NH2OH.HCl (117 mg, 1.7 mmol) and TEA (0.5 mL, 3.9 mmol) at 20° C. under N2 atmosphere. The reaction was stirred at the same temperature for 16 hrs. LCMS showed the reaction was completed. The mixture was poured into H2O (10 mL) and extracted with DCM (10 mL×3). The organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)benzaldehyde oxime (4) (0.410 g, yield: 78.9%) as white solid. 1H NMR (400 MHz, CHCl3-d) δ=8.13 (s, 1H), 7.52 (d, J=8.4 Hz, 2H), 7.37 (s, 1H), 7.23 (d, J=8.4 Hz, 2H), 7.12 (s, 2H), 4.15 (t, J=5.6 Hz, 2H), 3.86 (t, J=6.5 Hz, 2H), 2.28 (quin, J=6.1 Hz, 2H), 1.65 (s, 6H). MS(M+H+) m/z: clcd. 399.0. found 399.8.
  • tert-Butyl ((3-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl)isoxazol-5-yl)methyl)carbamate (6)
  • To a solution of 4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl) propan-2-yl)benzaldehyde oxime (4) (320 mg, 0.80 mmol) in DCM (10 mL) was added aqueous NaClO (7.0%, 1.3 g, 1.2 mmol) dropwise at 0° C. The reaction was stirred for 5 min. Tert-butyl prop-2-yn-1-ylcarbamate (5) (120 mg, 0.80 mmol) and TEA (0.22 mL, 1.6 mmol) were added to the reaction mixture at 0° C. The mixture was stirred at 0° C.-25° C. for 16 hours. LCMS showed ˜50% of desired product and ˜10% of starting material 2 remained. The mixture was poured into H2O (10 mL), extracted with DCM (10 mL×2). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure and purified by MPLC to give tert-butyl ((3-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl)isoxazol-5-yl)methyl) carbamate (0.22 g, yield: 51%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ=7.72 (d, J=8.60 Hz, 2H) 7.31 (d, J=8.60 Hz, 2H) 7.10-7.20 (m, 2H) 6.48 (s, 1H) 5.03 (br s, 1H) 4.48 (br d, J=6.17 Hz, 2H) 4.16 (t, J=5.73 Hz, 2H) 3.86 (t, J=6.39 Hz, 2H) 2.29 (q, J=6.06 Hz, 2H) 1.68 (s, 6H) 1.47 (s, 9H). MS(M+H+) m/z: clcd. 552.1. found 553.2.
  • (3-(4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl)isoxazol-5-yl) methanamine hydrochloride (7)
  • A solution of tert-butyl ((3-(4-(2-(3,5-dichloro-4-(3-chloropropoxy) phenyl)propan-2-yl)phenyl)isoxazol-5-yl)methyl)carbamate (6) (190 mg, 0.34 mmol) in HCl/EtOAc (4 M, 3 mL, 12 mmol) was stirred at 20° C. for 0.5 hour. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure to give (3-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl)isoxazol-5-yl)methanamine hydrochloride (7) (60 mg, yield: 36%) as brown oil. MS(M+H+) m/z: clcd. 452.1. found 453.1.
  • N-((3-(4-(2-(3,5-Dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl)isoxazol-5-yl)methyl)methanesulfonamide (A22)
  • To a solution of [3-[4-[1-[3,5-dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenyl]isoxazol-5-yl]methanamine (7) (60 mg, 0.13 mmol) in DCM (3 mL) was added TEA (40 mg, 0.40 mmol) and MsCl (18 mg, 0.16 mmol) under N2 atmosphere at 0° C. The reaction was stirred at 20° C. for 5 hrs. TLC showed the reaction was completed. The mixture was poured into H2O (5 mL) and extracted with DCM (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (TFA) to give N-((3-(4-(2-(3,5-dichloro-4-(3-chloropropoxy)phenyl)propan-2-yl)phenyl)isoxazol-5-yl)methyl) methanesulfonamide (A22) (5 mg, yield: 7.11%) as brown oil. LCMS purity (220 nm): 89.4%. 1H NMR (400 MHz, CHCl3-d) δ=7.73 (br d, J=7.9 Hz, 2H), 7.31 (br d, J=8.2 Hz, 2H), 7.14 (s, 2H), 6.60 (s, 1H), 4.89-4.80 (m, 1H), 4.54 (d, J=6.2 Hz, 2H), 4.16 (t, J=5.6 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 2.99 (s, 3H), 2.33-2.25 (m, 2H), 1.68 (s, 6H). LCMS(M+H+) m/z: clcd. 530.0. found 531.0.
  • Example 6: Synthesis of N-(tert-Butyl)-3,5-dichloro-4-(2-chloroethoxy)-N-(4-((4-(methyl-sulfonyl)oxazol-5-yl)methoxy)phenyl)aniline (A31)
  • Figure US20200123117A1-20200423-C00328
    Figure US20200123117A1-20200423-C00329
  • tert-Butyl-(4-iodophenoxy)-dimethyl-silane (2)
  • To a solution of 4-iodophenol (1) (3.0 g, 13.6 mmol) and imidazole (1.9 g, 27.3 mmol) in DCM (30 mL) was added TBSCl (1.8 g, 12.3 mmol) at 25° C. Then the mixture was stirred at the same temperature for 16 hours. TLC showed the reaction was completed. The reaction mixture was washed with water (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give tert-butyl-(4-iodophenoxy)-dimethyl-silane (2) (2.5 g, yield: 54.8%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ 7.57-7.44 (m, 2H), 6.67-6.55 (m, 2H), 1.02-0.93 (m, 9H), 0.19 (s, 6H).
  • 3,5-Dichloro-4-(2-chloroethoxy)aniline (5)
  • To a solution of 4-amino-2,6-dichloro-phenol (3) (5.0 g, 28.1 mmol) in DMF (50 mL) was added NaH (60% purity, 1.1 g, 28.1 mmol) in portions at 0° C. under N2 atmosphere. After stirring at the same temperature for 15 min, 1-bromo-2-chloro-ethane (4) (4.4 g, 30.9 mmol) was added and the resulting mixture was stirred at 25° C. for 16 hours. TLC showed the reaction was completed. The resulting mixture was quenched with saturated aqueous NH4Cl solution (100 mL) and extracted with EtOAc (50 mL×3). The combined organic layers were washed with brine (50 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 3,5-dichloro-4-(2-chloroethoxy)aniline (5) (2.5 g, yield: 37%) as yellow solid. 1H NMR (400 MHz, CHCl3-d) δ 6.64-6.55 (m, 2H), 4.18 (t, J=6.4 Hz, 2H), 3.84 (t, J=6.4 Hz, 2H), 3.69-3.58 (m, 2H).
  • N-tert-butyl-3,5-dichloro-4-(2-chloroethoxy)aniline (7)
  • To a solution of 3,5-dichloro-4-(2-chloroethoxy)aniline (5) (1.0 g, 4.16 mmol) and tert-butyl 2,2,2-trichloroethanimidate (6) (2.27 g, 10.4 mmol) in MeNO2 (10 mL) was added Copper(II) trifluoromethanesulfonate (1.5 g, 4.16 mmol) under N2 atmosphere. Then the resulting mixture was stirred at 25° C. for 16 hours. TLC showed the reaction completed. The mixture was quenched with aqueous NaHCO3 solution (5 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give the N-tert-butyl-3,5-dichloro-4-(2-chloroethoxy)aniline (7) (460 mg, yield: 37.3%) as a brown gum. 1H NMR (400 MHz, CHCl3-d) δ 6.64 (s, 2H), 4.19 (t, J=6.4 Hz, 2H), 3.84 (t, J=6.4 Hz, 2H), 1.37-1.29 (m, 9H).
  • N-tert-Butyl-N-[4-[tert-butyl(dimethyl)silyl]oxyphenyl]-3,5-dichloro-4-(2-chloroethoxy)aniline (8)
  • To a mixture of N-tert-butyl 3,5-dichloro-4-(2-chloro ethoxy) aniline (7) (210 mg, 0.71 mmol), tert-butyl-(4-iodophenoxy)-dimethyl-silane (237 mg, 0.71 mmol) and t-BuONa (162 mg, 1.77 mmol) in toluene (5 mL) was added palladium (I) tritertubutyl phosphine bromide (55 mg, 0.07 mmol) under N2 atmosphere. Then the resulting mixture was stirred at 110° C. for 16 hours. TLC showed the reaction was completed. The resulting mixture was cooled down and quenched with water (2 mL) and extracted with EtOAc (2 mL×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give N-tert-butyl-N-[4-[tert-butyl(dimethyl) silyl]oxyphenyl]-3,5-dichloro-4-(2-chloroethoxy)aniline (8): (125 mg, yield: 35.1%) as yellow solid. 1H NMR (400 MHz, CHCl3-d) δ 6.99-6.93 (m, 2H), 6.86-6.77 (m, 2H), 6.70 (s, 2H), 4.18 (t, J=6.4 Hz, 2H), 3.83 (t, J=6.5 Hz, 2H), 1.37 (s, 9H), 1.04-0.94 (m, 9H), 0.26-0.21 (m, 6H).
  • 4-[N-tert-Butyl3,5-dichloro-4-(2-chloroethoxy)anilino]phenol (9)
  • To a solution of N-tert-butylN-[4-[tert-butyl(dimethyl)silyl]oxyphenyl]-3,5-dichloro-4-(2-chloroethoxy)aniline (8) (150 mg, 0.29 mmol) in THF (5 mL) was added TBAF (1 M, 1.0 mL, 1 mmol). Then the resulting mixture was stirred at 25° C. for 2 hours. TLC showed the reaction was completed. The mixture was quenched with water (5 mL) and extracted with EtOAc (3 mL×3). The combined organic layers were washed with water (5 mL×2), dried over Na2SO4, filtered and concentrated to give the 4-[N-tert-butyl3,5-dichloro-4-(2-chloroethoxy)anilino]phenol (9) (110 mg, yield: 94.9%) as yellow gum. 1H NMR (400 MHz, CHCl3-d) δ 7.03-6.93 (m, 2H), 6.87-6.79 (m, 2H), 6.72 (s, 2H), 4.18 (t, J=6.4 Hz, 2H), 3.83 (t, J=6.4 Hz, 2H), 1.37 (s, 9H).
  • N-tert-Butyl-3,5-dichloro-4-(2-chloroethoxy)-N-[4-[(4-methylsulfonyloxazol-5-yl)-methoxy]-phenyl]aniline (A31)
  • To a mixture of 4-[N-tert-butyl-3,5-dichloro-4-(2-chloroethoxy)anilino]phenol (9) (110 mg, 0.283 mmol) and Cs2CO3 (277 mg, 0.85 mmol) in DMF (5 mL) was added 5-(chloromethyl)-4-methylsulfonyl-oxazole (10) (83 mg, 0.42 mmol). Then the resulting mixture was stirred at 40° C. for 2 hours. LCMS showed the reaction was completed. The mixture was cooled down, quenched with water (5 mL) and extracted with EtAOc (5 mL×3). The combined organic layers were washed with brine (5 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (TFA) to give the N-tert-butyl-3,5-dichloro-4-(2-chloroethoxy)-N-[4-[(4-methylsulfonyl-oxazol-5-yl)-methoxy]-phenyl]aniline (A31) (36.5 mg, yield: 23.5%) as yellow solid. HPLC purity (220 nm): 91.7%. 1H NMR (400 MHz, CHCl3-d) δ 8.01 (s, 1H), 7.06-7.02 (m, 2H), 6.99-6.94 (m, 2H), 6.73 (s, 2H), 5.41 (s, 2H), 4.17 (t, J=6.4 Hz, 2H), 3.82 (t, J=6.4 Hz, 2H), 3.20 (s, 3H), 1.35 (s, 9H). LCMS (M+Na+) m/z: calcd 546.1. found 569.1.
  • Example 7: Synthesis of 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)-N-((4-(methylsulfonyl)oxazol-5-yl)methyl)aniline hydrochloride (A32)
  • Figure US20200123117A1-20200423-C00330
  • 4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)-N-(diphenylmethylene)aniline (3)
  • To a mixture of 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl trifluoromethanesulfonate (1) (5.0 g, 10.2 mmol), diphenylmethanimine (2) (1.8 g, 10.2 mmol), Cs2CO3 (6.6 g, 20.3 mmol) and BINAP (1.3 g, 5.08 mmol) in 1,4-dioxane (100 mL) was added Pd(OAc)2 (0.5 g, 2.0 mmol) at 25° C. under N2 atmosphere. The mixture was stirred at 65° C. for 2 hrs. TLC showed the reaction was completed. The resulting solution was cooled down, quenched with H2O (100 mL), extracted with EtOAc (30 mL×2). The combined organic layers were washed with brine (40 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)-N-(diphenylmethylene)aniline (4.5 g, yield: 84.6%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ 7.78-7.74 (m, 2H), 7.63-7.57 (m, 2H), 7.45-7.38 (m, 2H), 7.32-7.27 (m, 2H), 7.14-7.07 (m, 2H), 7.03-6.94 (m, 4H), 6.66 (d, J=8.6 Hz, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.58 (s, 6H).
  • 4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)aniline hydrochloride (4)
  • To a solution of 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)-N-(diphenyl methylene)aniline (3) (4.5 g, 8.6 mmol) in THF (50 mL) was added aqueous HCl (2 M, 15 mL) and the mixture was stirred at 25° C. for 15 min. TLC showed the reaction was completed. The mixture was concentrated under reduced pressure and purified by silica-gel column chromatography to give 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)aniline hydrochloride (2.5 g, yield: 81.0%) as yellow solid. 1H NMR (400 MHz, CHCl3-d) δ 10.54 (br s, 2H), 7.48 (br d, J=8.6 Hz, 2H), 7.31-7.27 (m, 2H), 7.09 (s, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.3 Hz, 2H), 1.63 (s, 6H).
  • 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)-N-((4-(methylsulfonyl)oxazol-5-yl)methyl)aniline hydrochloride (A32)
  • To a suspension of 5-(chloromethyl)-4-(methylsulfonyl)oxazole (5) (200 mg, 0.5 mmol) and Ag2CO3 (564 mg, 0.2 mmol) in DMF (2 mL) was added 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)aniline (4) (382 mg, 0.1 mmol), and the mixture was stirred at 65° C. for 2 hours. TLC showed the reaction was completed. The resulting mixture was cooled down, poured into H2O (6 mL), extracted with EtOAc (2 mL×2). The combined organic layers were washed with brine (4 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (HCl) to give 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)-N-((4-(methylsulfonyl)oxazol-5-yl) methyl)aniline hydrochloride (A32) (20 mg, yield: 3.8%) as white solid. 1H NMR (400 MHz, CHCl3-d) δ 7.85 (s, 1H), 7.16-7.09 (m, 4H), 7.04-6.93 (m, 2H), 4.80 (s, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.16 (s, 3H), 1.61 (s, 6H). LCMS (M+H+) m/z: calcd: 516.0. found 517.0.
  • Example 8: Synthesis of 5-(1-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl) phenoxy)ethyl)-4-(methylsulfonyl)oxazole (A35)
  • Figure US20200123117A1-20200423-C00331
  • 4-(Methylthio)oxazole-5-carboxylic acid (2)
  • To a solution of ethyl 4-methylsulfanyloxazole-5-carboxylate (1) (1 g, 80% purity, 4.3 mmol) in THF (10 mL) and H2O (4 mL) was added NaOH (300 mg, 8.6 mmol) at 25° C., and the mixture was stirred at the same temperature for 2 hours. LCMS showed the reaction was completed. The mixture was acidified to pH=3 with aqueous HCl solution (3 M, 3 mL) and extracted with EtOAc (15 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 4-(methylthio)oxazole-5-carboxylic acid (2) (0.7 g, 80% purity, yield: 82.3%) as white solid. 1H NMR (400 MHz, CHCl3-d) δ 7.99 (s, 1H), 2.65 (s, 3H). LCMS (M+1) m/z: calcd: 531.0. found 532.0.
  • N-Methoxy-N-methyl-4-(methylthio)oxazole-5-carboxamide (4)
  • To a mixture of 4-(methylthio)oxazole-5-carboxylic acid (2) (0.7 g, 4.4 mmol) in THF (5 mL) and DMF (5 mL) was added HATU (2.5 g, 6.6 mmol) at 25° C. under N2 atmosphere. After stirring at the same temperature for 15 minutes, N-methoxymethanamine hydrochloride (3) (0.6 g, 5.7 mmol) and TEA (3.1 mL, 22.0 mmol) were added and the resulting mixture stirred at 25° C. for 2 hours. TLC showed the reaction was completed. The reaction was diluted with H2O (10 mL), extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give N-methoxy-N-methyl-4-(methylthio)oxazole-5-carboxamide (4) (0.5 g, yield: 56.2%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.90 (s, 1H), 4.12 (q, J=7.1 Hz, 1H), 3.81 (s, 3H), 3.30 (s, 3H), 2.58 (s, 3H).
  • 1-(4-(Methylthio)oxazol-5-yl)ethenone (5)
  • To a mixture of N-methoxy-N-methyl-4-(methylthio)oxazole-5-carboxamide (4) (0.4 g, 1.9 mmol) in THF (8 mL) was added MeMgBr (3 M, 0.8 mL, 2.4 mmol) at 0° C. under N2 atmosphere, and the resulting mixture was stirred at the same temperature for 1 hour. TLC showed the reaction was completed. The reaction was quenched with saturated aqueous NH4Cl (4 mL), extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 1-(4-(methylthio)oxazol-5-yl)ethenone (5) (0.4 g, 75% purity, yield: 96.2%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.89 (s, 1H), 2.62 (s, 3H), 2.50 (s, 3H).
  • 1-(4-(Methylthio)oxazol-5-yl)ethanol (6)
  • To a mixture of 1-(4-(methylthio)oxazol-5-yl)ethanone (5) (0.4 g, 2.3 mmol) in THF (8 mL) was added NaBH4 (0.1 g, 2.7 mmol) at 0° C. under N2 atmosphere, and the mixture was stirred at the same temperature for 1 hour. TLC showed the reaction was completed. The reaction was quenched with saturated aqueous NH4Cl (4 mL), extracted with EtOAc (5 ml×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 1-(4-(methylthio)oxazol-5-yl)ethanol (6) (0.2 g, yield: 54.9%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.83 (s, 1H), 5.12 (br d, J=5.5 Hz, 1H), 2.44 (s, 3H), 1.58 (d, J=6.6 Hz, 3H).
  • 5-(1-(4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)ethyl)-4-(methylthio)oxazole (8)
  • To a mixture of 1-(4-(methylthio)oxazol-5-yl)ethanol (6) (0.2 g, 1.1 mmol), 4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenol (7) (0.4 g, 1.1 mmol) and PPh3 (0.6 g, 2.3 mmol) in THF (2 mL) was added DEAD (0.3 g, 1.7 mmol) at 0° C. under N2 atmosphere, and the mixture was stirred at 25° C. for 16 hours. TLC showed the reaction was completed. The reaction was quenched with H2O (5 mL), extracted with EtOAc (6 mL×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give the 5-(1-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)ethyl)-4-(methylthio)oxazole (8) (50 mg, yield: 8.83%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.85-7.82 (m, 1H), 7.11-7.03 (m, 4H), 6.88 (d, J=8.8 Hz, 2H), 5.51 (q, J=6.6 Hz, 1H), 4.24 (t, J=6.4 Hz, 2H), 3.84 (t, J=6.4 Hz, 2H), 2.35 (s, 3H), 1.69 (d, J=6.6 Hz, 3H), 1.58 (s, 6H).
  • 5-(1-(4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)ethyl)-4-(methylsulfonyl)oxazole (A35)
  • To a mixture of 5-(1-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)ethyl)-4-(methylthio)oxazole (8) (50 mg, 0.1 mmol) was added mCPBA (80% purity, 64 mg, 0.3 mmol) in DCM (3 mL) at 25° C., and the mixture was stirred at the same temperature for 16 hours. LCMS showed the reaction was completed. The reaction was quenched with H2O (5 mL), extracted with EtOAc (6 mL×3). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (TFA) 5-(1-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)ethyl)-4-(methylsulfonyl)oxazole (21.7 mg, yield: 40.8%) as white solid. HPLC purity (220 nm): 98.5%. 1H NMR (400 MHz, CHCl3-d) δ=7.94 (s, 1H), 7.14-7.03 (m, 4H), 6.92 (d, J=8.9 Hz, 2H), 6.10 (q, J=6.5 Hz, 1H), 4.26 (t, J=6.3 Hz, 2H), 3.86 (t, J=6.3 Hz, 2H), 3.06 (s, 3H), 1.74 (d, J=6.7 Hz, 3H), 1.59 (s, 6H). LCMS (M+H+) m/z: calcd: 531.0. found 532.0, 534.0.
  • Example 9: Synthesis of N-(4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)oxazol-2-yl)methanesulfonamide (A38)
  • Figure US20200123117A1-20200423-C00332
  • (2-Chlorooxazol-4-yl)methanol (2)
  • To a solution of ethyl 2-chlorooxazole-4-carboxylate (1) (1.0 g, 5.7 mmol) in DCM (20 mL) was added DIBAL-H (1 M in toluene 8.5 mL, 8.5 mmol) dropwise at 0° C. under N2 atmosphere. The mixture was stirred at the same temperature for 1 hour. TLC showed the reaction was completed. The mixture was quenched with saturated aqueous NH4Cl (20 mL), then extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by MPLC to give (2-chlorooxazol-4-yl)methanol (2) (470 mg, yield: 62%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ 7.59 (s, 1H), 4.58 (d, J=0.8 Hz, 2H).
  • (2-Chlorooxazol-4-yl)methyl methanesulfonate (3)
  • To a mixture of (2-chlorooxazol-4-yl)methanol (2) (0.47 g, 3.52 mmol) and TEA (1 mL, 7.04 mol) in DCM (15 mL) was added methanesulfonyl chloride (444 mg, 3.87 mmol) at 0° C. The mixture was stirred at 0-25° C. for 3 hours. TLC showed the reaction was completed. The mixture was poured into H2O (30 mL), extracted with DCM (10 mL×2). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduce pressure to give (2-chlorooxazol-4-yl)methyl methanesulfonate (3) (800 mg, 70% purity) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.77 (s, 1H), 5.14 (d, J=0.8 Hz, 2H), 3.08 (s, 3H).
  • 2-Chloro-4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)oxazole (5)
  • To a solution of (2-chlorooxazol-4-yl)methyl methanesulfonate (3) (0.70 g, 3.31 mmol) in DMF (10 mL) was added 4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenol (4) (1.19 g, 3.31 mmol) and Cs2CO3 (2.16 g, 6.62 mmol). The resulting mixture was stirred at 25° C. for 10 hours. LCMS showed the reaction was completed. The mixture was poured into H2O (30 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (20 mL×3), dried over Na2SO4, filtered and concentrated under reduce pressure. The residue was purified by silica gel column chromatography to give 2-chloro-4-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]oxazole (0.50 g, yield: 31.8%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 7.68 (s, 1H), 7.15-7.11 (m, 4H), 6.92-6.87 (m, 2H), 4.97 (d, J=0.8 Hz, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.63 (s, 6H). LCMS (M+H+) m/z: calcd: 473.0. found 474.1.
  • N-(4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)oxazol-2-yl)methanesulfonamide (A38)
  • To a solution of 2-chloro-4-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]oxazole (5) (10 mg, 0.02 mmol) in 1,4-dioxane (0.2 mL) was added methanesulfonamide (2.4 mg, 0.02 mmol), Brettphos Pd G3 (2 mg, w20%) and t-BuONa (3 mg, 0.03 mmol). The mixture was stirred at 80° C. for 10 hours under N2 atmosphere. LCMS showed 5% desired MS and 90% starting material. The resulting 20 reaction mixtures were cooled down and combined. The mixture was filtered and the filtrate concentrated under reduced pressure. The residue was purified by prep-HPLC (TFA) to give N-[[5-bromo-4-[4-[1-[3,5-dichloro-4-(3-chloropropoxy)phenyl]-1-methyl-ethyl]phenyl]oxazol-2-yl]methyl]methanesulfonamide (2 mg, yield: 1.8%) as pale yellow solid. LCMS (220 nm): 85.79%. 1H NMR (400 MHz, CHCl3-d) δ 7.16 (d, J=8.8 Hz, 2H), 7.12 (s, 2H), 7.08 (s, 1H), 6.86 (d, J=8.8 Hz, 2H), 4.86 (s, 2H), 4.27 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.09 (s, 3H), 1.64 (s, 6H). LCMS (M+H+) m/z: calcd: 532.04. found 533.0.
  • Example 10: Synthesis of N-[3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1H-pyrazol-4-yl]methanesulfonamide (A40)
  • Figure US20200123117A1-20200423-C00333
    Figure US20200123117A1-20200423-C00334
  • Methyl 4-nitro-1-tetrahydropyran-2-yl-pyrazole-3-carboxylate (3)
  • To a solution of methyl 4-nitro-1H-pyrazole-5-carboxylate (1) (2.00 g, 1.17 mmol) in DCM (20 mL) was added 3,4-dihydro-2H-pyran (2) (3.11 g, 14.0 mmol) and p-Toluenesulfonic acid (0.20 g, 1.17 mmol) at 20° C. The mixture was stirred at the same temperature for 4 hours. LCMS showed the reaction was completed. The reaction was poured into water (100 mL) and extracted with DCM (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give methyl 4-nitro-1-tetrahydropyran-2-yl-pyrazole-3-carboxylate (3) (2.7 g, yield: 90.5%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ ppm 8.39 (s, 1H), 5.44 (dd, J=9.04, 2.87 Hz, 1H), 4.06-4.15 (m, 1H), 4.00 (s, 3H), 3.74 (ddd, J=11.69, 9.04, 4.41 Hz, 1H), 2.23 (dd, J=12.79, 3.09 Hz, 1H), 1.86-2.07 (m, 3H), 1.63-1.77 (m, 4H), 1.57 (s, 6H). LCMS (220 nm): 68% Exact Mass 255.09; found 256.0.
  • (4-nitro-1-tetrahydropyran-2-yl-pyrazol-3-yl) methanol (4)
  • To a solution of methyl 4-nitro-1-tetrahydropyran-2-yl-pyrazole-3-carboxylate (3) (1.00 g, 3.92 mmol) in THF (10 mL) was added DIBALH (7.8 mL, 7.8 mmol, 1.0 mol/L in toluene) at −78° C. under N2 atmosphere and the mixture was stirred at 20° C. for 4 hours. LCMS showed the reaction was completed. The mixture was quenched with water (20 mL) and extracted with EtOAc (30 mL×5). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (4-nitro-1-tetrahydropyran-2-yl-pyrazol-3-yl)methanol (70.0%, 0.90 g, yield: 70.8%) as colorless oil. HPLC purity (220 nm): 69%. 1H NMR (400 MHz, CHCl3-d) δ ppm 8.39 (s, 1H), 5.37 (dd, J=9.17, 2.69 Hz, 1H), 4.93 (s, 2H), 4.06-4.14 (m, 1H), 3.70-3.77 (m, 1H), 2.13-2.22 (m, 1H), 1.87-2.09 (m, 2H), 1.64-1.77 (m, 4H). LCMS (M+H+) m/z: calcd: 227.09. found 250.2.
  • 3-(Chloromethyl)-4-nitro-1-tetrahydropyran-2-yl-pyrazole (5)
  • To a solution of (4-nitro-1-tetrahydropyran-2-yl-pyrazol-3-yl) methanol (4) (1.3 g, 5.72 mmol) in DCM (13 mL) was added triethylamine (1.2 g, 1.14 mmol) at 20° C. Methanesulfonyl chloride (0.8 g, 6.87 mmol) was dropwise at 0° C. The reaction was stirred at 20° C. for 4 hours. TLC showed the reaction was completed. The mixture was quenched with water (30 mL) and extracted with DCM (30 mL×3). The combined organic layers were washed with brine (40 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 3-(chloromethyl)-4-nitro-1-tetrahydropyran-2-yl-pyrazole (5) (60.0%, yield: 30.9%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ ppm 8.39 (s, 1H), 5.38 (dd, J=9.26, 2.65 Hz, 1H), 4.88 (s, 2H), 3.75 (m, 2H), 1.86 (m, 2H), 1.67-1.77 (m, 4H).
  • 3-[[4-[1-[3,5-Dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-nitro-1-tetrahydropyran-2-yl-pyrazole (7)
  • To a solution of 3-(chloromethyl)-4-nitro-1-tetrahydropyran-2-yl-pyrazole (5) (0.20 g, 0.814 mmol) in DMF (2 mL) was added CS2CO3 (0.4 g, 1.22 mmol) and 4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenol (0.18 g, 0.49 mmol) (6) at 20° C. The mixture was stirred for 2 hours at 60° C. 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×5), dried over Na2SO4, filtered and concentrated under reduced pressure to give 3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-nitro-1-tetrahydropyran-2-yl-pyrazole 3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-nitro-1-tetrahydropyran-2-yl-pyrazole (7) (50.0% purity, 0.20 g, yield: 21.6%) as colorless oil. LCMS (M+Na+) m/z: calcd: 567.1. found 590.2.
  • 3-[[4-[1-[3,5-Dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1-tetrahydropyran-2-yl-pyrazol-4-amine (8)
  • To a solution of 3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-nitro-1-tetrahydropyran-2-yl-pyrazole (7) (1.0 g, 1.76 mmol) in THF/H2O (6 mL/3 mL) was added NH4Cl (0.282 g, 5.27 mmol) at 20° C., Zn (0.575 g, 8.79 mmol) was added to the mixture at 0° C. The mixture was stirred at 20° C. for 8 hours. 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 (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC (TFA) to give the 3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1-tetrahydropyran-2-yl-pyrazol-4-amine (8) (60 mg, yield: 6.3%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ ppm 7.07-7.17 (m, 4H) 6.89-6.99 (m, 2H), 5.23 (d, J=9.48 Hz, 1H), 5.10 (s, 2H), 4.24-4.30 (m, 2H), 4.05 (d, J=11.47 Hz, 1H), 3.83-3.89 (m, 2H), 3.63-3.72 (m, 1H), 2.06 (m, 2H), 2.02 (s, 1H), 1.62 (s, 6H). LCMS (M+H+) m/z: calcd: 537.14. found 538.2.
  • N-[3-[[4-[1-[3,5-Dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1-tetrahydropyran-2-yl-pyrazol-4-yl]methanesulfonamide (9)
  • To a solution of 3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1-tetrahydropyran-2-yl-pyrazol-4-amine (8) (60 mg, 0.111 mmol) in DCM (0.5 mL) was added pyridine (17.6 mg, 0.223 mmol) at 20° C. Mesyl chloride (0.0129 mL, 0.167 mmol) was added dropwise to the reaction mixture at 0° C. The mixture was stirred at 20° C. for 4 hours. LCMS showed the reaction was completed. The reaction mixture was quenched with water (5 mL) and extracted with DCM (1 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give N-[3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1-tetrahydropyran-2-yl-pyrazol-4-yl]methanesulfonamide (9) (70 mg, yield: 81.5%) as colorless oil. HPLC purity (220 nm): 79%. LCMS (M+Na+) m/z: calcd: 615.1. found 616.2.
  • N-[3-[[4-[1-[3,5-Dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1H-pyrazol-4-yl]methanesulfonamide (A40)
  • A solution of N-[3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1-tetrahydropyran-2-yl-pyrazol-4-yl]methanesulfonamide (9) (70 mg, 0.113 mmol) in HCl/EtOAc (4M, 2 mL) was stirred at 20° C. for 2 hours. LCMS showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give N-[3-[[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-1H-pyrazol-4-yl]methanesulfonamide(A40) (11.6 mg, yield: 18.2%) as white solid. 1H NMR (400 MHz, CHCl3-d) δ ppm 7.71 (s, 1H), 7.10-7.17 (m, 4H), 6.89-6.94 (m, 2H), 6.22 (s, 1H), 5.21 (s, 2H), 4.26 (t, J=6.39 Hz, 2H), 3.86 (t, J=6.28 Hz, 2H), 2.90 (s, 3H), 1.62 (s, 6H). LCMS (M+Na+) m/z: calcd: 531.06. found 532.1.
  • Example 11: Synthesis of N-(4-((4-(2-(3, 5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy) methyl) pyrimidin-5-yl)methanesulfonamide (A41)
  • Figure US20200123117A1-20200423-C00335
  • N-(4-Methylpyrimidin-5-yl)methanesulfonamide (2)
  • To a solution of 4-methylpyrimidin-5-amine (1) (3.0 g, 27.5 mmol) and pyridine (10.9 g, 0.137 mol) in DCM (30 mL) was added methanesulfonyl chloride (6.3 g, 55.0 mmol) at 0° C. and the resulting mixture was stirred at 20° C. for 3 hours. TLC showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give N-(4-methylpyrimidin-5-yl)methanesulfonamide (2) (1.0 g, 19.4%) as yellow solid. 1H NMR (400 MHz, CHCl3-d) δ ppm 8.98 (s, 1H), 8.76 (s, 1H), 6.68 (br s, 1H), 3.13 (s, 3H), 2.61 (s, 3H).
  • tert-Butyl (4-methylpyrimidin-5-yl)(methylsulfonyl)carbamate (3)
  • To a solution of N-(4-methylpyrimidin-5-yl) methanesulfonamide (2) (1.0 g, 5.34 mmol) and TEA (1.1 g, 10.7 mmol), DMAP (196 mg, 1.60 mmol) in THF (15 mL) was added tert-butoxycarbonyl tert-butyl carbonate (1.75 g, 8.01 mmol) at 0° C. and the resulting mixture was stirred at 50° C. for 3 hours. TLC showed the reaction was completed. The resulting mixture was cooled down and quenched with water (20 mL). The mixture was extracted with EtOAc (10 mL×5). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give tert-butyl N-(4-methylpyrimidin-5-yl)-N-methylsulfonyl-carbamate (3) (1.0 g, yield: 65.2%) as yellow solid. 1H NMR (400 MHz, CHCl3-d) δ ppm 9.07 (s, 1H), 8.46 (s, 1H), 3.53 (s, 3H), 2.56 (s, 3H), 1.49 (s, 9H).
  • tert-Butyl (4-(bromomethyl)pyrimidin-5-yl)(methylsulfonyl)carbamate (4)
  • To a mixture of tert-butyl N-(4-methylpyrimidin-5-yl)-N-methylsulfonyl-carbamate (3) (1.0 g, 3.48 mmol) and NBS (929 mg, 5.22 mmol) in CCl4 (20 mL) was added AIBN (495 mg, 3.48 mmol) under N2 atmosphere, and the resulting mixture was stirred at 100° C. for 16 hours. LCMS showed the reaction was completed. The mixture was cooled down and quenched with water (40 mL), and then extracted with DCM (10 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give tert-butyl N-(bromomethyl) pyrimidin-5-yl)-N-methylsulfonyl-carbamate (4) (170 mg, yield: 13.3%) as yellow solid. 1H NMR (400 MHz, CHCl3-d) δ ppm 9.20 (br s, 1H), 8.60 (br s, 1H), 4.59 (d, J=10.64 Hz, 1H), 4.42 (d, J=10.64 Hz, 1H), 3.57-3.60 (m, 3H), 1.48-1.51 (m, 9H). LCMS (M+H+) m/z: 367.0; found 368.0.
  • tert-Butyl (4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy) methyl) pyrimidin-5-yl)(methylsulfonyl)carbamate (6)
  • To a mixture of 4-(1-(3,5-dichloro-4-(2-chloroethoxy)phenyl)-1-methyl-ethyl)phenol (5) (134 mg, 0.37 mmol) and Cs2CO3 (181 mg, 0.56 mmol) in DMF (5 mL) was added tert-butyl N-(4-(bromomethyl)pyrimidin-5-yl)-N-methylsulfonyl-carbamate (4) (170 mg, 0.37 mmol) at 0° C. under N2 atmosphere, and the resulting mixture was stirred at the same temperature for 3 hours. TLC showed the reaction was completed. The mixture was poured into water (10 mL) and extracted with EtOAc (5 mL×5). The combined organic layers were washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give tert-butyl N-(4-((4-(1-(3,5-dichloro-4-(2-chloroethoxy)phenyl)-1-methyl-ethyl)phenoxy)methyl)pyrimidin-5-yl)-N-methylsulfonyl-carbamate (6) (50 mg, yield: 20.9%) as light oil. 1H NMR (400 MHz, CHCl3-d) δ ppm 9.18 (s, 1H), 8.62 (s, 1H), 7.12 (s, 2H), 7.09 (s, 2H), 6.85-6.91 (m, 2H), 5.15-5.30 (m, 2H), 4.24 (t, J=6.28 Hz, 2H), 3.85 (t, J=6.28 Hz, 2H), 3.52 (s, 3H), 1.60 (s, 6H), 1.37 (s, 9H).
  • N-(4-((4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl) pyrimidin-5-yl)methanesulfonamide (A41)
  • A mixture of tert-butyl N-(4-((4-(1-(3,5-dichloro-4-(2-chloroethoxy)phenyl)-1-methyl-ethyl)phenoxy)methyl)pyrimidin-5-yl)-N-methylsulfonyl-carbamate (6) (50 mg, 0.062 mmol) in DCM (5.0 mL) and TFA (0.5 mL) was stirred at 20° C. for 1 hour. LCMS showed the reaction was completed. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC (TFA) to give N-(4-((4-(1-(3,5-dichloro-4-(2-chloroethoxy)phenyl)-1-methyl-ethyl)phenoxy)methyl)pyrimidin-5-yl) methanesulfonamide (A41) (8 mg, yield: 23.7%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ ppm 9.01 (d, J=4.40 Hz, 2H), 7.81 (br s, 1H), 7.16 (d, J=8.93 Hz, 2H), 7.10 (s, 2H), 6.93 (d, J=8.93 Hz, 2H), 5.36 (s, 2H), 4.26 (t, J=6.36 Hz, 2H), 3.86 (t, J=6.36 Hz, 2H), 3.03 (s, 3H), 1.62 (s, 6H). LCMS (M+H+) m/z: 545.05; found 546.0. HPLC purity (220 nm): 84.4%.
  • Example 12: Synthesis of N-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)-2-(methylsulfonamido)oxazole-4-carboxamide (A49)
  • Figure US20200123117A1-20200423-C00336
  • Ethyl 2-(methylsulfonamido)oxazole-4-carboxylate (2)
  • To a solution of methane sulfonamide (3.3 g, 34.2 mmol) in DMF (30 mL) was added NaH (1.3 g, 60% purity, 34.2 mmol) in portions at 0° C. under N2 atmosphere. The mixture was stirred at 0° C. for 0.5 hour, and then ethyl 2-chlorooxazole-4-carboxylate (1) (3.0 g, 17.1 mmol) was added to the mixture. The resulting mixture was stirred at 60° C. for 10 hours. LCMS showed the reaction was completed. The reaction was quenched with H2O (10 mL), and directly purified by reversed phase MPLC (TFA) to give ethyl 2-(methanesulfon-amido)oxazole-4-carboxylate (2) (1.17 g, yield: 29.2%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ 7.61 (s, 1H), 4.39 (q, J=7.2 Hz, 2H), 3.12 (s, 3H), 1.37 (t, J=7.2 Hz, 3H). LCMS (M+H)+ m/z: clcd 234.03. found 235.1.
  • 2-(Methylsulfonamido)oxazole-4-carboxylic acid (3)
  • To a mixture of ethyl 2-(methanesulfonamido)oxazole-4-carboxylate (2) (100 mg, 0.43 mmol) in THF (3 mL) and H2O (1 mL) was added LiOH.H2O (72 mg, 1.7 mmol) at 25° C. The mixture was stirred at the same temperature for 10 hours. LCMS showed the reaction was completed. The mixture was adjusted pH to 4 by aqueous HCl (2 M), and then concentrated under reduce pressure to give 2-(methanesulfonamido)oxazole-4-carboxylic acid (3) (90 mg, 65% purity) as yellow solid. 1H NMR (400 MHz, MeOH-d4) δ 7.94 (s, 1H), 3.08 (s, 3H). LCMS (M−H) m/z: clcd 206.0. found 204.9.
  • N-(4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)-2-(methylsulfonamido)oxazole-4-carboxamide (A49)
  • To a solution of 2-(methane-sulfonamido)oxazole-4-carboxylic acid (3) (60 mg, 0.3 mmol) in DMF (3 mL) was added 4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]aniline (4) (104 mg, 0.3 mmol), HATU (133 mg, 0.35 mmol) and TEA (0.12 mL, 0.9 mmol) at 25° C. The mixture was stirred at the same temperature for 3 hours. LCMS showed the reaction was completed, the mixture was quenched with H2O (1 mL), and directly purified by prep-HPLC (TFA), to give N-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]phenyl]-2-(methanesulfonamido)oxazole-4-carboxamide (A49) (23.2 mg, yield: 14.6%) as white solid.
  • 1H NMR (400 MHz, CHCl3-d) δ 8.44 (s, 1H), 7.91 (s, 1H), 7.56 (d, J=8.8 Hz, 2H), 7.21 (d, J=8.8 Hz, 2H), 7.13 (s, 2H), 4.27 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.32 (s, 3H), 1.65 (s, 6H). LCMS (M+H+) m/z: clcd 545.03. found 546.0.
  • Example 13: Synthesis of 5-((4-(2-(3,5-dichloro-4-(3,3,3-trifluoropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-4-(methylsulfonyl)oxazole (A54)
  • Figure US20200123117A1-20200423-C00337
  • 2,6-Dichloro-4-(2-(4-hydroxyphenyl)propan-2-yl)phenol (3)
  • To a solution of 2,6-dichloro-4-[1-(4-hydroxyphenyl)-1-methyl-ethyl]phenol (1) (0.2 g, 0.67 mmol) in DMF (2 mL) were added 1,1,1-trifluoro-3-iodo-propane (2) (1.5 g, 6.7 mmol) and Cs2CO3 (0.66 g, 2.0 mmol). The resulting reaction was stirred at 60° C. for 16 hours. TLC showed the reaction was completed. The mixture was cooled down and poured into water (10 mL), extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL×3), dried over Na2SO4, filtered and concentrated. The residue was purified by p-TLC to give 4-[1-[3,5-dichloro-4-(3,3,3-trifluoropropoxy)phenyl]-1-methyl-ethyl]phenol (3) (80 mg, yield: 30.2%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ 7.13 (s, 2H), 7.09-7.05 (m, 2H), 6.78-6.75 (m, 2H), 4.71 (s, 1H), 4.21 (t, J=6.8 Hz, 2H), 2.78-2.66 (m, 2H), 1.62 (s, 6H).
  • 5-((4-(2-(3,5-Dichloro-4-(3,3,3-trifluoropropoxy)phenyl)propan-2-yl)phenoxy)methyl)-4-(methylsulfonyl)oxazole (A54)
  • To a mixture of 4-[1-[3,5-dichloro-4-(3,3,3-trifluoro propoxy)phenyl]-1-methyl-ethyl]phenol (3) (40 mg, 0.10 mmol) and 5-(chloromethyl)-4-methylsulfonyl-oxazole (4) (24 mg, 0.12 mmol) in DMF (0.5 mL) was added Cs2CO3 (66 mg, 0.20 mmol) and the mixture was stirred at 25° C. for 16 hours. LCMS showed the reaction was completed. The mixture was poured into water (2 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-TLC to give 5-[[4-[1-[3,5-dichloro-4-(3,3,3-trifluoropropoxy)phenyl]-1-methyl-ethyl]phenoxy]methyl]-4-methylsulfonylo-xazole (A54) (18 mg, yield: 29.9%) as yellow oil. LCMS purity: (220 nm): 93.3%. 1H NMR (400 MHz, CHCl3-d) δ 8.00 (s, 1H), 7.16-7.12 (m, 4H), 6.94 (d, J=8.8 Hz, 2H), 5.42 (s, 2H), 4.22 (t, J=6.8 Hz, 2H), 3.19 (s, 3H), 2.78-2.64 (m, 2H), 1.62 (s, 6H). LCMS (M+NH4+) m/z: calcd 551.1. found 569.0.
  • Example 14: Synthesis of 2-(2-chloroethoxy)-5-(2-(3-cyano-4-((4-(methylsulfonyl)oxazol-5-yl)methoxy) phenyl)propan-2-yl)benzonitrile (A63)
  • Figure US20200123117A1-20200423-C00338
  • Methyl 3-bromo-4-(2-chloroethoxy)benzoate (2)
  • To a solution of methyl 3-bromo-4-hydroxybenzoate (1) (15 g, 65 mmol) in DMF (100 mL) was added 1-bromo-2-chloro-ethane (11 g, 78 mmol) and Cs2CO3 (32 g, 97 mmol) under N2 atmosphere at 20° C. Then the reaction mixture was stirred at the same temperature for 16 hours. TLC showed the reaction was completed. The mixture was poured into H2O (100 mL), extracted with EtOAc (100 mL×2). The combined organic layers were washed with brine (100 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by MPLC to give methyl 3-bromo-4-(2-chloroethoxy)benzoate (2) (13 g, yield: 67%) as white solid. 1H NMR (400 MHz, CHCl3-d) δ 8.29-8.21 (m, 1H), 8.02-7.90 (m, 1H), 6.91 (d, J=8.6 Hz, 1H), 4.35 (t, J=6.1 Hz, 2H), 3.95-3.86 (m, 5H).
  • Methyl 4-(2-chloroethoxy)-3-cyanobenzoate (3)
  • To a solution of methyl 3-bromo-4-(2-chloroethoxy)benzoate (2) (0.6 g, 2.0 mmol) in DMF (20 mL) was added Zn(CN)2 (0.72 g, 6.1 mmol), Xantphos (0.1 g, 0.2 mmol) and Pd2(dba)3 (0.18 g, 0.2 mmol) under N2 atmosphere at 20° C. The reaction mixture was stirred at 130° C. for 3 hours. TLC showed the reaction was completed. Thirteen batches of reaction were combined, and then poured into H2O (50 mL). The mixture was extracted with EtOAc (80 mL×3). The combined organic layers were washed with brine (80 mL×4), dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude product was purified by MPLC to give methyl 4-(2-chloroethoxy)-3-cyanobenzoate (3) (4.60 g, yield: 72.2%) as brown solid. 1H NMR (400 MHz, CHCl3-d) δ 8.27 (s, 1H), 8.22 (dd, J=2.1, 8.9 Hz, 1H), 7.02 (d, J=8.8 Hz, 1H), 4.41 (s, 2H), 3.92 (s, 3H), 3.92-3.88 (m, 2H).
  • 2-(2-Chloroethoxy)-5-(2-hydroxypropan-2-yl)benzonitrile (4)
  • To a solution of MeMgBr (3 M, 24 mL, 72 mmol) in THF (20 mL) was added methyl 4-(2-chloroethoxy)-3-cyanobenzoate (3) (4.3 g, 18 mmol) in THF (40 mL) dropwise under N2 atmosphere at 0° C. Then the reaction was stirred at 20° C. for 3 hours. LCMS showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl (20 mL) and extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over Na2SO4, filtered, concentrated under reduced pressure. The residue was purified by MPLC to give 2-(2-chloroethoxy)-5-(2-hydroxypropan-2-yl)benzonitrile (4) (2.9 g, yield: 67.4%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ=7.71 (d, J=2.4 Hz, 1H), 7.67 (dd, J=2.3, 8.7 Hz, 1H), 6.98-6.92 (m, 1H), 4.34 (t, J=6.1 Hz, 2H), 3.87 (t, J=6.2 Hz, 2H), 1.57 (s, 6H). LCMS (M+H+) m/z: calcd 239.1. found: 240.1.
  • 2-(2-Chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (5)
  • To a mixture of 2-(2-chloroethoxy)-5-(2-hydroxypropan-2-yl)benzonitrile (4) (2.9 g, 12.1 mmol) and phenol (1.4 g, 14.5 mmol) in CCl4 (40 mL) was added BF3-Et2O (3.0 mL, 24 mmol) dropwise under N2 atmosphere at 0° C. Then the reaction was stirred at 20° C. for 3 hours. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NaHCO3 (40 mL), extracted with DCM (20 mL×2). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (5) (2.77 g, yield: 72.5%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ 7.46 (s, 1H), 7.39-7.33 (dd, J=8.8 Hz, 1H), 7.05 (d, J=8.6 Hz, 2H), 6.88-6.83 (d, J=8.8 Hz, 1H), 6.80-6.73 (d, J=8.4 Hz, 2H), 5.00 (s, 1H), 4.31 (t, J=6.2 Hz, 2H), 3.85 (t, J=6.2 Hz, 2H), 1.63 (s, 6H).
  • 5-(2-(3-Bromo-4-hydroxyphenyl)propan-2-yl)-2-(2-chloroethoxy)benzonitrile (6)
  • To a solution of 2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (5) (1.35 g, 4.27 mmol) in EtOH (30 mL) was added Br2 (0.22 mL, 4.27 mmol) dropwise under N2 atmosphere at 0° C. Then the reaction was stirred at 20° C. for 3 hours, and then at 40° C. for another 6 hours. LCMS showed 23% of starting material and 73% of desired product in reaction mixture. The mixture was poured into saturated aqueous Na2SO3 (20 mL) then concentrated under reduced to remove EtOH. The resulting solution was extracted with EtOAc (20 mL×2). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced. The residue was purified by MPLC to give 5-(2-(3-bromo-4-hydroxyphenyl)propan-2-yl)-2-(2-chloroethoxy)benzonitrile (6) (1.16 g, yield: 68.7%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ=7.45 (d, J=2.4 Hz, 1H), 7.37-7.31 (d, J=8.8 Hz, 1H), 7.30-7.28 (m, 1H), 7.03-6.98 (m, 1H), 6.96-6.92 (m, 1H), 6.87 (d, J=8.8 Hz, 1H), 5.44 (br s, 1H), 4.32 (t, J=6.2 Hz, 2H), 3.86 (t, J=6.2 Hz, 2H), 1.63 (s, 6H). LCMS (M+H+) m/z: calcd 393.0. found: 393.8.
  • 2-(2-Chloroethoxy)-5-(2-(3-cyano-4-hydroxyphenyl)propan-2-yl)benzonitrile (7)
  • To a solution of 5-(2-(3-bromo-4-hydroxyphenyl)propan-2-yl)-2-(2-chloroethoxy) benzonitrile (6) (0.60 g, 1.52 mmol) in NMP (5 mL) was added CuCN (0.68 g, 7.6 mol) under N2 atmosphere at 20° C. The reaction was stirred at 180° C. for 0.5 hour. TLC showed the reaction was completed. The mixture was poured into H2O (20 mL), extracted with EtOAc (10 mL×2). The combined organic layers were washed with brine (10 mL×6), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by MPLC to give 2-(2-chloroethoxy)-5-(2-(3-cyano-4-hydroxyphenyl)propan-2-yl)benzonitrile (7) (130 mg, yield: 25.1%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ=7.44-7.41 (m, 1H), 7.35 (d, J=2.4 Hz, 1H), 7.32 (dd, J=2.5, 8.9 Hz, 1H), 7.23 (dd, J=2.4, 8.8 Hz, 1H), 6.91 (d, J=7.3 Hz, 1H), 6.90-6.88 (dd, J=2.5, 8.9 Hz, 1H), 6.10 (br, 1H), 4.37-4.28 (t, J=6.2 Hz, 2H), 3.87 (t, J=6.2 Hz, 2H), 1.64 (s, 6H).
  • 2-(2-Chloroethoxy)-5-(2-(3-cyano-4-((4-(methylsulfonyl)oxazol-5-yl)methoxy) phenyl)propan-2-yl) benzonitrile (A63)
  • To a solution of 2-(2-chloroethoxy)-5-(2-(3-cyano-4-hydroxyphenyl)propan-2-yl)benzonitrile (7) (130 mg, 0.38 mmol) in DMF (2 mL) was added 5-(chloromethyl)-4-(methylsulfonyl)oxazole (G) (75 mg, 0.38 mol) and Cs2CO3 (249 mg, 0.76 mmol) under N2 atmosphere. The reaction was stirred at 0° C. for 3 hours. LCMS showed the reaction was completed. The mixture was diluted with EtOAc (5 mL) and poured into H2O (5 mL). The aqueous phase was extracted with EtOAc (5 mL×2). The combined organic layers were washed with brine (5 mL×4), then dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by prep-HPLC(TFA) to give 2-(2-chloroethoxy)-5-(2-(3-cyano-4-((4-(methylsulfonyl)oxazol-5-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A63) (53 mg, yield: 27.8%) as white solid. LCMS purity (220 nm): 91.1%. 1H NMR (400 MHz, CHCl3-d) δ=8.04 (s, 1H), 7.41 (br s, 2H), 7.39-7.29 (m, 2H), 7.15-7.04 (m, 1H), 6.94-6.85 (d, J=8.9 Hz, 1H), 5.51 (s, 2H), 4.33 (br t, J=6.0 Hz, 2H), 3.87 (br t, J=6.0 Hz, 2H), 3.25 (s, 3H), 1.64 (s, 6H). LCMS (M+H+) m/z: calcd 499.1. found 500.1.
  • Example 15: Synthesis of N-((2-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl) phenyl)amino)oxazol-5-yl)methyl)methanesulfonamide (A75)
  • Figure US20200123117A1-20200423-C00339
  • Ethyl 2-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)amino)oxazole-5-carboxylate (3)
  • To a solution of 4-[1-[3,5-dichloro-4-(2-chloroethoxy)-phenyl]-1-methyl-ethyl]aniline (1) (1.00 g, 2.5 mmol) in DMF (10 mL) was added Ag2CO3 (1.05 g, 3.81 mol) and ethyl 2-chlorooxazole-5-carboxylate (2) (0.668 g, 3.81 mmol) under N2 atmosphere at 20° C. Then the mixture was stirred at 70° C. for 16 hours. TLC showed the reaction was completed. The mixture was cooled down, poured into H2O (50 mL) and extracted with EtOAc (25 mL×2). The organic layers were combined and washed with brine (20 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give ethyl 2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]anilino]oxazole-5-carboxylate (3) (0.7 g, yield: 56.2%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ 7.62 (s, 1H), 7.46 (s, 1H), 7.43 (d, J=8.8 Hz, 2H), 7.20 (d, J=8.8 Hz, 2H), 7.14 (s, 2H), 4.37 (q, J=7.2 Hz, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.64 (s, 6H), 1.38 (t, J=7.2 Hz, 3H).
  • (2-((4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)amino)oxazol-5-yl)methanol (4)
  • To a solution of ethyl 2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy) phenyl]-1-methyl-ethyl]anilino]oxazole-5-carboxylate (3) (0.6 g, 1.2 mmol) in DCM (10 mL) was added DIBAL-H (1 M in toluene, 2.4 mL, 2.4 mmol) under N2 atmosphere at 0° C. Then the resulting mixture was stirred at 20° C. for 3 hours. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl (20 mL) and extracted with DCM (10 mL×3). The organic layers were washed with brine (15 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give [2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]anilino]oxazol-5-yl]-methanol (4) (410 mg, yield: 74.6%) as brown oil. 1H NMR (400 MHz, CHCl3-d) δ 7.38 (d, J=8.8 Hz, 2H), 7.17 (d, J=8.8 Hz, 2H), 7.14 (s, 2H), 6.81 (s, 1H), 4.60 (s, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.63 (s, 6H).
  • tert-Butyl ((2-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)-amino)oxazol-5-yl)methyl)(methylsulfonyl)carbamate (5)
  • To a mixture of [2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]anilino]oxazol-5-yl]methanol (4) (50 mg, 0.1 mmol) and tert-butyl N-methylsulfonylcarbamate (21 mg, 0.1 mmol) in THF (3 mL) was added PPh3 (58 mg, 0.22 mmol) and DEAD (29 mg, 0.16 mmol). The mixture was stirred at 25° C. for 10 hours under N2 atmosphere. LCMS showed 30% desired product was detected. The mixture was concentrated and purified by prep-TLC to give tert-butyl N-[[2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]anilino]oxazol-5-yl]methyl]-N-methylsulfonyl-carbamate (5) (25 mg, yield: 36.0%) as colorless oil. 1H NMR (400 MHz, CHCl3-d) δ 7.39 (d, J=8.8 Hz, 2H), 7.16 (d, J=8.8 Hz, 2H), 7.13 (s, 2H), 6.84 (s, 1H), 4.86 (s, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.24 (s, 3H), 1.63 (s, 9H), 1.53 (s, 6H).
  • N-((2-((4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)amino)oxazol-5-yl)methyl)methanesulfonamide (A75)
  • A solution of tert-butyl N-[[2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]anilino]oxazol-5-yl]methyl]-N-methylsulfonyl-carbamate (5) (25 mg, 0.04 mmol) in DCM (2 mL) and TFA (0.2 mL) was stirred at 25° C. for 3 hours. LCMS showed the reaction was completed. The mixture was concentrated and purified by prep-HPLC (TFA) to give N-[[2-[4-[1-[3,5-dichloro-4-(2-chloroethoxy)phenyl]-1-methyl-ethyl]anilino]oxazol-5-yl]methyl]methanesulfonamide (4.6 mg, yield: 21.9%) as yellow oil. LCMS purity (220 nm): 86%. 1H NMR (400 MHz, CHCl3-d) δ 7.36-7.33 (m, 2H), 7.25-7.22 (m, 2H), 7.11 (s, 2H), 7.02 (s, 1H), 5.18 (s, 1H), 4.36 (s, 2H), 4.27 (t, J=6.4 Hz, 2H), 3.87 (t, J=6.4 Hz, 2H), 3.00 (s, 3H), 1.64 (s, 6H). LCMS (M+H+) m/z: calcd: 531.0. found 531.6.
  • Example 16: Synthesis of 3-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-1,5,5-trimethylimidazolidine-2,4-dione (B2)
  • Figure US20200123117A1-20200423-C00340
  • Methyl 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzoate (2)
  • To a solution of 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl trifluoromethanesulfonate (1) (600 mg, 1.7 mmol) and TEA (1 mL, 5.0 mmol) in MeOH (15 mL) was added Pd(dppf)Cl2 (200 mg, 0.3 mmol). The resulting mixture was stirred at 50 Psi of CO atmosphere and the mixture at 60° C. for 16 hours. TLC showed the reaction was completed. The mixture was poured into H2O (30 mL), extracted with EtOAc (10 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-TLC to give methyl 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzoate (2) (300 mg, yield: 44.5%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 8.00-7.90 (m, 2H), 7.27-7.23 (m, 2H), 7.09 (s, 2H), 4.28-4.21 (m, 2H), 3.90 (s, 3H), 3.87-3.81 (m, 2H), 1.68-1.62 (m, 6H).
  • (4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)methanol (3)
  • To a solution of methyl 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzoate (2) (300 mg, 0.7 mmol) in THF (3 mL) was added DIBAL-H (1.00 M, 2 mL) at 0° C. and the mixture was stirred at 0-25° C. for 2 hours. TLC showed the reaction was completed. The mixture was poured into H2O (15 mL), extracted with EtOAc (5 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)methanol (3) (170 mg, yield: 60.9%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=7.32 (d, J=8.3 Hz, 2H), 7.24-7.18 (m, 2H), 7.17-7.11 (m, 2H), 4.75-4.63 (m, 2H), 4.31-4.22 (m, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.70-1.61 (m, 6H).
  • 1,3-Dichloro-2-(2-chloroethoxy)-5-(2-(4-(chloromethyl)phenyl)propan-2-yl)benzene (4)
  • To a solution of (4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)methanol (3) (40 mg, 0.1 mmol) in DCM (2 mL) was added SOCl2 (0.1 mL, 1.1 mmol) at 0° C. and the mixture was stirred at 0-25° C. for 2 hours. TLC showed the reaction was completed. The mixture was poured into H2O (10 mL), extracted with DCM (3 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 1,3-dichloro-2-(2-chloroethoxy)-5-(2-(4-(chloromethyl)phenyl)propan-2-yl)benzene (4) (40 mg, yield: 95.3%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=7.36-7.30 (m, 2H), 7.20 (d, J=8.4 Hz, 2H), 7.15-7.15 (m, 1H), 7.14 (s, 1H), 4.59 (s, 2H), 4.27 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.68-1.62 (m, 6H).
  • 3-(4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-1,5,5-trimethylimida zolidine-2,4-dione (B2)
  • To a mixture of 1,5,5-trimethylimidazolidine-2,4-dione (5) (20 mg, 0.2 mmol) and K2CO3 (70 mg, 0.5 mmol) in DMF (3 mL) was added 1,3-dichloro-2-(2-chloroethoxy)-5-(2-(4-(chloromethyl)phenyl)propan-2-yl)benzene (4) (50 mg, 0.1 mmol) at 25° C. and the mixture was stirred at the same temperature for 2 hours. LCMS showed the reaction was completed. The mixture was poured into H2O (10 mL), extracted with EtOAc (5 mL×2). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (HCl) to give 3-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-1,5,5-trimethylimidazolidine-2,4-dione (B2) (20 mg, yield: 31.8%) as colorless oil. LCMS purity (220 nm): 96.1%. 1H NMR (400 MHz, CHCl3-d) δ=7.30-7.25 (m, 1H), 7.28-7.25 (m, 1H), 7.30-7.25 (m, 1H), 7.15-7.10 (m, 2H), 7.10-7.08 (m, 2H), 4.66-4.57 (m, 2H), 4.24 (t, J=6.4 Hz, 2H), 3.89-3.77 (m, 2H), 2.87 (s, 3H), 1.65-1.54 (m, 6H), 1.41-1.34 (m, 6H). LCMS (M+H+) m/z: calcd: 496.1. found 497.1.
  • Example 17: Synthesis of 3-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-5,5-dimethyl-1-(methylsulfonyl)imidazolidine-2,4-dione (B3)
  • Figure US20200123117A1-20200423-C00341
  • Methyl 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzoate (2)
  • To a solution of 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl trifluoromethanesulfonate (1) (600 mg, 1.7 mmol) and TEA (1 mL, 5.0 mmol) in MeOH (15 mL) was added Pd(dppf)Cl2 (200 mg, 0.3 mmol). The resulting mixture was stirred at 50 Psi of CO atmosphere and the mixture at 60° C. for 16 hours. TLC showed the reaction was completed. The mixture was poured into H2O (30 mL), extracted with EtOAc (10 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-TLC to give methyl 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzoate (2) (300 mg, yield: 44.5%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ 8.00-7.90 (m, 2H), 7.27-7.23 (m, 2H), 7.09 (s, 2H), 4.28-4.21 (m, 2H), 3.90 (s, 3H), 3.87-3.81 (m, 2H), 1.68-1.62 (m, 6H).
  • (4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)methanol (3)
  • To a solution of methyl 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzoate (2) (300 mg, 0.7 mmol) in THF (3 mL) was added DIBAL-H (1.00 M, 2 mL) at 0° C. and the mixture was stirred at 0-25° C. for 2 hours. TLC showed the reaction was completed. The mixture was poured into H2O (15 mL), extracted with EtOAc (5 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)methanol (3) (170 mg, yield: 60.9%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=7.32 (d, J=8.3 Hz, 2H), 7.24-7.18 (m, 2H), 7.17-7.11 (m, 2H), 4.75-4.63 (m, 2H), 4.31-4.22 (m, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.70-1.61 (m, 6H).
  • 1,3-Dichloro-2-(2-chloroethoxy)-5-(2-(4-(chloromethyl)phenyl)propan-2-yl)benzene (4)
  • To a solution of (4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenyl)methanol (3) (40 mg, 0.1 mmol) in DCM (2 mL) was added SOCl2 (0.1 mL, 1.1 mmol) at 0° C. and the mixture was stirred at 0-25° C. for 2 hours. TLC showed the reaction was completed. The mixture was poured into H2O (10 mL), extracted with DCM (3 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 1,3-dichloro-2-(2-chloroethoxy)-5-(2-(4-(chloromethyl)phenyl)propan-2-yl)benzene (4) (40 mg, yield: 95.3%) as yellow oil. 1H NMR (400 MHz, CHCl3-d) δ=7.36-7.30 (m, 2H), 7.20 (d, J=8.4 Hz, 2H), 7.15-7.15 (m, 1H), 7.14 (s, 1H), 4.59 (s, 2H), 4.27 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.68-1.62 (m, 6H).
  • 3-(4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-5,5-dimethylimidazolidine-2,4-dione (6)
  • To a mixture of 1,3-dichloro-2-(2-chloroethoxy)-5-(2-(4-(chloromethyl)phenyl)propan-2-yl)benzene (4) (100 mg, 0.2 mmol) and 5,5-dimethylimidazolidine-2,4-dione (5) (30 mg, 0.2 mmol) in DMF (5 mL) was added K2CO3 (100 mg, 0.8 mmol) at 25° C. and the mixture was stirred at 25° C. for 16 hours. LCMS showed the reaction was completed. The mixture was poured into H2O (15 mL), extracted with EtOAc (5 mL×2). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 3-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-5,5-dimethylimidazolidine-2,4-dione (6) (40 mg, yield: 64.8%) as yellow oil. LCMS purity (220 nm): 97.7%. 1H NMR (400 MHz, CHCl3-d) δ=7.16-7.08 (m, 4H), 6.83 (d, J=8.8 Hz, 2H), 5.36 (s, 1H), 5.27 (s, 1H), 4.51 (s, 2H), 4.45 (s, 2H), 4.17-4.12 (m, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.31 (s, 3H), 2.28 (quin, J=6.1 Hz, 2H), 1.62 (s, 6H), 1.49 (s, 9H). LCMS (M+H+) m/z: calcd: 482.1. found 483.2.
  • 3-(4-(2-(3,5-Dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-5,5-dimethyl-1-(methylsulfonyl)imidazolidine-2,4-dione (B3)
  • To a solution of 3-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-5,5-dimethylimidazolidine-2,4-dione (6) ((40 mg, 0.1 mmol) in THF (2 mL) was added Mesyl chloride (0.1 mL, 0.2 mmol) and NaH (60.0%, 6 mg, 0.2 mmol) at 0° C. and the mixture was stirred at 80° C. for 16 hours. TLC showed the reaction was completed. The reaction was quenched with saturated aqueous NH4Cl (10 mL) and extracted with EtOAc (3 mL×2). The combined organic layers were washed with brine (3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (HCl) to give 3-(4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)benzyl)-5,5-dimethyl-1-(methylsulfonyl)imidazolidine-2,4-dione (5 mg, yield: 10.8%) as yellow oil. LCMS purity (220 nm): 81.8%. 1H NMR (400 MHz, CHCl3-d) δ=7.31-7.28 (m, 2H), 7.20-7.14 (m, 2H), 7.14-7.11 (m, 2H), 4.71-4.65 (m, 2H), 4.27 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.38 (s, 3H), 1.76-1.71 (m, 6H), 1.64 (s, 6H). LCMS (M+H+) m/z: calcd: 526, found: 527.
  • Example 18: Synthesis of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((4-(methylsulfonyl)oxazol-5-yl) methoxy)phenyl)propan-2-yl)benzonitrile (A28)
  • Figure US20200123117A1-20200423-C00342
  • To a mixture of 3-chloro-2-(2-chloro ethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (900 mg, 2.6 mmol) in DMF (10 mL) was added Cs2CO3 (167 mg, 5.1 mmol) and 5-(chloromethyl)-4-(methylsulfonyl)oxazole (750 mg, 3.9 mmol) under N2 atmosphere at 20° C. The mixture was stirred at 40° C. for 1 h. TLC showed the reaction was completed. The mixture was cooled down to room temperature, poured into H2O (40 mL) and extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine (20 mL×4), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by MPLC and then triturated with MTBE: EtOAc=10 mL: 2 mL to give 3-chloro-2-(2-chloroethoxy) 5-(2-(4-((4-(methylsulfonyl) xazol-5-yl) ethoxy)phenyl) propan-2-yl)benzonitrile (310 mg, yield: 23.2%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.00 (s, 1H), 7.43 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.13 (d, J=8.4 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 5.42 (s, 2H), 4.43 (t, J=6.4 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 3.21 (s, 3H), 1.64 (s, 6H). LCMS (220 nm): 97.8%. Exact Mass: 508.1; found 509.1, 511.1.
  • Example 19: Synthesis of 2-(2-chloroethoxy)-3-methyl-5-(2-(4-((4-(methylsulfonyl)oxazol-5-yl)methoxy) phenyl)proan-2-yl)benzonitrile (A29)
  • Figure US20200123117A1-20200423-C00343
  • Methyl 4-hydroxy-3-iodo-5-methylbenzoate (2)
  • To a solution of methyl 4-hydroxy-3-methyl-benzoate (3.0 g, 18.0 mmol) in DCM (30 mL) was added NIS (4.0 g, 18.0 mmol) at 0° C. under N2 atmosphere. The mixture was stirred at 20° C. for 16 hrs. TLC showed the reaction was completed. The mixture was poured into water (20 mL), extracted with DCM (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give methyl 4-hydroxy-3-iodo-5-methyl-benzoate (3.6 g, yield: 68.3%) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.21 (d, J=1.2 Hz, 1H), 7.80 (s, 1H), 5.70 (s, 1H), 3.89 (s, 3H), 2.34 (s, 3H).
  • Methyl 4-(2-chloroethoxy)-3-iodo-5-methylbenzoate (4)
  • To a mixture of methyl 4-hydroxy-3-iodo-5-methyl-benzoate (3.6 g, 12.3 mmol) and 1-bromo-2-chloro-ethane (1.9 g, 13.6 mmol) in DMF (80 mL) was added Cs2CO3 (8.0 g, 24.7 mmol) and the mixture was stirred at 70° C. for 16 hrs. TLC showed the reaction was completed. The mixture was cooled down and poured into water (80 mL), extracted with EtOAc (50 mL×3). The combined organic layers were washed with brine (50 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give methyl 4-(2-chloroethoxy)-3-iodo-5-methyl-benzoate (2.3 g, yield: 52.6%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ=8.31 (d, J=1.6 Hz, 1H), 7.87-7.86 (m, 1H), 4.17 (t, J=6.0 Hz, 2H), 3.89-3.95 (m, 5H), 2.41 (s, 3H).
  • Methyl 4-(2-chloroethoxy)-3-cyano-5-methylbenzoate (5)
  • To a solution of methyl 4-(2-chloroethoxy)-3-iodo-5-methyl-benzoate (2.1 g, 6.0 mmol) in NMP (20 mL) was added CuCN (637 mg, 7.1 mmol) under N2 atmosphere and the mixture was stirred at 160° C. for 2 hrs. LCMS showed the reaction was completed. The mixture was cooled down and poured into water (60 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (30 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give methyl 4-(2-chloroethoxy)-3-cyano-5-methyl-benzoate (0.7 g, yield: 46.6%) as yellow solid. 1H NMR (400 MHz, CHCl3-d): δ 8.13 (d, J=2.0 Hz, 1H), 8.09 (d, J=1.2 Hz, 1H), 4.54 (t, J=5.6 Hz, 2H), 3.93 (s, 3H), 3.89 (t, J=5.6 Hz, 2H), 2.39 (s, 3H). LCMS (220 nm): 78.1%. Exact Mass: 253.1; found: 254.1.
  • 2-(2-chloroethoxy)-5-(2-hydroxypropan-2-yl)-3-methylbenzonitrile (6)
  • To a mixture of methyl 4-(2-chloroethoxy)-3-cyano-5-methyl-benzoate (500 mg, 2.1 mmol) and TEA (1.7 mL, 11.8 mmol) in THF (10 mL) was added MeMgBr (3M, 9.8 mmol, 3.3 mL) dropwise at 0° C. under N2 atmosphere and the mixture was stirred at 0° C. for 2 hrs. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl (20 mL), extracted with EtOAc (10 mL×4), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give 2-(2-chloroethoxy)-5-(2-hydroxypropan-2-yl)-3-methylbenzonitrile (250 mg, yield: 50.0%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.55 (s, 2H), 4.38 (t, J=5.6 Hz, 2H), 3.87 (t, J=5.6 Hz, 2H), 2.36 (s, 3H), 1.57 (s, 6H).
  • 2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)-3-methylbenzonitrile (7)
  • To a solution of 2-(2-chloroethoxy)-5-(2-hydroxy-2-methyl-ethyl)-3-methyl-benzonitrile (250 mg, 1.0 mmol) and phenol (110 mg, 1.2 mmol) in DCM (3 mL) was added BF3-Et2O (0.5 mL, 2 mmol) at −78° C. under N2 atmosphere and the mixture was stirred at the same temperature for 2 hrs. TLC showed the reaction was completed. The mixture was poured into water (5 mL) and extracted with DCM (5 mL×3). The combined organic layers were washed with brine (15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give 2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl) propan-2-yl)-3-methylbenzonitrile (220 mg, yield: 67.7%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ=7.29 (d, J=2.4 Hz, 1H), 7.21 (d, J=2.0 Hz, 1H), 7.06 (d, J=6.4 Hz, 2H), 6.76 (d, J=6.8 Hz, 2H), 4.73 (s, 1H), 4.36 (t, J=6.0 Hz, 2H), 3.86 (t, J=6.0 Hz, 2H), 2.29 (s, 3H), 1.63 (s, 6H)
  • 2-(2-chloroethoxy)-3-methyl-5-(2-(4-((4-(methylsulfonyl)oxazol-5-yl)methoxy)phenyl)proan-2-yl)benzonitrile (A29)
  • To a mixture of 2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)-3-methylbenzonitrile (100 mg, 0.3 mmol) and 5-(chloromethyl)-4-methylsulfonyl-oxazole (60 mg, 0.3 mmol) in DMF (3 mL) was added Cs2CO3 (200 mg, 0.6 mmol) at 20° C. and the mixture was stirred at 40° C. for 4 hrs. LCMS showed the reaction was completed. The mixture was cooled down, filtered and purified by prep-HPLC (TFA) to give 2-(2-chloroethoxy)-3-methyl-5-(2-(4-((4-(methylsulfonyl)oxazol-5-yl)methoxy)phenyl)propan-2-yl)benzonitrile (26 mg, yield: 17.5%) as yellow oil. 1H NMR (400 MHz, MeOD): δ 8.39 (s, 1H), 7.33-7.35 (m, 1H), 7.31-7.32 (m, 1H), 7.16 (d, J=8.8 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 5.41 (s, 2H), 4.32 (t, J=5.6 Hz, 2H), 3.88 (t, J=5.4 Hz, 2H), 3.18 (s, 3H), 2.28 (s, 3H), 1.64 (s, 6H). LCMS: (220 nm): 98.8%. Exact Mass: 488.1; found 489.0.
  • Example 20: Synthesis of N-(4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy) methyl)pyrimidin-2-yl)methanesulfonamide (A74)
  • Figure US20200123117A1-20200423-C00344
  • 4-(bromomethyl)-2-chloropyrimidine (2)
  • To a mixture of 2-chloro-4-methyl-pyrimidine (5.0 g, 38.9 mmol) and NBS (10.4 g, 58.3 mmol) in CCl4 (100 mL) was added AIBN (2.18 g, 19.4 mmol) at 20° C. and the mixture was stirred at 100° C. for 16 hrs. LCMS showed most of the starting material consumed. The mixture was cooled down, added to water (200 mL) and extracted with DCM (100 mL×5). The combined organic phases were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by MPLC to give 4-(bromomethyl)-2-chloropyridine (2.0 g, yield: 24.8%) as brown oil. 1H NMR (400 MHz, CDCl3) δ=8.65 (d, J=5.2 Hz, 1H), 7.45 (d, J=5.2 Hz, 1H), 4.43 (s, 2H).
  • 2-chloro-4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl) pyrimidine (4)
  • To a mixture of 4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenol (0.2 g, 0.6 mmol) and 4-(bromomethyl)-2-chloropyrimidine (127 mg, 0.6 mmol) in MeCN (5 mL) was added K2CO3 (115 mg, 0.83 mmol) and the mixture was stirred at 25° C. for 16 hrs. LCMS showed the reaction was completed. The resulting mixture was quenched with saturated aqueous NH4Cl (5 mL), extracted with EtOAc (5 mL×3). The combined organic layers we washed with brine (10 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give 2-chloro-4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)pyrimidine (270 mg, yield: 82.9%) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.66 (d, J=5.2 Hz, 1H), 7.56 (d, J=5.2 Hz, 1H), 7.16 (d, J=8.8 Hz, 2H), 7.13 (s, 2H), 6.88 (d, J=8.8 Hz, 2H), 5.15 (s, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 1.63 (s, 6H).
  • N-(4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy) methyl)pyrimidin-2-yl)methanesulfonamide (A74)
  • To a mixture of 2-chloro-4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)pyrimidine (200 mg, 0.3 mmol), methanesulfonamide (62.6 mg, 0.66 mmol), Cs2CO3 (322 mg, 0.99 mmol) in anhydrous dioxane (8 mL) was added Pd2(dba)3 (30.1 mg, 0.03 mmol) and Xantphos (38.1 mg, 0.06 mmol) and the mixture was stirred at 100° C. for 16 hrs under N2 atmosphere. LCMS showed the reaction was completed. The mixture was cooled down, poured into water (10 mL), extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by p-HPLC(TFA) to obtain the desired product N-(4-((4-(2-(3,5-dichloro-4-(2-chloroethoxy)phenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (61.4 mg, yield: 34.2%) as light red solid. 1H NMR (400 MHz, CDCl3) δ=8.61 (d, J=5.2 Hz, 1H), 7.29 (d, J=5.2 Hz, 1H), 7.16-7.11 (m, 4H), 6.88 (d, J=8.8 Hz, 2H), 5.09 (s, 2H), 4.26 (t, J=6.4 Hz, 2H), 3.86 (t, J=6.4 Hz, 2H), 3.47 (s, 3H), 1.63 (s, 6H). LCMS: (220 nm): 98.1%. Exact Mass: 543.1; found 544.
  • Example 21: Synthesis of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylsulfonyl)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A93)
  • Figure US20200123117A1-20200423-C00345
  • 2-(((tert-butyldiphenylsilyl)oxy)methyl)thiazole (2)
  • To a mixture of thiazol-2-ylmethanol (1.0 g, 8.7 mmol) and imidazole (1.2 g, 17.4 mmol) in DCM (50 mL) was added TBDPSCl (2.9 g, 10.4 mmol) in portions at 25° C. The mixture was stirred at the same temperature for 2 hrs. TLC showed the reaction was completed. The reaction was quenched with water (80 mL) and extracted with DCM (50 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduce pressure. The residue was purified by MPLC to give 2-(((tert-butyldiphenylsilyl)oxy)methyl)thiazole (3.0 g, yield: 97.7%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ=7.73-7.70 (m, 4H), 7.48-7.45 (m, 1H), 7.44-7.38 (m, 6H), 7.31 (d, J=3.2 Hz, 1H), 5.02 (s, 2H), 1.14 (s, 9H).
  • 2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-(methylthio)thiazole (3)
  • To a solution of 2-(((tert-butyldiphenylsilyl)oxy)methyl)thiazole (4.2 g, 11.9 mmol) in THF (80 mL) was added n-BuLi (2.50 M, 9.50 mL) dropwise at −78° C. under N2 atmosphere. The mixture was stirred at the same temperature for 30 min. Then 1,2-dimethyldisulfane (2.2 g, 23.8 mmol) was added to the mixture dropwise at −78° C. under N2 atmosphere and stirred for 30 min. TLC showed the reaction was completed. The reaction mixture was poured into saturated aqueous NH4Cl (100 mL) and extracted with EtOAc (50 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduce pressure to give 2-(((tert-butyldiphenylsilyl)oxy)methyl)-5-(methylthio)thiazole (4.6 g, yield: 96.9%) as brown oil. 1H NMR (400 MHz, CDCl3) δ=7.74-7.67 (m, 4H), 7.60 (s, 1H), 7.48-7.38 (m, 6H), 4.92 (s, 2H), 2.50 (s, 3H), 1.13 (s, 9H).
  • (5-(methylthio)thiazol-2-yl)methanol (4)
  • To a solution of 2-(((tert-butyldiphenylsilyl) oxy) methyl)-5-(methylthio)thiazole (5.4 g, 13.5 mmol) in anhydrous THF (27 mL) was added TBAF (1.00 M, 27.0 mL) dropwise at 20° C. and stirred at the same temperature for 1 hour. TLC showed the reaction was completed. The reaction was quenched with water (50 mL). The aqueous layer was extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (30 mL×3), dried over Na2SO4, filtered and concentrated under reduce pressure. The residue was purified by MPLC to give (5-(methylthio)thiazol-2-yl)methanol (1.4 g, yield: 64.3%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.63 (s, 1H), 4.91 (d, J=5.6 Hz, 2H), 2.72 (t, J=6.0 Hz, 1H), 2.49 (s, 3H).
  • 2-(chloromethyl)-5-(methylthio)thiazole (5)
  • To a solution of (5-(methylthio)thiazol-2-yl)methanol (0.4 g, 2.5 mmol) in DCM (5 mL) was added SOCl2 (0.5 mL, 7.4 mmol) at 20° C., then DMF (0.1 mL) was added. The mixture was stirred at the same temperature for 1 h. TLC showed the reaction was completed. The reaction was concentrated under reduce pressure. The residue was quenched with water (10 mL) and extracted with DCM (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduce pressure to give 2-(chloromethyl)-5-(methylthio)thiazole (0.4 g, yield: 89.7%) as brown oil. 1H NMR (400 MHz, CDCl3) δ=7.63 (s, 1H), 4.82 (s, 2H), 2.52 (s, 3H).
  • 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylthio)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (6)
  • To a mixture of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl) propan-2-yl)benzonitrile (100 mg, 0.3 mmol) and 2-(chloromethyl)-5-(methylthio)thiazole (103 mg, 0.6 mmol) in DMF (3 mL) was added Cs2CO3 (186 mg, 0.6 mmol) and the mixture was stirred at 20° C. for 2 hrs. LCMS showed the reaction was completed. The resulting mixture was quenched with water (10 mL) and extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL×2), dried over Na2SO4, filtered and concentrated under reduce pressure. The residue was purified by MPLC to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylthio)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (80 mg, yield: 56.8%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.66 (s, 1H), 7.44 (s, 1H), 7.33 (s, 1H), 7.12 (br d, J=8.4 Hz, 2H), 6.95 (br d, J=8.8 Hz, 2H), 5.30 (s, 2H), 4.42 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 2.50 (s, 3H), 1.65 (s, 6H).
  • 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylthio)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A93)
  • A suspension of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylthio)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (80 mg, 0.16 mmol) and m-CPBA (80.0%, 105 mg, 0.49 mmol) in DCM (3 mL) was stirred at 20° C. for 1 h. LCMS showed the reaction was completed. The resulting mixture was poured into saturated aqueous NaHCO3 (3 mL) and extracted with DCM (3 mL×3). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduce pressure. The residue was purified by prep-HPLC (TFA) to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylsulfonyl)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (64.2 mg, yield: 75.4%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.31 (s, 1H), 7.45 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.16 (d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 5.40 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.25 (s, 3H), 1.66 (s, 6H). LCMS (220 nm): 99.5%. Exact Mass: 524.0; found 525.1, 527.1.
  • Example 22: Synthesis of N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy) methyl)pyrimidin-2-yl)methanesulfonamideN-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl) propan-2-yl) phenoxy) methyl)pyrimidin-2-yl)methanesulfonamide (A109)
  • Figure US20200123117A1-20200423-C00346
  • 2-chloro-4-(chloromethyl)pyrimidine (2)
  • To a mixture of 2-chloro-4-methyl-pyrimidine (50.0 g, 398 mmol) and NCS (77.9 g, 583 mmol) in MeCN (250 mL) was added benzoyl benzenecarboperoxoate (28.3 g, 117 mmol) in portions at 20° C. and the mixture was stirred at 100° C. for 16 hrs under N2 atmosphere. TLC showed most of the starting material consumed and two new spots appeared. The mixture was cooled down to room temperature, poured into water (500 mL) and extracted with EtOAc (200 mL×3). The organic layers were combined and washed with brine (200 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 2-chloro-4-(chloromethyl) pyrimidine (22 g, yield: 31.2%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=8.69 (d, J=5.2 Hz, 1H), 7.54 (d, J=5.0 Hz, 1H), 4.61 (s, 2H).
  • 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((2-chloropyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (4)
  • To a mixture of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (18.0 g, 51.4 mmol) and 2-chloro-4-(chloromethyl) pyrimidine (10.1 g, 61.7 mmol) in DMF (150 mL) was added Cs2CO3 (33.5 g, 103.4 mmol) at 20° C. and the mixture was stirred at the same temperature for 16 hrs. LCMS showed the reaction was completed. The reaction mixture was poured into H2O (300 mL) and extracted with EtOAc (150 mL×3). The combined organic layers were washed with brine (150 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-(2-chloroethoxy)-5-(2-(4-((2-chloropyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (15.5 g, yield: 63.3%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.67 (d, J=5.2 Hz, 1H), 7.56 (d, J=5.2 Hz, 1H), 7.45 (d, J=2.4 Hz, 1H), 7.35-7.29 (m, 1H), 7.13 (d, J=8.8 Hz, 2H), 6.90 (d, J=8.8 Hz, 2H), 5.16 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 1.65 (s, 6H).
  • N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl) pyrimidin-2-yl)methanesulfonamide (A109)
  • To a mixture of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((2-chloropyrimidin-4-yl)methoxy)phenyl)propan-2-yl)benzonitrile (15.5 g, 32.5 mmol), methane sulfonamide (9.3 g, 97.5 mmol), Cs2CO3 (21.2 g, 65.0 mmol) and Xantphos (1.88 g, 3.25 mmol) in 1,4-dioxane (450 mL) was added Pd2(dba)3 (3.0 g, 3.3 mmol) at 20° C. and the mixture was stirred at 90° C. for 6 hrs under N2 atmosphere. LCMS showed the reaction was completed. The mixture was cooled down to room temperature, poured into water (300 mL) and extracted with EtOAc (300 mL×3). The combined organic layers were washed with brine (300 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the crude product and then further purified by p-HPLC (TFA) to give N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy) methyl)pyrimidin-2-yl)methanesulfonamide (5.30 g, yield: 30.1%) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=10.02 (br s, 1H), 8.69 (d, J=5.2 Hz, 1H), 7.45 (d, J=2.4 Hz, 1H), 7.34-7.31 (m, 1H), 7.30 (d, J=5.2 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 6.91 (d, J=8.8 Hz, 2H), 5.13 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.47 (s, 3H), 1.65 (s, 6H). LCMS (220 nm): 99.0%. Exact Mass: 534.09; found 535.1, 537.0.
  • Example 23: Synthesis of N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)ethyl)oxazol-2-yl)methanesulfonamide (A122)
  • Figure US20200123117A1-20200423-C00347
    Figure US20200123117A1-20200423-C00348
  • 2-Chloro-4-(1-ethoxyvinyl)-5-methylpyrimidine (2)
  • To a mixture of 2,4-dichloro-5-methyl-pyrimidine (10.0 g, 61.3 mmol) and tributyl(1-ethoxyvinyl)stannane (22.2 g, 0.0613 mol) in DMF (200 mL) was added Pd(PPh3)2Cl2 (2.15 g, 3.1 mmol) at 20° C. and the mixture was stirred at 100° C. for 5 hrs under N2 atmosphere. TLC showed the reaction was completed. The reaction was quenched by KF solution (30 g, 0.172 mol dissolved 500 mL in water), filtered and extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (100 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 2-chloro-4-(1-ethoxyvinyl)-5-methyl-pyrimidine (6.0 g, yield: 41.5%) as yellow oil. 1H NMR (400 MHz, CDCl3): δ 8.40 (s, 1H), 5.01 (d, J=2.4 Hz, 1H), 4.55 (d, J=2.4 Hz, 1H), 3.94 (q, J=7.2 Hz, 2H), 2.38 (s, 3H), 1.41 (t, J=7.2 Hz, 3H).
  • Ethyl 2-chloro-5-methylpyrimidine-4-carboxylate (3)
  • O3 (gas) was bubbled into a solution of 2-chloro-4-(1-ethoxyvinyl)-5-methyl-pyrimidine (2) (purity: 50.0%, 6.0 g, 0.0151 mol) in DCM (60.0 mL) at −30° C. for 0.5 hour. TLC showed the reaction was completed. 02 and N2 was bubbled into the above solution at 10° C. for 0.5 hr. (CH3)2S (10.0 mL, 0.0151 mol) was added into the reaction and the mixture was stirred for 0.5 hr at 20° C. TLC showed the reaction was completed. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give ethyl 2-chloro-5-methylpyrimidine-4-carboxylate (1.70 g, yield: 56.1%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ 8.62 (s, 1H), 4.48 (q, J=7.2 Hz, 2H), 2.50 (s, 3H), 1.44 (t, J=7.2 Hz, 3H).
  • Ethyl 2-(N-(2,4-dimethoxybenzyl)methylsulfonamido)-5-methylpyrimidine-4-carboxylate (4)
  • To a mixture of ethyl 2-chloro-5-methylpyrimidine-4-carboxylate (3) (0.20 g, 0.997 mmol) and N-[(2,4-dimethoxyphenyl)methyl]methanesulfonamide (0.49 g, 1.99 mmol) in DMF (5.0 mL) was added NaH (60.0%, 0.076 g, 1.99 mmol) at 0° C. The mixture was stirred at 60° C. for 6 hrs. LCMS showed the reaction was completed. The mixture was quenched by saturated aqueous NH4Cl (10 mL) and extracted with EtOAc (15 mL×3). The combined organic layers were combined and washed with brine (15 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give ethyl 2-(N-(2,4-dimethoxybenzyl)methylsulfonamido)-5-methylpyrimidine-4-carboxylate (0.180 g, yield: 44.1%) as colorless oil. 1H NMR (400 MHz, CDCl3): δ 8.53 (s, 1H), 7.33-7.29 (m, 1H), 6.42-6.37 (m, 2H), 5.29 (s, 2H), 4.44 (q, J=7.2 Hz, 2H), 3.78 (s, 3H), 3.66 (s, 3H), 3.36 (s, 3H), 2.44 (s, 3H), 1.43 (t, J=7.2 Hz, 3H).
  • N-(2,4-Dimethoxybenzyl)-N-(4-(hydroxymethyl)-5-methylpyrimidin-2-yl)methanesulfonamide (5)
  • To a solution of ethyl 2-(N-(2,4-dimethoxybenzyl)methylsulfonamido)-5-methylpyrimidine-4-carboxylate (4) (0.18 g, 0.44 mmol)) in THF (5 mL) was added LiBH4 (0.038 g, 1.8 mmol) in portions at 0° C. The mixture was stirred at 20° C. for 2 hrs. TLC showed the reaction was completed. The mixture was quenched by saturated aqueous NH4Cl (20 mL), extracted with EtOAc (10 mL×2). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give N-(2,4-dimethoxybenzyl)-N-(4-(hydroxymethyl)-5-methylpyrimidin-2-yl) methanesulfonamide (0.080 g, yield: 50%) as colorless oil. 1H NMR (400 MHz, CDCl3): δ 8.29 (s, 1H), 7.25 (br s, 1H), 6.45-6.41 (m, 2H), 5.31 (s, 2H), 4.63 (d, J=4.8 Hz, 2H), 4.03 (t, J=4.8 Hz, 1H), 3.78 (s, 3H), 3.74 (s, 3H), 3.41 (s, 3H), 2.12 (s, 3H).
  • (2-(N-(2,4-Dimethoxybenzyl)methylsulfonamido)-5-methylpyrimidin-4-yl)methyl methanesulfonate (6)
  • To a solution of N-(2,4-dimethoxybenzyl)-N-(4-(hydroxymethyl)-5-methylpyrimidin-2-yl)methanesulfonamide (5) (0.10 g, 0.272 mmol) in DCM (2 mL) was added TEA (0.0759 mL, 0.544 mmol) and MsCl (0.047 g, 0.41 mmol) at 0° C. The mixture was stirred at 20° C. for 0.5 hr under N2 atmosphere. LCMS showed the reaction was completed. The reaction was quenched by saturated aqueous NH4Cl solution (2 mL), extracted with DCM (3 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure to give (2-(N-(2,4-dimethoxybenzyl)methylsulfonamido)-5-methylpyrimidin-4-yl)methyl methanesulfonate (0.15 g, crude). 1H NMR (400 MHz, CDCl3): δ 8.39 (s, 1H), 7.24 (s, 1H), 6.39 (s, 2H), 5.29-5.28 (m, 2H), 4.54 (s, 2H), 3.78 (s, 6H), 3.62 (s, 3H), 3.28 (s, 3H), 2.80 (s, 3H).
  • N-(4-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-5-methylpyrimidin-2-yl)-N-(2,4-dimethoxybenzyl)methanesulfonamide (7)
  • To a solution of (2-(N-(2,4-dimethoxybenzyl)methylsulfonamido)-5-methylpyrimidin-4-yl)methyl methanesulfonate (6) (0.10 g, 0.22 mmol) in DMF (2.0 mL) was added Cs2CO3 (0.15 g, 0.45 mmol) and 3-chloro-2-(2-chloroethoxy)-5-[1-(4-hydroxyphenyl)-1-methyl-ethyl]benzonitrile (Int H) (0.079 g, 0.22 mmol) at 20° C. The mixture was stirred at 20° C. for 10 hrs. TLC showed the reaction was completed. The mixture was quenched with H2O (3 mL), extracted with EtOAc (3 mL×3). The combined organic layers were washed with brine (4 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure to give N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-5-methylpyrimidin-2-yl)-N-(2,4-dimethoxybenzyl)methanesulfonamide (0.10 g, crude) as yellow oil which was used directly without further purification.
  • N-(4-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-5-methylpyrimidin-2-yl)methanesulfonamide (A122)
  • To a solution of N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-5-methylpyrimidin-2-yl)-N-(2,4-dimethoxybenzyl)methanesulfonamide (7) (0.05 g, crude) in DCM (1 mL) was added TFA (0.0159 mL, 0.214 mmol) and the mixture was stirred at 20° C. for 15 min. LCMS showed the reaction was completed. The mixture was quenched with H2O (2 mL), extracted with DCM (3 mL×3). The combined organic layers were washed with brine (2 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-5-methylpyrimidin-2-yl)methanesulfonamide (purify: 100%, 7 mg, yield: 17.8%) as white solid. 1H NMR (400 MHz, CDCl3): δ 8.41 (s, 2H), 8.25-8.30 (br, 1H), 7.44 (d, J=2.4 Hz, 1H), 7.31 (d, J=2.4 Hz, 1H), 7.11 (d, J=6.8 Hz, 2H), 6.92 (d, J=6.8 Hz, 2H), 5.12 (s, 2H), 4.42 (t, J=6.1 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 3.41 (s, 3H), 2.35 (s, 3H), 1.64 (s, 6H). LCMS: (220 nm):100%. Exact Mass: 548.1; found 549.2, 551.1.
  • Example 24: Synthesis of N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-2-yl)methanesulfonamide (A134)
  • Figure US20200123117A1-20200423-C00349
  • 5-(2-(4-((2-Aminothiazol-4-yl)methoxy)phenyl)propan-2-yl)-3-chloro-2-(2-chloro ethoxy)benzonitrile (3)
  • To a mixture of 4-(chloromethyl)thiazol-2-aminehydrochloride (1) (200 mg, 1.08 mmol) and 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (2) (378 mg, 1.08 mmol) in DMF (5 mL) was added Cs2CO3 (1.4 g, 4.32 mol) at 20° C. and the mixture was stirred at 130° C. for 10 hrs. LCMS showed the reaction was completed. The reaction was cooled down, quenched with water (10 mL). The mixture was extracted with DCM (5 mL×3). The combined organic layers were washed with brine (5 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by HPLC (FA) to obtain 5-(2-(4-((2-aminothiazol-4-yl)methoxy)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy) benzonitrile (3) (15.0 mg, yield: 3%) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=7.44 (s, 1H), 7.32 (s, 1H), 7.11 (br d, J=6.6 Hz, 2H), 6.93 (br s, 2H), 6.53 (br s, 1H), 5.06-4.83 (m, 2H), 4.42 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 1.64 (s, 6H).
  • N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-2-yl)methanesulfonamide (A134)
  • To a mixture of 5-(2-(4-((2-aminothiazol-4-yl)methoxy)phenyl)propan-2-yl)-3-chloro-2-(2-chloroethoxy)benzonitrile (3) (15.0 mg, 0.03 mmol) and TEA (0.01 mL, 0.10 mmol) in DCM (1 mL) was added methanesulfonyl chloride (4.46 mg, 0.03 mmol) and the mixture was stirred at 25° C. for 30 min (two Ms groups are installed in LCMS). Then Cs2CO3 (21.1 mg, 0.0649 mol) in H2O (0.5 mL) was added into the reaction. The mixture was stirred at 50° C. for 10 min. LCMS showed the reaction was completed. The reaction was quenched with water (2 mL) and extracted with DCM (3 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 prep-HPLC (HCOOH) to obtain N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-2-yl) methanesulfonamide (A134) (1.8 mg, yield: 10.3%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.44 (d, J=2.2 Hz, 1H), 7.32 (d, J=2.2 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 6.90 (d, J=8.8 Hz, 2H), 6.49 (s, 1H), 4.98 (s, 2H), 4.43 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 3.01 (s, 3H), 1.65 (s, 6H). LCMS: (220 nm): 94.9%. Exact Mass: 539.1; found 540.0/542.0.
  • Example 25: Synthesis of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((4-(methylsulfonyl)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A136)
  • Figure US20200123117A1-20200423-C00350
  • (4-Bromothiazol-2-yl)methanol (2)
  • To a solution of 4-bromothiazole-2-carbaldehyde (10.4 g, 0.0542 mol) in MeOH (150 mL) was added NaBH4 (4.10 g, 0.108 mol) at 0° C. The mixture was stirred at 25° C. for 2 hrs. TLC showed the reaction was completed and one new spot was observed. The mixture was quenched with water (70 mL), stirred for 0.5 hr and concentrated to remove most of MeOH. The aqueous layer was extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (SiO2, Petroleum ether/Ethyl acetate=10: to 3:1) to give (4-bromothiazol-2-yl)methanol (10.0 g, 0.0515 mol, yield: 95.2%) as white solid. 1H NMR (400 MHz, CDCl3) δ 7.23 (s, 1H), 4.96 (d, J=6.0 Hz, 2H), 2.79 (t, J=6.0 Hz, 1H).
  • 4-Bromo-2-(((tert-butyldimethylsilyl)oxy)methyl)thiazole (3)
  • To a solution of (4-bromothiazol-2-yl)methanol (11.7 g, 0.0603 mol) in DCM (120 mL) was added imidazole (8.21 g, 0.121 mol) and tert-butyl-chloro-dimethyl-silane (10.0 g, 0.0663 mol). The mixture was stirred at 25° C. for 1 hr. TLC showed the starting material was consumed and one new spot was observed. The reaction was poured into water (100 mL), extracted with DCM (70 mL×2). The combined organic layers were washed with brine (70 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give (4-bromothiazol-2-yl)methoxy-tert-butyl-dimethyl-silane (17.0 g, 0.0551 mol, yield: 91.5%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.18 (s, 1H), 4.95 (s, 2H), 0.96 (s, 9H), 0.14 (s, 6H).
  • 2-(((tert-Butyldimethylsilyl)oxy)methyl)-4-(methylthio)thiazole (4)
  • To a solution of (4-bromothiazol-2-yl)methoxy-tert-butyl-dimethyl-silane (11.5 g, 0.0373 mol) in MTBE (300 mL) was added n-BuLi (2.50 M, 17.9 mL, 0.0448 mol) dropwise at −78° C. under N2 atmosphere. The mixture was stirred at −78° C. for 30 min. Then MeSSMe (14.3 g, 0.152 mol) was added at −78° C. and stirred at the same temperature for 1 hr. LCMS showed 70% desired MS was detected. The mixture was quenched with sat. NH4Cl solution (100 mL), extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give crude tert-butyl-dimethyl-[(4-methylsulfanylthiazol-2-yl)methoxy]silane (90.0%, 10.0 g, 0.0327 mol, yield: 87.6%) as brown oil. 1H NMR (400 MHz, CDCl3) δ 6.87 (s, 1H), 4.95 (s, 2H), 2.53 (s, 3H), 0.96 (s, 9H), 0.14 (s, 6H).
  • (4-(Methylthio)thiazol-2-yl)methanol (5)
  • To a solution of (4-bromothiazol-2-yl)methoxy-tert-butyl-dimethyl-silane (12.0 g, 0.0389 mol) in THF (150 mL) was added TBAF (1.00 M, 46.7 mL, 0.0467 mol) at 0° C. The mixture was stirred at 20° C. for 1 hr. TLC showed the reaction was completed. The reaction was poured into water (50 mL), extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give (4-methylsulfanylthiazol-2-yl) methanol (3.60 g, 0.0223 mol, yield: 57.4%) as yellow solid. 1H NMR (400 MHz, CDCl3) δ 6.89 (s, 1H), 4.94 (br d, J=2.0 Hz, 2H), 2.95 (br s, 1H), 2.54 (s, 3H).
  • (4-(Methylthio)thiazol-2-yl)methyl methanesulfonate (6)
  • To a mixture of (4-methylsulfanylthiazol-2-yl)methanol (3.40 g, 0.0211 mol) and TEA (0.432 mL, 0.00310 mol) in DCM (50 mL) was added MsCl (1.96 mL, 0.0253 mol) at 0° C. The mixture was stirred at 0° C. for 1 hr. TLC showed the starting material was consumed and one new spot was observed. The reaction was poured into water (30 mL), extracted with DCM (15 mL×3). The combined organic layers were washed with brine (40 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (4-methylsulfanylthiazol-2-yl)methyl methanesulfonate (5.40 g, 0.0226 mol, crude) as yellow oil. 1H NMR (400 MHz, CDCl3) δ 6.99 (s, 1H), 5.45 (s, 2H), 3.10 (s, 3H), 2.55 (s, 3H).
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-((4-(methylthio)thiazol-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (7)
  • To a suspension of (4-methylsulfanylthiazol-2-yl)methyl methanesulfonate (80.0%, 3.08 g, 0.0103 mol) and Cs2CO3 (5.58 g, 0.0171 mol) in DMF (20 mL) was added 3-chloro-2-(2-chloroethoxy)-5-[1-(4-hydroxyphenyl)-1-methyl-ethyl]benzonitrile (3.00 g, 0.00857 mol) at 20° C. The mixture was stirred at 20° C. for 16 hrs. LCMS showed the reaction was completed. The reaction was poured into H2O (50 mL), extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (30 mL×3), filtered and concentrated under reduced pressure to give 3-chloro-2-(2-chloroethoxy)-5-[1-methyl-1-[4-[(4-methylsulfanylthiazol-2-yl)methoxy]phenyl]ethyl]benzonitrile (3.20 g, 0.00648 mol, yield: 75.7%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ 7.44 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.12 (d, J=8.4 Hz, 2H), 6.99-6.91 (m, 3H), 5.34 (s, 2H), 4.42 (t, J=6.4 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 2.56 (s, 3H), 1.64 (s, 6H).
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-((4-(methylsulfonyl)thiazol-2-yl)methoxy)phenyl) propan-2-yl)benzonitrile (A136)
  • To a solution of 3-chloro-2-(2-chloroethoxy)-5-[1-methyl-1-[4-[(4-methylsulfanylthiazol-2-yl)methoxy]phenyl]ethyl]benzonitrile (3.00 g, 0.00608 mol) in DCM (50 mL) was added m-CPBA (80.0%, 5.25 g, 0.0243 mol) at 0° C. The mixture was stirred at 25° C. for 3 hrs. LCMS showed the reaction was completed and 60% desired product was detected. The mixture was quenched with water (30 mL). The mixture was extracted with DCM (20 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give 3-chloro-2-(2-chloroethoxy)-5-[1-methyl-1-[4-[(4-methylsulfonylthiazol-2-yl)methoxy]phenyl]ethyl]benzonitrile (A136) (1.71 g, 0.00325 mol, yield: 53.5%) as white solid. 1H NMR (400 MHz, CDCl3) δ 8.22 (s, 1H), 7.45 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.0 Hz, 1H), 7.15 (d, J=6.8 Hz, 2H), 6.95 (d, J=6.8 Hz, 2H), 5.39 (s, 2H), 4.43 (t, J=6.4 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.23 (s, 3H), 1.66 (s, 6H).
  • Example 26: Synthesis of N-(2-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-4-yl)methanesulfonamide (A137)
  • Figure US20200123117A1-20200423-C00351
  • Ethyl 2-methylthiazole-4-carboxylate (2)
  • To a mixture of ethyl 2-methyloxazole-4-carboxylate (1) (10.0 g, 58.4 mmol) and AIBN (0.959 g, 5.84 mmol) in CCl4 (80 mL) was added NBS (12.5 g, 70.1 mmol) at 20° C. and the mixture was stirred at 80° C. for 16 hrs. TLC showed a new spot appeared and trace of starting material was remained. The reaction was poured into H2O (150 mL) and extracted with DCM (80 mL×2). The combined organic layers were washed with brine (80 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel to give ethyl 2-methyloxazole-4-carboxylate (2) (4.6 g, Yield: 31.5%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=8.23 (s, 1H), 4.77 (s, 2H), 4.41 (q, J=6.8 Hz, 2H), 1.42 (t, J=7.2 Hz, 3H).
  • Ethyl 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl) phenoxy) methyl)thiazole-4-carboxylate (3)
  • A solution of 3-chloro-2-(2-chloroethoxy)-5-(1-(4-hydroxyphenyl)-1-methyl-ethyl)benzonitrile (int H) (0.5 g, 1.43 mmol), ethyl 2-(bromomethyl) thiazole-4-carboxylate (2) (0.428 g, 1.71 mmol) and Cs2CO3 (0.93, 2.86 mmol) in DMF (10 mL) was stirred at 20° C. for 16 hrs. LCMS showed the reaction was completed. The reaction was poured into H2O (15 mL) and extracted with EtOAc (15 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 to give ethyl 2-((4-(1-(3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl)-1-methyl-ethyl)phenoxy)methyl)thiazole-4-carboxylate (3) (0.6 g, Yield: 80.9%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=8.27 (s, 1H), 7.44 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.12 (d, J=8.8 Hz, 2H), 5.41 (s, 2H), 4.44-4.09 (m, 4H), 3.88 (t, J=6.4 Hz, 2H), 1.65 (s, 1H), 1.43 (t, J=7.6 Hz, 3H).
  • 2-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxylic acid (4)
  • A solution of ethyl 2-((4-(1-(3-chloro-4-(2-chloroethoxy)-5-cyano-phenyl)-1-methyl-ethyl)phenoxy)methyl)thiazole-4-carboxylate (3) (0.5 g, 0.963 mmol) and LiOH.H2O (0.162 g, 3.85 mmol) in THF (3 mL) and H2O (3 mL) was stirred at 20° C. for 16 hrs. LCMS showed the reaction was completed. The reaction was poured into H2O (8 mL). The pH of the aqueous layer was adjusted to 3-4 by adding aqueous HCl solution (3M). The aqueous layer was separated and extracted with EtOAc (8 mL×3). The combined organic layers were washed with brine (8 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxylic acid (4) (0.4 g, yield: 84.6%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.34 (s, 1H), 7.44 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.14 (d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 5.41 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 1.65 (s, 6H).
  • tert-Butyl (2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl) phenoxy)methyl)thiazol-4-yl)carbamate (5)
  • To a solution of 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxylic acid (4) (0.1 g, 0.204 mmol) and TEA (0.0851 mL, 0.000611 mol) in toluene (1.0 mL) was added t-BuOH (1.0 mL) and DPPA (0.132 mL, 0.611 mmol) under N2 atmosphere. The mixture was stirred at 90° C. for 4 hrs. LCMS showed the starting material was consumed. The mixture was poured into H2O (6 mL), extracted with DCM (3 mL×2). And the combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel tert-butyl (2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-4-yl)carbamate (5) (0.11 g, yield: 96.1%) as white oil. 1H NMR (400 MHz, CDCl3) δ=7.43 (d, J=2.4 Hz, 1H), 7.33 (d, J=2.4 Hz, 1H), 7.25 (s, 1H), 7.11 (d, J=8.8 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 5.25 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 1.65 (s, 6H), 1.53 (s, 9H).
  • 5-(2-(4-((4-Aminothiazol-2-yl)methoxy)phenyl)propan-2-yl)-3-chloro-2-propoxybenzonitrile (6)
  • To a solution of tert-butyl (2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-4-yl)carbamate (5) (0.1 g, 0.0178 mmol) in DCM (1 mL) was added TFA (0.1 mL) and stirred at 25° C. for 2 hrs. LCMS showed the reaction was completed. The reaction was poured into saturated aqueous NaHCO3 solution (4 mL) and extracted with DCM (2 mL×3). The combined organic layers were washed with brine (2 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 5-(2-(4-((4-aminothiazol-2-yl)methoxy)phenyl)propan-2-yl)-3-chloro-2-propoxybenzonitrile (6) (30 mg, Yield: 35.1%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.44 (d, J=2.0 Hz, 1H), 7.33 (d, J=2.4 Hz, 1H), 7.25 (s, 1H), 7.11 (d, J=8.8 Hz, 2H), 6.94 (d, J=8.8 Hz, 2H), 5.24 (s, 2H), 4.42 (t, J=2.4 Hz, 2H), 3.88 (t, J=2.4 Hz, 2H), 1.65 (s, 6H)
  • N-(2-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazol-4-yl)methanesulfonamide (A137)
  • To a solution of 5-(2-(4-((4-aminothiazol-2-yl)methoxy)phenyl)propan-2-yl)-3-chloro-2-propoxybenzonitrile (6) (50 mg, 0.108 mmol) and TEA (21.9 mg, 0,216 mmol) in DCM (2 mL) was added methanesulfonyl chloride (18.6 mg, 0.0649 mmol) dropwisely at 0° C. under nitrogen and the mixture was stirred at 25° C. for 2 hrs. (two Ms groups were installed). Then the mixture was added Cs2CO3 (0.141 g, 0.433 mmol) and H2O (1 mL) and stirred at 25° C. for 2 hrs. LCMS showed the reaction was completed. The reaction was poured into H2O (4 mL). The aqueous layer was extracted with EtOAc (4 mL×3). The combined organic layers were washed with brine (4 m), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC (HCOOH) to give product N-(2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy) methyl)thiazol-4-yl)methanesulfonamide (A137) (10 mg, yield: 17.1%) as white solid. 1H NMR (400 MHz, CDCl3) δ=7.44 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 7.04 (s, 1H), 7.00 (s, 1H), 6.94 (d, J=8.8 Hz, 2H), 5.29 (s, 2H), 4.46 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.12 (s, 3H), 1.65 (s, 6H). LCMS: (220 nm): 93.78%. Exact Mass: 540.5; found: 540.1/542.1.
  • Example 27: Synthesis of 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxamide (A171)
  • Figure US20200123117A1-20200423-C00352
  • To a solution of 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxylic acid (4) (20 mg, 0.0407 mmol), NH4Cl (5.4 mg, 0.102 mmol), HATU (0.0186 g, 0.0488 mmol) in DCM (1.00 mL) was added TEA (8.2 mg, 0.0814 mmol) at 20° C. The mixture was stirred at same temperature for 2 hrs. LCMS showed the starting material was consumed. The mixture was poured into H2O (4 mL) and extracted with DCM (3 mL×2). The combined organic layers were washed with brine (4 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified prep-HPLC (HCOOH) to give 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxamide (A171) (9.2 mg, yield: 46.1%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.19 (s, 1H), 7.44 (d, J=2.2 Hz, 1H), 7.32 (d, J=2.2 Hz, 1H), 7.14 (br d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 5.60 (br s, 1H), 5.34 (s, 2H), 4.43 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 1.66 (s, 6H). LCMS (220 nm): 99.5%. Exact Mass: 489.0; found 490.
  • Example 28: Synthesis of 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carbonitrile (A172)
  • Figure US20200123117A1-20200423-C00353
  • To a solution 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carboxamide (A171) (20 mg, 0.0408 mmol) and pyridine (3.27 mg, 0.0413 mmol) in dioxane (2.0 mL) was added TFAA (17.4 mg, 0.00827 mmol) under N2 at 0° C. Then the reaction was stirred at 20° C. for 16 hrs. LCMS showed the reaction was completed. The reaction was poured into H2O (4 mL). The reaction mixture were separated and extracted with EtOAc (4 mL×3). The organic layers were washed with brine (4 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC (HCOOH) to give the product 2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)thiazole-4-carbonitrile (A172) (10 mg, yield: 52.1%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.05 (s, 1H), 7.44 (d, J=2.2 Hz, 1H), 7.31 (d, J=2.2 Hz, 1H), 7.15 (d, J=8.8 Hz, 2H), 6.94 (d, J=8.8 Hz, 2H), 5.37 (s, 2H), 4.43 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 1.65 (s, 6H). LCMS (220 nm): 100.0%. Exact Mass: 471.0; found 472.0.
  • Example 29: Synthesis of N-(4-(aminomethyl)-6-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (A164)
  • Figure US20200123117A1-20200423-C00354
  • 2,4-Dichloro-6-(chloromethyl)pyrimidine (2)
  • A suspension of 6-(chloromethyl)pyrimidine-2,4-diol (1) (1.50 g, 9.34 mmol) in POCl3 (10 mL) was stirred at 100° C. for 3 hrs. TLC showed the reaction was completed. The solid was dissolved and the reaction became brown. The resulting mixture was cooled down, concentrated under reduced pressure. The residue was quenched with water (20 mL) and extracted with EtOAc (10 mL×2). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC (petroleum ether/EtOAc=10/1) to obtain 2,4-dichloro-6-(chloromethyl)pyrimidine (2) (1.5 g, yield: 81.3%) as light yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.55 (s, 1H), 4.59 (s, 2H).
  • 2-((2,6-Dichloropyrimidin-4-yl)methyl)isoindoline-1,3-dione (4)
  • A suspension of 2,4-dichloro-6-(chloromethyl)pyrimidine (2) (1.5 g, 7.60 mmol) and potassium 1,3-dioxoisoindolin-2-ide (3) (1.55 g, 8.36 mmol) in DMF (20 mL) was stirred at 20° C. for 2 hrs. LCMS showed the reaction was completed. The reaction was quenched with water (60 mL) and extracted with EtOAc (40 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by R-MPLC to obtain 2-((2,6-dichloropyrimidin-4-yl)methyl)isoindoline-1,3-dione (4) (1.5 g, yield: 64.1%) as yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=8.00 (s, 1H), 7.96-7.92 (m, 2H), 7.92-7.87 (m, 2H), 4.92 (s, 2H).
  • 2-((2-Chloro-6-vinylpyrimidin-4-yl)methyl)isoindoline-1,3-dione (5)
  • A suspension of 2-((2,6-dichloropyrimidin-4-yl)methyl)isoindoline-1,3-dione (4) (1.5 g, 4.87 mmol), potassium; trifluoro(vinyl)boranuide (0.587 g, 4.38 mmol), TEA (0.746 mL, 5.36 mmol) and Pd(dppf)Cl2 (0.249 g, 0.34 mmol) in n-PrOH (80 mL) was stirred at 100° C. for 2 hrs under N2 atmosphere. LCMS showed the reaction was completed. Then the resulting mixture was quenched with saturated aqueous NH4Cl (50 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (80 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to obtain 2-((2-chloro-6-vinylpyrimidin-4-yl)methyl)isoindoline-1,3-dione (5) (˜80.0% in H NMR, 0.8 g) as yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=7.96-7.93 (m, 2H), 7.92-7.88 (m, 2H), 7.72 (s, 1H), 6.82-6.73 (m, 1H), 6.55 (dd, J=1.0, 17.4 Hz, 1H), 5.83 (dd, J=1.0, 10.7 Hz, 1H), 4.90 (s, 2H).
  • 2-Chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidine-4-carbaldehyde (6)
  • To a solution of 2-((2-chloro-6-vinylpyrimidin-4-yl)methyl)isoindoline-1,3-dione (5) (80.0%, 0.7 g, 1.87 mmol) in DCM (20 mL) was bubbled with O3 at −78° C. for 15 min. Then the solution was bubbled with O2 for 10 min at same temperature until the reaction turned colorless. Me2S (2 mL) was added at −78° C. and the mixture was stirred at 20° C. for 1 hr. TLC showed the reaction was completed. The resulting mixture was quenched with water (20 mL) and extracted with DCM (10 mL×2). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give 2-chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidine-4-carbaldehyde (6) (470 mg, yield: 83.4%) as yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=9.96 (s, 1H), 7.95 (dd, J=3.2, 5.4 Hz, 2H), 7.82 (dd, J=2.8, 5.6 Hz, 2H), 7.63 (s, 1H), 5.08 (s, 2H).
  • 2-((2-Chloro-6-(hydroxymethyl)pyrimidin-4-yl)methyl)isoindoline-1,3-dione (7)
  • To a solution of 2-chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidine-4-carbaldehyde (6) (490 mg, 1.62 mmol) in THF (10 mL) was added NaBH4 (24.7 mg, 0.65 mmol) in water (0.5 mL) dropwise at 0° C. The reaction was stirred at same temperature for 15 min. LCMS showed the reaction was completed. The residue was partitioned between EtOAc (10 mL) and water (10 mL). The aqueous layer was extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 2-((2-chloro-6-(hydroxymethyl)pyrimidin-4-yl)methyl)isoindoline-1,3-dione (7) (430 mg, yield: 78.5%) as light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=7.98-7.94 (m, 2H), 7.93-7.89 (m, 2H), 7.61 (s, 1H), 5.76 (t, J=5.9 Hz, 1H), 4.95 (s, 2H), 4.54 (d, J=5.9 Hz, 2H).
  • (2-Chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-4-yl)methyl methanesulfonate (8)
  • To a solution of 2-((2-chloro-6-(hydroxymethyl)pyrimidin-4-yl)methyl)isoindoline-1,3-dione (7) (410 mg, 1.35 mmol)) and TEA (0.3 mL, 2.03 mmol) in DCM (10 mL) was added MsCl (170 mg, 1.49 mmol) at 0° C. The reaction was stirred at same temperature for 20 min. TLC showed the reaction was completed. The reaction was quenched with saturated aqueous NH4Cl (10 mL) and extracted with DCM (10 mL×2). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (2-chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-4-yl)methylmethanesulfonate (8) (500 mg, yield: 97.0%) as yellow oil, which was used for next step directly. 1H NMR (400 MHz, CDCl3) δ=7.94 (dd, J=2.8, 5.4 Hz, 2H), 7.83-7.79 (m, 2H), 7.35 (s, 1H), 5.25 (s, 2H), 5.01 (s, 2H), 3.15 (s, 3H).
  • 3-Chloro-5-(2-(4-((2-chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-4-yl)methoxy) phenyl)propan-2-yl)-2-(2-chloroethoxy)benzonitrile (9)
  • A suspension of (2-chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-4-yl)methyl methanesulfonate (8) (500 mg, 1.31 mmol), 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (int H) (436 mg, 1.24 mmol) and Cs2CO3 (853 mg, 2.62 mmol) in DMF (10 mL) was stirred at 20° C. for 16 hrs. Then the mixture was stirred at 40° C. for 0.5 hrs. LCMS showed the reaction was completed. The resulting mixture was quenched with saturated aqueous NH4Cl solution (10 mL) and water (10 mL). The aqueous layer was extracted with EtOAc (10 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to obtain 3-chloro-5-(2-(4-((2-chloro-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-4-yl)methoxy)phenyl)propan-2-yl)-2-(2-chloroethoxy)benzonitrile (9) (300 mg, 36.0%) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=7.92 (dd, J=3.0, 5.4 Hz, 2H), 7.80 (dd, J=3.0, 5.4 Hz, 2H), 7.46-7.43 (m, 2H), 7.33 (d, J=2.4 Hz, 1H), 7.11 (d, J=8.8 Hz, 2H), 6.86 (d, J=8.8 Hz, 2H), 5.11 (s, 2H), 4.99 (s, 2H), 4.43 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 1.65 (s, 6H).
  • N-(4-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2-yl)methanesulfonamide (10)
  • A suspension of N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2-yl)methanesulfonamide (10) (180 mg, 0.28 mmol), methanesulfonamide (80.8 mg, 0.85 mmol), Cs2CO3 (184 mg, 0.57 mmol), Xantphos (32.8 mg, 0.056 mmol) and Pd2(dba)3 (25.9 mg, 0.028 mmol) in dioxane (6 mL) was stirred at 100° C. for 2 hrs. TLC showed the reaction was completed. The resulting mixture was cooled down, quenched with saturated aqueous NH4Cl (5 mL) and water (5 mL). The aqueous layer was extracted with EtOAc (5 mL×3). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to obtain N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2-yl)methanesulfonamide (10) (150 mg, yield: 76.3%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.92 (dd, J=3.2, 5.4 Hz, 2H), 7.79 (dd, J=3.2, 5.4 Hz, 2H), 7.45 (d, J=2.2 Hz, 1H), 7.34 (d, J=2.6 Hz, 1H), 7.25 (s, 1H), 7.12 (d, J=8.8 Hz, 2H), 6.89 (d, J=8.8 Hz, 2H), 5.05 (s, 2H), 4.96 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.25 (s, 3H), 1.66 (s, 6H).
  • N-(4-(Aminomethyl)-6-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (A164)
  • A solution of N-(4-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)-6-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2-yl)methanesulfonamide (10) (50 mg, 0.072 mmol) and NH2NH2H2O (80.0%, 0.0218 mL, 0.36 mmol) in MeOH (1 mL) and DCM (0.5 mL) was stirred at 20° C. for 1 hr. LCMS showed the reaction was completed. The resulting mixture was adjusted pH to 4-5 by adding aqueous HCl (1 N) and the residue was purified by prep-HPLC (HCl) to obtain N-(4-(aminomethyl)-6-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl) phenoxy)methyl)pyrimidin-2-yl)methanesulfonamide (A164) (5.54 mg, yield: 13.6%) as white solid. 1H NMR (400 MHz, DMSO-d6) δ=11.52 (s, 1H), 8.45 (s, 3H), 7.63 (s, 1H), 7.58 (s, 1H), 7.39 (s, 1H), 7.20 (d, J=8.4 Hz, 2H), 6.98 (d, J=8.8 Hz, 2H), 5.14 (s, 2H), 4.41 (t, J=4.8 Hz, 2H), 4.22 (s, 2H), 3.95 (t, J=4.8 Hz, 2H), 3.42 (s, 3H), 1.63 (s, 6H). LCMS (220 nm): 99.7%. Exact Mass: 563.12; found 564.1.
  • Example 30: Synthesis of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylsulfonyl)furan-2-yl) methoxy)phenyl)propan-2-yl)benzonitrile (A168)
  • Figure US20200123117A1-20200423-C00355
  • Methyl 5-(methylsulfonyl)furan-2-carboxylate (2)
  • A suspension of methyl 5-nitrofuran-2-carboxylate (1.0 g, 4.9 mmol), sodium methanesulfonate (115 mg, 9.8 mmol) and CuI (139 mg, 7.3 mmol) in DMSO (30 mL) was stirred at 100° C. for 6 hrs under N2 atmosphere. TLC showed the reaction was completed. The mixture was cooled down. The reaction was diluted with EtOAc (30 mL) and quenched with water (70 mL). The mixture was filtered, and the filtrate was extracted with EtOAc (10 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to give methyl 5-(methylsulfonyl)furan-2-carboxylate (0.3 g, yield: 30.1%) as brown oil. 1H NMR (400 MHz, CDCl3) δ=7.25 (d, J=3.4 Hz, 1H), 7.22 (d, J=3.4 Hz, 1H), 3.96 (s, 3H), 3.23 (s, 3H).
  • (5-(Methylsulfonyl)furan-2-yl)methanol (3)
  • To a solution of methyl 5-(methylsulfonyl)furan-2-carboxylate (0.3 g, 1.5 mmol) in THF (3 mL) was added LiBH4 (128 mg, 5.9 mmol) under N2 atmosphere at 20° C. The mixture was stirred at 50° C. for 1 hr. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl (5 mL) and extracted with EtOAc (3 mL×3). The combined organic layers were washed with brine (3 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (5-(methylsulfonyl)furan-2-yl)methanol (220 mg, yield: 85.0%) as brown oil. 1H NMR (400 MHz, CDCl3) δ=7.14 (d, J=3.4 Hz, 1H), 6.48 (d, J=3.4 Hz, 1H), 4.72 (d, J=6.0 Hz, 2H), 3.17 (s, 3H), 1.97 (t, J=6.4 Hz, 1H).
  • (5-(Methylsulfonyl)furan-2-yl)methyl methanesulfonate (4)
  • To a solution of (5-(methylsulfonyl)furan-2-yl)methanol (110 mg, 0.6 mmol) in DCM (2 mL) was added TEA (0.18 g, 1.87 mmol) and MsCl (107 mg, 0.936 mol) at 0° C. under N2 atmosphere. The mixture was stirred at 20° C. for 30 min. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl solution (2 mL). The mixture was extracted with DCM (2 mL×3). The combined organic layers were washed with brine (1 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (5-(methylsulfonyl)furan-2-yl)methyl methanesulfonate (80.0%, 150 mg, yield: 75.6%) as light yellow solid. 1H NMR (400 MHz, CDCl3) δ=7.17 (d, J=3.4 Hz, 1H), 6.70 (d, J=3.4 Hz, 1H), 5.25 (s, 2H), 3.19 (s, 3H), 3.07 (s, 3H)
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylsulfonyl)furan-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A168)
  • To a solution of (5-(methylsulfonyl)furan-2-yl)methyl methanesulfonate (150 mg, 0.6 mmol) in DMF (2.00 mL) was added 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (207 mg, 0.6 mmol) and Cs2CO3 (384 mg, 1.2 mmol) under N2 atmosphere at 20° C. The mixture was stirred at 20° C. for 16 hrs. LCMS showed the reaction was completed. The mixture was poured into H2O (5 mL). The mixture was extracted with EtOAc (3 mL×2). The combined organic layers were washed with brine (2 mL×4), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((5-(methylsulfonyl)furan-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A168) (62 mg, yield: 20.7%) as white solid. 1H NMR (400 MHz, CDCl3) δ=7.44 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.17 (d, J=3.2 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 6.90 (d, J=8.8 Hz, 2H), 6.59 (d, J=3.2 Hz, 1H), 5.07 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.18 (s, 3H), 1.65 (s, 6H). LCMS: (220 nm): 32.43%. Exact Mass: 507.1; found 508.1.
  • Example 31: Synthesis of (R)—N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)ethyl)pyrimidin-2-yl)methanesulfonamide (A169) and (S)—N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)ethyl)pyrimidin-2-yl)methanesulfonamide (A170)
  • Figure US20200123117A1-20200423-C00356
    Figure US20200123117A1-20200423-C00357
  • 2-Chloro-4-(1-ethoxyvinyl)pyrimidine (2)
  • To a solution of 2, 4-dichloropyrimidine (1) (20.0 g, 0.134 mol) in DMF (250 mL) was added tributyl(1-ethoxyvinyl)stannane (53.3 g, 0.148 mol), Pd(PPh3)4 (7.7 g, 0.0067 mol) and KF (23.4 g, 0.403 mol) at 20° C. under N2. The mixture was stirred at 100° C. for 2 hrs under N2 atmosphere. TLC showed the reaction was completed. The mixture was poured into water (500 mL). The reaction mixture was filtered and the filter cake was washed with 200 mL water, dried in vacuum to give 2-chloro-4-(1-ethoxyvinyl)pyrimidine (2) (25.0 g, crude) as yellow solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.79 (d, J=5.0 Hz, 1H), 7.64 (d, J=5.0 Hz, 1H), 5.54 (d, J=2.2 Hz, 1H), 4.72 (d, J=2.2 Hz, 1H), 3.95 (q, J=6.8 Hz, 2H), 1.34 (t, J=7.0 Hz, 3H).
  • 1-(2-Chloropyrimidin-4-yl)ethanone (3)
  • To a solution of 2-chloro-4-(1-ethoxyvinyl) pyrimidine (2) (25 g, crude) in MeCN (100 mL) was added HCl (10.0%, 100 mL) at 20° C. The mixture was stirred at 75° C. for 1 hour. TLC showed the reaction was completed. The mixture was concentrated to remove MeCN. The mixture was extracted with EtOAc (50 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC (petroleum ether:EtOAc=10:1 to 5:1) to give 1-(2-chloropyrimidin-4-yl)ethanone (3) (11.0 g, over two steps yield: 52.3%) as white solid. 1H NMR (400 MHz, CDCl3) δ ppm 8.79 (d, J=4.89 Hz, 1H), 7.77 (d, J=4.89 Hz, 1H), 2.61-2.68 (m, 3H).
  • 1-(2-Chloropyrimidin-4-yl)ethanol (4)
  • To a solution of 1-(2-chloropyrimidin-4-yl)ethanone (3) (11.0 g, 0.0703 mol) in THF (50.0 mL) and H2O (50.0 mL) was added NaBH4 (2.92 g, 0.0773 mol) at 0° C. The mixture was stirred for 1 hour at 20° C. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl (100 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column (petroleum ether:EtOAc=10:1 to 5:1) to give 1-(2-chloropyrimidin-4-yl)ethanol (4) (5.50 g, yield: 49.4%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ ppm 8.62 (d, J=5.0 Hz, 1H), 7.38 (d, J=5.0 Hz, 1H), 4.84-4.94 (m, 1H), 2.98 (d, J=5.0 Hz, 1H), 1.55 (d, J=6.8 Hz, 2H), 1.52-1.53 (m, 1H).
  • 1-(2-Chloropyrimidin-4-yl)ethyl methanesulfonate (5)
  • To a solution of 1-(2-chloropyrimidin-4-yl)ethanol (4) (14.0 g, 0.0883 mol) and TEA (24.6 mL, 0.177 mol) in DCM (200 mL) was added methanesulfonyl chloride (15.2 g, 0.132 mol) at 0° C. The mixture was stirred at 20° C. for 1 hr. LCMS showed the reaction was completed. The mixture was poured into water (400 mL) and extracted with DCM (100 mL×3). The combined organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 1-(2-chloropyrimidin-4-yl)ethyl methanesulfonate (5) (19.0 g, 0.0803 mol, yield: 90.9%) as yellow oil.
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-(1-(2-chloropyrimidin-4-yl)ethoxy)phenyl)propan-2-yl)benzonitrile (6)
  • To a solution of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (int H) (20.0 g, 0.0571 mol) in DMF (250 mL) was added 1-(2-chloropyrimidin-4-yl)ethyl methanesulfonate (5) (18.9 g, 0.0799 mol) and Cs2CO3 (37.2 g, 0.114 mol) at 20° C. The mixture was stirred at 20° C. for 20 hrs and 30° C. for 4 hrs. TLC showed the reaction was completed. The mixture was poured into saturated aqueous NH4Cl solution (200 mL) and water (200 mL). The aqueous layer was extracted with EtOAc (300 mL×3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC (petroleum ether:EtOAc=10:1 to 5:1) to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(1-(2-chloropyrimidin-4-yl)ethoxy)phenyl)propan-2-yl)benzonitrile (6) (14.8 g, 0.0302 mol, yield: 52.8%) as yellow oil.
  • 1H NMR (400 MHz, CDCl3) δ=8.60 (d, J=5.2 Hz, 1H), 7.45-7.41 (m, 2H), 7.29 (d, J=2.4 Hz, 1H), 7.06 (d, J=8.8 Hz, 2H), 6.78 (d, J=8.8 Hz, 2H), 5.33-5.28 (m, 1H), 4.42 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 1.69 (d, J=6.6 Hz, 3H), 1.62 (s, 6H).
  • (R)—N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)ethyl)pyrimidin-2-yl)methanesulfonamide (A169) and (S)—N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)ethyl)pyrimidin-2-yl)methanesulfonamide (A170)
  • A suspension of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-(1-(2-chloropyrimidin-4-yl)ethoxy)phenyl)propan-2-yl)benzonitrile (6) (8.00 g, 0.0163 mol), methanesulfonamide (4.65 g, 0.0489 mol), Cs2CO3 (10.6 g, 0.0326 mol), Xantphos (1.89 g, 0.00326 mol) and Pd2(dba)3 (1.49 g, 0.00163 mol) in dioxane (250 mL) was stirred at 100° C. for 2 hrs under N2 atmosphere. TLC showed the reaction was completed. The resulting mixture was quenched with saturated aqueous NH4Cl (100 mL) and water (100 mL). The aqueous layer was extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by MPLC to obtain the crude product. The mixture was purified by p-HPLC (FA) and SFC to give (R)—N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl) phenoxy)ethyl)pyrimidin-2-yl)methanesulfonamide (1.77 g, yield: 19.8%, peak 1 in SFC was named as A169)1H NMR (400 MHz, CDCl3) δ=8.56 (d, J=5.1 Hz, 1H), 8.35 (br s, 1H), 7.43 (d, J=2.2 Hz, 1H), 7.29 (d, J=2.1 Hz, 1H), 7.17 (d, J=5.1 Hz, 1H), 7.05 (d, J=8.7 Hz, 2H), 6.79 (d, J=8.8 Hz, 2H), 5.23 (q, J=6.7 Hz, 1H), 4.42 (t, J=6.2 Hz, 2H), 3.88 (t, J=6.2 Hz, 2H), 3.49 (s, 3H), 1.67 (d, J=6.6 Hz, 3H), 1.62 (s, 6H). LCMS (220 nm): 98.4%. Exact Mass: 548.1; found 549.1. (S)—N-(4-(1-(4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)ethyl)pyrimidin-2-yl)methanesulfonamide (1.23 g, yield: 13.7%, peak 2 in SFC was named as A170) as light yellow solid. 1H NMR (400 MHz, CDCl3) δ=8.69 (br s, 1H), 8.58 (d, J=5.3 Hz, 1H), 7.43 (d, J=1.6 Hz, 1H), 7.29 (d, J=1.6 Hz, 1H), 7.17 (d, J=5.1 Hz, 1H), 7.05 (br d, J=8.6 Hz, 2H), 6.79 (d, J=8.7 Hz, 2H), 5.23 (q, J=6.3 Hz, 1H), 4.42 (t, J=6.1 Hz, 2H), 3.88 (t, J=6.1 Hz, 2H), 3.49 (s, 3H), 1.67 (d, J=6.6 Hz, 3H), 1.62 (s, 6H). LCMS (220 nm): 98.6%. Exact Mass: 548.1; found 549.1.
  • Example 32: Synthesis of N-(2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl) propan-2-yl)phenoxy)methyl)pyrimidin-4-yl)methanesulfonamide (A184)
  • Figure US20200123117A1-20200423-C00358
  • 2-(Bromomethyl)-4-chloropyrimidine (2)
  • To a mixture of 4-chloro-2-methyl-pyrimidine (10.0 g, 7.8 mmol) and AIBN (5.1 g, 31.1 mmol) in CCl4 (100 mL) was added NBS (16.6 g, 93.3 mmol) at 25° C. and the mixture was stirred at 80° C. for 16 hrs. LCMS showed the reaction was completed. The reaction mixture was cooled down to room temperature, poured into H2O (150 mL) and extracted with DCM (80 mL×2). The organic layers were combined and washed with brine (80 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 2-(bromomethyl)-4-chloropyrimidine (1.7 g, yield 10.5%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=8.64 (d, J=5.6 Hz, 1H), 7.29 (d, J=5.2 Hz, 1H), 4.56 (s, 2H).
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-((4-chloropyrimidin-2-yl)methoxy)phenyl)propan-2-yl) benzonitrile (4)
  • To a mixture of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl) propan-2-yl)benzonitrile (300 mg, 0.9 mmol) and 2-(bromomethyl)-4-chloropyrimidine (444 mg, 2.1 mmol) in DMF (5 mL) was added NaH (60%, 39 mg, 1.0 mmol) in portions at 0° C. under N2 and the mixture was stirred at 0° C. for 4 hrs. LCMS showed the reaction was completed. The reaction was poured into H2O (10 mL), extracted with EtOAc (8 mL×3). The combined organic layers were washed with brine (12 mL×4), dried over 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-((4-chloropyrimidin-2-yl) methoxy)phenyl)propan-2-yl) benzonitrile (300 mg, Yield: 73.5%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=8.68 (d, J=5.2 Hz, 1H), 7.43 (d, J=2.4 Hz, 1H), 7.33-7.32 (m, 2H), 7.09 (d, J=8.8 Hz, 2H), 6.94 (d, J=9.2 Hz, 2H), 5.27 (s, 2H), 4.42 (t, J=6.4 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 1.64 (s, 6H).
  • N-(2-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl) pyrimidin-4-yl)methanesulfonamide (A184)
  • To a solution of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((4-chloropyrimidin-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (100 mg, 0.2 mmol) in dioxane (2 mL) was added methanesulfonamide (60 mg, 6.3 mmol), Xantphos (24.3 mg, 0.06 mmol), Cs2CO3 (137 mg, 0.4 mmol) and Pd2(dba)3 (19.2 mg, 0.02 mmol) at 100° C. under N2 atmosphere. The mixture was stirred at 100° C. for 5 hrs. LCMS showed the mixture was completed. The reaction was cooled down to room temperature, quenched with water (4 mL) and extracted with DCM (3 mL×3). The combined organic layers were washed with brine (6 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude was purified by silica gel column chromatography and prep-HPLC (FA) to give N-(2-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrimidin-4-yl) methanesulfonamide (A184) (8.4 mg, yield: 7.84%) as while solid. 1H NMR (400 MHz, DMSO) δ=8.31-8.36 (m, 1H), 7.63 (d, J=2.4 Hz, 1H), 7.55 (d, J=2.4 Hz, 1H), 7.13 (d, J=8.8 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 7.06 (s, 1H), 6.93 (d, J=8.8 Hz, 2H), 5.16 (s, 2H), 4.43 (t, J=6.4 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 3.39 (s, 3H), 1.61 (s, 6H). LCMS (220 nm): 98.6%. Exact Mass: 534.10; found 535.1.
  • Example 33: Synthesis of N-(6-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrazin-2-yl)methanesulfonamide (A185)
  • Figure US20200123117A1-20200423-C00359
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-((6-chloropyrazin-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (3)
  • To a solution of (6-chloropyrazin-2-yl)methanol (1) (0.500 g, 3.46 mmol), 3-chloro-2-(2-chloroethoxy)-5-(1-(4-hydroxyphenyl)-1-methyl-ethyl)benzonitrile (2) (0.606 g, 1.73 mmol) and triphenylphosphane (1.09 g, 4.15 mmol) in THF (10.0 mL) was added DIAD (0.817 mg, 4.15 mmol) at 0° C. and stirred at 25° C. for 3 hrs under N2. TLC showed the reaction was completed. The mixture was poured into water (20 mL) and extracted with EtOAc (15 mL×3), the combined organic layers were washed with brine (5 mL×2), dried over Na2SO4, filtered and concentrated to give the crude product which was purified by silica gel column chromatography to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((6-chloropyrazin-2-yl)methoxy)phenyl)propan-2-yl)benzonitrile (3) (purity: 90.0%, 0.400 g, yield: 21.8%) as colorless oil. 1H NMR (400 MHz, CDCl3) δ=8.75 (s, 1H), 8.58 (s, 1H), 7.44 (d, J=2.4 Hz, 1H), 7.33 (d, J=2.0 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 5.22 (s, 2H), 4.43 (t, J=6.4 Hz, 2H), 3.88 (t, J=6.0 Hz, 2H), 1.65 (s, 6H).
  • N-(6-((4-(2-(3-Chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl) pyrazin-2-yl)methanesulfonamide (A185)
  • To a solution of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((6-chloropyrazin-2-yl)methoxy)phenyl)propan-2-yl) benzonitrile (3) (120 mg, 0.25 mmol) in 1,4-Dioxane (3.00 mL) was added methanesulfonamide (71.8 mg, 0.76 mmol), tris(dibenzylideneacetone)dipalladium(0) (14.5 mg, 0.025 mmol), Xantphos (14.6 mg, 0.025 mmol), and Cs2CO3 (0.164 g, 0.50 mmol) at 20° C. under N2. The mixture was stirred at 100° C. for 3 hrs. LCMS showed the reaction was completed. The reaction was poured into water (10 mL) and extracted with EtOAc (3 mL×3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give N-(6-((4-(2-(3-chloro-4-(2-chloroethoxy)-5-cyanophenyl)propan-2-yl)phenoxy)methyl)pyrazin-2-yl)methanesulfonamide (A185) (38 mg, purity: 96.0%, yield: 27.1%) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.58 (s, 1H), 8.48 (s, 1H), 7.45 (d, J=2.4 Hz, 1H), 7.32 (d, J=2.4 Hz, 1H), 7.14 (d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 5.16 (s, 2H), 4.43 (t, J=6.0 Hz, 2H), 3.88 (t, J=6.4 Hz, 2H), 3.33 (s, 3H), 1.66 (s, 6H). LCMS (220 nm): 96.8%. Exact Mass: 534.09; found 535.0.
  • Example 34: Synthesis of 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((2-methoxyoxazol-5-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A112)
  • Figure US20200123117A1-20200423-C00360
  • (2-Methoxyoxazol-5-yl)methanol (2)
  • To a mixture of ethyl 2-chlorooxazole-5-carboxylate (300 mg, 1.7 mmol) in MeOH (5 mL) was added NaOMe (138 mg, 2.6 mmol) at 25° C. and the mixture was stirred at 25° C. for 2 hrs. Then NaBH4 (259 mg, 6.8 mmol) was added to the mixture and stirred at 25° C. for another 2 hrs. TLC showed the starting material was consumed and one new spot was observed. The mixture was filtered and concentrated under reduced pressure. The residue was purified by silica-gel column chromatography to give (2-methoxyoxazol-5-yl)methanol (110 mg, yield: 49.9%) as yellow oil. 1H NMR (400 MHz, CD3OD) δ=6.74 (s, 1H), 4.44 (s, 2H), 4.04 (s, 3H).
  • 3-Chloro-2-(2-chloroethoxy)-5-(2-(4-((2-methoxyoxazol-5-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A112)
  • To a mixture of (2-methoxyoxazol-5-yl)methanol (35 mg, 0.27 mmol), 3-chloro-2-(2-chloroethoxy)-5-(2-(4-hydroxyphenyl)propan-2-yl)benzonitrile (95 mg, 0.27 mmol) and (C6H5)3P (142 mg, 0.5 mmol) in THF (4 mL) was added DIAD (0.1 mL, 0.5 mmol) at 25° C. under N2 atmosphere. The mixture was stirred at 25° C. for 2 hrs. LCMS showed the starting material was consumed and one new peak was observed. The mixture was poured into water (15 mL), extracted with EtOAc (4 mL×2). The combined organic layers were washed with brine (6 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC (FA) to give 3-chloro-2-(2-chloroethoxy)-5-(2-(4-((2-methoxyoxazol-5-yl)methoxy)phenyl)propan-2-yl)benzonitrile (A112) (9.05 mg, yield: 7.6%) as yellow oil. 1H NMR (400 MHz, CDCl3) δ=7.43 (d, J=2.4 Hz, 1H), 7.33 (d, J=2.4 Hz, 1H), 7.11 (d, J=8.8 Hz, 2H), 6.91 (d, J=2.4 Hz, 2H), 6.85 (s, 1H), 4.92 (s, 2H), 4.42 (t, J=6.4 Hz, 2H), 4.08 (s, 3H), 3.88 (t, J=6.0 Hz, 2H), 1.65 (s, 6H). LCMS (220 nm): 98.1%. Exact Mass: 460.1; found: 461.2.
  • Characterization of representative compounds synthesized are shown in Table C.
  • TABLE C
    Characterization of compounds
    Compound
    ID 1HNMR and LCMS
    A185 1H NMR (400 MHz, CDCl3): δ = 8.58 (s, 1H), 8.48 (s, 1H), 7.45 (d, J = 2.4
    Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.14 (d, J = 8.8 Hz, 2H), 6.95 (d, J = 8.8
    Hz, 2H), 5.16 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88 (t, J = 6.4 Hz, 2H), 3.33
    (s, 3H), 1.66 (s, 6H). LCMS (220 nm): 96.8%. Exact Mass: 534.09; found
    535.0, 537.0.
    A189 1H NMR (400 MHz, CDCl3): δ = 8.67 (s, 1H), 8.53 (br d, J = 6.0 Hz, 1H),
    8.02 (br d, J = 6.0 Hz, 1H), 7.87 (d, J = 8.4 Hz, 2H), 7.38 (d, J = 8.4 Hz, 2H),
    7.13 (s, 2H), 4.28 (t, J = 6.0 Hz, 2H), 3.87 (t, J = 6.8 Hz, 2H), 3.42 (s, 3H),
    1.69 (s, 6H).
    LCMS (220 nm): 95.2%. Exact Mass: 556.1; found 557.0/559.0.
    A190 1H NMR (400 MHz, CDCl3): δ = 7.51 (br s, 1H), 7.44 (d, J = 2.0 Hz, 1H),
    7.31 (d, J = 2.4 Hz, 1H), 7.10 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 8.8 Hz, 2H),
    5.04 (s, 2H), 4.84 (s, 2H), 4.43 (t, J = 6.4 Hz, 2H), 4.03 (t, J = 6.0 Hz, 2H),
    3.88 (t, J = 2.0 Hz, 2H), 3.45 (s, 3H), 2.95 (t, J = 5.6 Hz, 2H), 1.64 (s, 6H).
    LCMS (220 nm): 97.8%. Exact Mass: 590.1; found: 591.1/593.1.
    A191 1H NMR (400MHz, CDCl3): δ = 9.34 (br s, 1H), 7.62 (d, J = 2.2 Hz, 1H), 7.57
    (d, J = 2.2 Hz, 1H), 7.18 (d, J = 8.8 Hz, 2H), 6.96 (d, J = 8.8 Hz, 2H), 5.17 (s,
    2H), 4.41 (t, J = 7.2 Hz, 2H), 4.34 (s, 2H), 3.95 (t, J = 7.2 Hz, 2H), 3.51-3.42
    (m, 2H), 3.30 (s, 3H), 3.03 (br s, 2H), 1.63 (s, 6H). LCMS (220 nm): 97.6%.
    Exact Mass: 589.1; found: 590.1/592.2.
    A192 1H NMR (400MHz, DMSO-d6) δ = 7.61 (d, J = 2.2 Hz, 1H), 7.56 (d, J = 2.2
    Hz, 1H), 7.40 (s, 1H), 7.17 (d, J = 8.8 Hz, 2H), 6.96 (d, J = 8.8 Hz, 2H), 5.14
    (s, 2H), 4.39 (t, J = 4.8 Hz, 2H), 4.28 (s, 2H), 3.94 (t, J = 4.8 Hz, 2H), 1.62 (s,
    6H). LCMS (220 nm): 99.7%. Exact Mass: 459.1; found: 460.1/462.1.
    A193 1H NMR (400 MHz, CDCl3) δ = 7.46 (d, J = 2.0 Hz, 1H), 7.31 (d, J = 2.4
    Hz, 1H), 7.13 (d, J = 8.4 Hz, 2H), 6.95-6.89 (m, 3H), 6.32 (d, J = 4.0 Hz,
    1H), 5.00 (s, 2H), 4.43 (t, J = 6.4 Hz, 2H), 3.94 (s, 3H), 3.88 (t, J = 6.4 Hz,
    2H), 3.14 (s, 3H), 1.66 (s, 6H).
    LCMS (220 nm): 99.3%. Exact Mass: 520.10; found 521.1, 523.1.
    A194 1H NMR (400MHz, CDCl3) δ = 7.73 (t, J = 7.6 Hz, 1H), 7.45 (d, J = 2.4 Hz,
    1H), 7.32 (d, J = 2.4 Hz, 1H), 7.24 (d, J = 7.6 Hz, 1H), 7.14 -7.10 (m, 3H),
    6.92 (d, J = 8.8 Hz, 2H), 5.09 (s, 2H), 4.42 (t, J = 6.0 Hz, 2H), 3.88 (t, J =
    6.8 Hz, 2H), 3.22 (s, 3H), 1.65 (s, 6H). LCMS (220 nm): 94.7%. Exact
    Mass: 533.1; found 534.1/536.1.
    A195 1H NMR (400MHz, CDCl3) δ = 8.17 (d, J = 5.6 Hz, 1H), 7.44 (d, J = 2.4 Hz,
    1H), 7.37 (s, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.12 (d, J = 8.8 Hz, 2H), 6.98 (d,
    J = 5.6 Hz, 1H), 6.89 (d, J = 8.8 Hz, 2H), 5.08 (s, 2H), 4.43 (t, J = 6.4 Hz, 2H),
    3.88 (t, J = 6.4 Hz, 2H), 3.12 (s, 3H), 1.65 (s, 6H) LCMS: (220 nm): 97.5%.
    Exact Mass: 533.1; found 534.1/536.1
    A196 1H NMR (400MHz, CDCl3) δ = 8.48 (br s, 1H), 7.44 (d, J = 2.8 Hz, 1H),
    7.31 (d, J = 2.4 Hz, 1H), 7.23 (s, 1H), 7.12 (d, J = 8.8 Hz, 2H), 7.09 (br s,
    1H), 6.93 (d, J = 8.8 Hz, 2H), 6.79 (br s, 1H), 5.17 (s, 2H), 4.42 (t, J = 6.0
    Hz, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.12 (s, 3H), 1.65 (s, 6H). LCMS (220
    nm): 97% Exact Mass: 533; found 534.1/536.1.
    A197 1H NMR (400MHz, CDCl3) δ = 8.51 (br d, J = 4.6 Hz, 2H), 7.45 (s, 1H), 7.32
    (s, 1H), 7.13 (br d, J = 8.6 Hz, 2H), 6.94 (br d, J = 8.2 Hz, 2H), 5.21 (s, 2H),
    4.43 (t, J = 6.2 Hz, 2H), 3.88 (t, J = 6.2 Hz, 2H), 3.33 (s, 3H), 1.65 (s, 6H).
    LCMS: (220 nm): 95.8 Exact Mass: 534.1; found 535.1/537.1
    A198 1H NMR (400MHz, CDCl3) δ = 8.67 (d, J = 5.0 Hz, 1H), 7.45 (d, J = 2.4 Hz,
    1H), 7.34 (d, J = 5.2 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.12 (d, J = 8.8 Hz, 2H),
    6.90 (d, J = 8.8 Hz, 2H), 5.13 (s, 2H), 4.43 (t, J = 6.2 Hz, 2H), 4.30 -4.26 (m,
    2H), 3.88 (t, J = 6.2 Hz, 2H), 3.36 -3.31 (m, 2H), 2.34 (quin, J = 6.2 Hz, 2H),
    1.85 (td, J = 5.8, 11.7 Hz, 2H), 1.65 (s, 6H) LCMS: (220 nm): 90.5%. Exact
    Mass: 574.1; found 575.2/9,577.2.
    A199 1H NMR (400MHz, CDCl3) δ = 8.59 (d, J = 5.0 Hz, 1H), 7.44 (s, 1H), 7.33
    (s, 1H), 7.21 (d, J = 5.0 Hz, 1H), 7.11 (br d, J = 8.4 Hz, 2H), 6.89 (br d, J = 8.4
    Hz, 2H), 5.11 (s, 2H), 4.43 (t, J = 6.4 Hz, 2H), 4.07 (t, J = 6.4 Hz, 2H), 3.88 (t,
    J = 6.4 Hz, 2H), 3.48 (t, J = 7.2 Hz, 2H), 2.52 (br t, J = 6.8 Hz, 2H), 1.65 (s, 6H)
    LCMS: (220 nm): 95.2%. Exact Mass: 560.1; found 561.1/563.1
    A200 1H NMR (400 MHz, CDCl3) δ = 8.88 (d, J = 5.2 Hz, 1H), 7.82 (d, J = 5.2
    Hz, 1H), 7.45 (d, J = 2.4 Hz, 1H), 7.30 (d, J = 2.4 Hz, 1H), 7.14 (d, J = 6.8
    Hz, 2H), 6.90 (d, J = 8.8 Hz, 2H), 5.21 (s, 2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88
    (t, J = 6.4 Hz, 2H), 1.66 (s, 6H). LCMS (220 nm): 91.1%. Exact Mass:
    466.1; found 467.0, 469.0.
    A201 1H NMR (400MHz, CDCl3) δ = 8.80 (brs, 1H), 8.64 (d, J = 5.2 Hz, 1H), 7.45
    (d, J = 2.4 Hz, 1H),7.32 (d, J = 2.4 Hz, 1H), 7.29 (d, J = 5.2 Hz, 1H), 5.11 (s,
    2H), 4.43 (t, J = 6.4 Hz, 2H), 3.88 (t, J = 5.6 Hz, 2H), 3.47 (s, 3H), 1.65 (s, 6H)
    LCMS: (220 nm): 98.53%. Exact Mass: 538.1; found 539.2/541.1
    A202 1H NMR (400MHz, CDCl3) δ = 7.66 (br s, 1H), 7.44 (d, J = 2.4 Hz, 1H), 7.31
    (d, J = 2.4 Hz, 1H), 7.10 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 8.8 Hz, 2H), 5.08 (s,
    2H), 4.43 (t, J = 6.0 Hz, 2H), 3.88 (t, J = 6.4 Hz, 2H), 3.45 (s, 3H), 3.02 -2.93
    (m, 4H), 2.16 -2.12 (m, 2H), 1.64 (s, 6H). LCMS (220 nm): 92.0%. Exact
    Mass: 574.1; found 575.1/577.1
    A203 1H NMR (400MHz, CDCl3) δ = 8.56 (s, 1H), 8.48 (br s, 1H), 7.45 (d, J = 2.4
    Hz, 1H), 7.34 (d, J = 1.6 Hz, 1H), 7.16 -7.11 (m, 2H), 7.10 -7.05 (m, 2H),
    5.70 -5.59 (m, 1H), 4.43 (t, J = 6.4 Hz, 2H), 3.89 (t, J = 6.4 Hz, 2H), 3.31 (s,
    3H), 3.19 -3.07 (m, 1H), 2.96 -2.85 (m, 1H), 2.72 -2.59 (m, 1H), 2.41-2.27
    (m, 1H), 1.66 (s, 6H) LCMS: (220 nm): 97.7% Exact Mass: 560.1;
    found 561.1./563.1
    A204 1H NMR (400MHz, CDCl3) δ = 10.14 (br s, 1H), 8.74 (s, 2H), 7.45 (d,
    J = 2.2 Hz, 1H), 7.33 (d, J = 2.2 Hz, 1H), 7.14 (d, J = 8.8 Hz, 2H), 6.91 (d,
    J = 8.8 Hz, 2H), 5.02 (s, 2H), 4.43 (t, J = 6.4 Hz, 2H), 3.88 (t, J = 6.4 Hz, 2H),
    3.49 (s, 3H), 1.66 (s, 6H) LCMS: (220 nm): 98.6% Exact Mass: 534.1; found
    535.0/537.0
    A205 1H NMR (400MHz, CDCl3) δ = 8.61 (d, J = 5.2 Hz, 1H), 7.27-7.29 (m, 1H),
    7.17-7.19 (m, 1H),7.13- 7.14 (m, 1H), 7.12 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.8
    Hz, 1H), 5.10 (s, 2H), 4.48 (t, J = 6.0 Hz, 2H), 3.83 (t, J = 5.6 Hz, 2H), 3.48 (s,
    3H), 1.64 (s, 6H) LCMS: (220 nm): 99.74%. Exact Mass: 518.1; found
    519.1/521.1
    A206 1H NMR (400MHz, CDCl3) δ = 8.62 (d, J = 5.2 Hz, 1H),8.60 (brs, 1H), 7.29
    (d, J = 5.2 Hz, 1H), 7.15 (d, J = 8.8 Hz, 1H),7.02 (d, J = 2.0 Hz, 1H), 6.89 (d,
    J = 9.2 Hz, 1H), 6.86 -6.83 (dd, J = 12.4 Hz, 2.0 Hz, 1H), 5.10 (s, 2H), 4.31 (t,
    J = 6.0 Hz, 2H), 3.81 (t, J = 6.0 Hz, 2H), 3.47 (s, 3H), 1.63 (s, 6H) LCMS:
    (220 nm): 99.51%. Exact Mass: 527.1; found 528.0/529.9
    A207 1H NMR (400MHz, CDCl3) δ = 7.63 (d, J = 0.8 Hz, 1H), 7.46 (d, J = 2.4 Hz,
    1H), 7.30 (d, J = 2.4 Hz, 1H), 7.16 -7.07 (m, 3H), 6.89 (d, J = 8.8 Hz, 2H),
    5.01 -4.96 (m, 1H), 4.96 (br s, 1H), 4.95 (s, 2H), 4.43 (t, J = 6.4 Hz, 2H),
    3.88 (t, J = 6.4 Hz, 2H), 1.65 (s, 6H) LCMS: (220 nm): 99.2% Exact Mass:
    508.1; found 509.0/ 511.0
    A208 1H NMR (400MHz, CDCl3) δ = 7.71 (s, 1H), 7.45 (d, J = 2.4 Hz, 1H), 7.32
    (d, J = 2.2 Hz, 1H), 7.26 (br s, 1H), 7.13 (d, J = 8.8 Hz, 2H), 6.89 (d, J = 8.8 Hz,
    2H), 4.96 (s, 2H), 4.43 (t, J = 6.2 Hz, 2H), 3.88 (t, J = 6.2 Hz, 2H), 3.18 (s,
    3H), 1.66 (s, 6H) LCMS: (220 nm): 99.7% Exact Mass: 507.1; found
    508.1/510.1
    A209 1H NMR (400 MHz, CDCl3) δ = 8.53 (d, J = 4.8 Hz, 1H), 7.44 (d, J = 2.4
    Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 7.21 (d, J = 5.2 Hz, 1H), 7.11 (d, J = 8.8
    Hz, 2H), 6.89 (d, J = 8.8 Hz, 2H), 5.09 (s, 2H), 4.42 (t, J = 6.0 Hz, 2H), 3.88
    (t, J = 6.0 Hz, 2H), 2.59 (s, 3H), 1.65 (s, 6H).
    A210 1H NMR (400MHz, CDCl3) δ = 7.79 (s, 1H), 7.70 (s, 1H), 7.47 (d, J = 1.8 Hz,
    1H), 7.34 (d, J = 1.8 Hz, 1H), 7.14 (d, J = 8.8 Hz, 2H), 6.91 (d, J = 8.6 Hz, 2H),
    5.07 (s, 2H), 4.44 (t, J = 6.2 Hz, 2H), 3.90 (t, J = 6.2 Hz, 2H), 3.23 (s, 3H),
    1.67 (s, 6H) LCMS: (220 nm): 99.4% Exact Mass: 523.1; found 524.1/526.1
    A211 1H NMR (400MHz, CDCl3) δ = 7.72 (s, 1H), 7.58 (s, 1H), 7.46 (d, J = 2.2 Hz,
    1H), 7.31 (d, J = 2.2 Hz, 1H), 7.12 (d, J = 8.8 Hz, 2H), 6.89 (d, J = 8.8 Hz, 2H),
    5.03 (s, 2H), 4.99 (s, 2H), 4.43 (t, J = 6.2 Hz, 2H), 3.88 (t, J = 6.2 Hz, 2H),
    1.65 (s, 6H). LCMS: (220 nm): 99.4% Exact Mass: 524.0; found 524.9/522.8
  • Biological Assays Example 35: 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 1A).
  • Luciferase Assay:
  • 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.
  • Statistical analyses were performed using GraphPad Prism (Version 6.01 for Windows; La Jolla, Calif., 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 1A shows IC50 ranges of Compounds from Tables A and B from the luciferase assay. FIG. 1 shows dose dependent curve of representative compounds shown in Table 1A. Compound X and EPI-002 have the following structures:
  • Figure US20200123117A1-20200423-C00361
  • The PSA-Luc % inhibition IC50 values of the representative compounds are shown in Table 1B.
  • LBD-Expressing Reporter Assay:
  • To assess if the representative compounds (Compounds A13, A28, A38, A74, A93, and A109) compete with ligand (agonist) for the ligand-binding domain (LBD) of steroid hormone receptors (which would be off-target), the GeneBLAzer assay was employed (Thermo Fisher). This assay uses cells that stably express a chimera fusion protein comprising the LBD of a steroid hormone receptor fused to the DNA-binding domain (DBD) of GAL4 along with beta-lactamase reporter under transcriptional control of an upstream activator sequence (UAS). When agonist binds to the LBD of the GAL4 (DBD)-steroid hormone (LBD) chimera fusion protein, the protein binds to the UAS, resulting in expression of beta-lactamase. LBDs of androgen receptor (AR), progesterone receptor-beta (PRIβ), glucocorticoid receptor (GR), estrogen receptor-α (ERα), and estrogen receptor-β (ERβ) were tested and IC50s calculated for inhibiting maximal activity attained with agonist. See Table 1C.
  • Cell Proliferation Assay:
  • Cell proliferation/viability was measured in LNCaP and PC3 cells with Alamar blue, and proliferation was measured in LN-CaP95 cells with BrdU incorporation. In LNCaP cells, AR specific proliferation is calculated by measuring the difference between control cells treated with or without 0.1 nM R1881. See Table 1D and FIG. 7.
  • AR-V7 Transcriptional Activity Assay:
  • Transcriptional activity of AR-V7 was measured in LNCaP cells transiently cotransfected with an expression vector for AR-V7 and the PSA-luciferase reporter and incubated with the representative compounds in the presence or absence of synthetic androgen, R1881. As a control, LNCaP cells were transfected with pcDNA plasmid (absent of AR-V7 insert) with or without R1881. See FIGS. 2A-2B.
  • The results from this Example supports that the compounds of the present disclosure are selective to AR NTD.
  • TABLE 1A
    IC50 Ranges of Various Compounds in
    Androgen-Induced PSA-Luciferase Assay
    Compound PSA-Luc
    ID IC50 Range
    A1 N/A
    A2 N/A
    A3 **
    A4 N/A
    A5 **
    A6 N/A
    A7 *
    A8 N/A
    A9 N/A
    A10 N/A
    A11 N/A
    A12 N/A
    A13 **
    A14 N/A
    A15 N/A
    A16 N/A
    A17 **
    A18 *
    A19 N/A
    A20 N/A
    A21 N/A
    A22 *
    A23 *
    A24 **
    A25 *
    A26 N/A
    A27 *
    A28 ***
    A29 ***
    A30 **
    A31 ***
    A32 *
    A33 N/A
    A34 **
    A35 **
    A36 N/A
    A37 N/A
    A38 **
    A39 N/A
    A40 **
    A41 *
    A42 **
    A43 N/A
    A44 N/A
    A45 **
    A46 N/A
    A47 N/A
    A48 N/A
    A49 *
    A50 **
    A51 N/A
    A52 *
    A53 *
    A54 **
    A55 N/A
    A56 **
    A57 ***
    A58 *
    A59 N/A
    A60 N/A
    A61 N/A
    A62 *
    A63 **
    A64 *
    A65 **
    A66 **
    A67 N/A
    A68 ***
    A69 N/A
    A70 ***
    A71 **
    A72 N/A
    A73 **
    A74 **
    A75 **
    A76 **
    A77 **
    A78 **
    A79 ***
    A80 ***
    A81 ***
    A82 N/A
    A83 N/A
    A84 N/A
    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 N/A
    A111 ***
    A112 ***
    A113 *
    A114 **
    A115 ***
    A116 **
    A117 **
    A118 **
    A119 ***
    A120 *
    A121 **
    A122 ***
    A123 ***
    A124 *
    A125 ***
    A126 **
    A127 N/A
    A128 N/A
    A129 N/A
    A130 **
    A131 **
    A132 **
    A133 ***
    A134 **
    A135 ***
    A136 **
    A137 ***
    A138 N/A
    A139 N/A
    A140 ***
    A141 ***
    A142 ***
    A143 *
    A144 N/A
    A145 **
    A146 N/A
    A147 *
    A148 **
    A149 ***
    A150 N/A
    A151 **
    A152 N/A
    A153 **
    A154 N/A
    A155 ***
    A156 ***
    A157 N/A
    A158 N/A
    A159 N/A
    A160 N/A
    A161 N/A
    A162 ***
    A163 N/A
    A164 **
    A165 N/A
    A166 N/A
    A167 N/A
    A168 ***
    A169 ***
    A170 **
    A171 **
    A172 ***
    A173 *
    A174 **
    A175 N/A
    A176 N/A
    A177 N/A
    A178 N/A
    A179 N/A
    A180 N/A
    A181 N/A
    A182 N/A
    A183 N/A
    A184 *
    A185 ***
    A186 N/A
    A187 N/A
    A188 N/A
    A189 **
    A190 **
    A191 **
    A192 ***
    A193 **
    A194 **
    A195 ***
    A196 **
    A197 **
    A198 ***
    A199 ***
    A200 **
    A201 **
    A202 **
    A203 **
    A204 ***
    A205 **
    A206 **
    A207 ***
    A208 ***
    A209 N/A
    A210 N/A
    A211 N/A
    B1 **
    B2 ***
    B3 **
    B4 N/A
    B5 **
    B6 *
    B7 *
    B8 **
    B9 ***
    B10 *
    Note:
    ***represent IC50 < 500 nM,
    **represents IC50 in the range of 500-2000 nM,
    *represents IC50 in the range of ≥2000 nM
  • TABLE 1B
    IC50 of Representative Compounds in
    Androgen-Induced PSA-Luciferase Assay
    PSA-Luc
    Compound ID IC50 (nM) n
    X 1054 3
    A13 592 8
    A28 400 5
    A38 631 6
    A74 658 6
    A93 205 4
    A109 535 2
    EPI-002 9580 2
    Enzalutamide 189 8
    Bicalutamide 306 2
  • TABLE 1C
    IC50 of Representative Compounds
    in LBD-Expressing Reporter Cell Lines
    Steroid Receptor LBD IC50 (μM)
    Compound ID AR ERα ERβ PR GR
    X 9.83 5.82 7.37 4.96 >10
    A13 >10 >10 >10 0.97 >10
    A28 >10 >10 >10 0.93 >10
    A38 >10 >10 >10 5.07 >10
    A74 >10 >10 >10 5.41 9.51
    A93 >10 >10 >10 5.04 >10
    A109 >10 >10 >10 5.61 >10
    EPI-002 >10 >10 >10 >10 >10
    Enzalutamide 0.54 >10 >10 4.49 >10
  • TABLE 1D
    IC50 of Represenatative Compounds on Cell Proliferation/Viability
    Cellular Proliferation/Viability IC50 (μM)
    Compound ID LNCaP PC-3 LNCaP95
    X 3.00 >10 4.00
    A13 0.85 >10 4.00
    A28 0.68 >10 6.43
    A74 1.16 >10 2.65
    A109 0.44 >10 3.78
    EPI-002 9.00 >10 ~20
    Enzalutamide 0.35 >10 >10
  • Example 36: 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 L) 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 L 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 M, [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 (t½) values and subsequently, the intrinsic clearance (Clint).

  • Cl int=(0.693)(1/t½(min))(g of liver/kg of body weight)(mL incubation/mg of microsomal protein)(45 mg of microsomal protein/g of liver weight)
  • Hepatocyte Stability Assay:
  • The termination of in vitro intrinsic clearance (Clint) for drug candidates in the early discovery stage is common practice in the pharmaceutical industry. The Clint values of drug candidates can help to confirm whether metabolism is the main clearance pathway when it is compared with the total body clearance in vivo. Hepatocytes are among the routinely used in vitro systems for measuring clearance. Hepatocytes have been reported to retain most of the phase I and phase II metabolic activities, and therefore, it is a useful tool to estimate the intrinsic clearance for potential drug candidates.
  • Metabolic stability of testing compound were be evaluated using human and mouse hepatocytes and can also be use rat, mouse, or other animal hepatocytes to predict intrinsic clearance. Cryopreserved hepatocytes were removed from the liquid nitrogen tank and thawed in a 37° C. water bath. As soon as the cells pulled away from the vial wall, they were decanted into 48 ml of warm HT medium. Cells were centrifuged for four minutes at 420 rpm (50 g). After removing the supernatant, pellet was re-suspended in warm DMEM medium. Cell density was counted by a hemacytometer.
  • The assay was carried out in 96-well microtiter plates. Compounds were incubated for 0, 60, 120, and 180 minutes at 37° C. with hepatocytes. Reaction mixtures (50 μL) contained a final concentration of 1 μM test compound, 0.5 million cells/mL hepatocytes in the DMEM medium. At each of the time points (for example, 0, 1, 2, and 3 hours), 200 μL of quench solution (100% acetonitrile with 0.1% formic acid) with internal standard was transferred to each well. Midazolam was included as a positive control to verify assay performance. Plates were sealed 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 (customizable): [Compound]=1 M, Positive control: midazolam and/or naloxone, [Hepatocyte]=0.5 million cells/Ml, time=0, 60, 120, and 180 min, t=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 was calculated as the disappearance of the test compound, compared to the 0-min control reaction incubations. Initial rates were calculated for the compound concentration and used to determine t1/2 values and subsequently, the intrinsic clearance, Clint.

  • Cl int=(0.693)(1/t½(min))(mL incubation/million cells)
  • Metabolic stability assay results of representative compounds from Tables A and B are listed in Table 2A, below. Metabolic stability assay results of representative compounds from Tables A and B for human hepatocytes are demonstrated in Table 2B. Liver microsome and hepatocyte half life of representative compounds are shown in Tables 2C and 2D.
  • TABLE 2A
    Stability Results of Representative Compounds from Tables A and B
    MH +/−
    MLM +/− NADPH: HLM +/− NADPH: NADPH: Clint
    Compound Clint (μL/min/mg Clint (μL/min/mg (μL/min/
    ID protein)1 protein)1 million cells)2
    A3 *** *** NA
    A5 NA *** NA
    B1 * ** NA
    A7 NA * NA
    A13 *** *** **
    A17 NA *** NA
    A18 NA * NA
    A21 NA *** NA
    A22 NA *** NA
    A23 NA ** NA
    A24 NA *** NA
    A25 NA *** NA
    A27 * *** NA
    B2 * * NA
    B3 *** *** NA
    B5 *** *** **
    A28 ** *** **
    A29 ** ** NA
    A30 NA ** **
    A31 NA ** NA
    A32 * ** NA
    A34 ** *** NA
    A35 ** *** NA
    A38 *** *** ***
    A40 NA ** NA
    A41 NA * NA
    A42 *** *** NA
    A45 *** *** NA
    A49 *** ** NA
    A50 NA ** NA
    A52 NA *** NA
    A53 NA *** NA
    A54 NA *** NA
    A56 NA *** NA
    A57 * ** NA
    A58 NA ** NA
    B8 NA * NA
    A62 NA *** NA
    A63 NA ** NA
    B9 ** ** NA
    A64 NA * NA
    A65 ** ** NA
    A66 *** *** NA
    A68 *** *** NA
    A70 * ** **
    A71 *** ** **
    A73 *** *** NA
    A74 *** *** NA
    A75 NA *** NA
    B6 NA ** NA
    A76 NA *** NA
    B7 *** *** NA
    A77 * ** NA
    B10 ** *** NA
    A78 *** *** NA
    A79 * *** *
    A80 ** *** *
    A81 * ** *
    A85 * *** *
    A86 ** ** *
    A87 ** *** *
    A88 * * *
    A89 * ** *
    A90 * *** *
    A91 *** *** **
    A92 ** ** *
    A93 *** *** ***
    A94 ** ** **
    A95 * ** *
    A96 * * *
    A97 ** ** *
    A98 *** *** ***
    A99 ** *** **
    A100 * * *
    A101 ** * **
    A102 *** ** ***
    A103 ** ** ***
    A104 * ** *
    A105 * ** *
    A106 NA ** NA
    A107 NA ** NA
    A108 * *** *
    A109 *** *** ***
    A110 NA NA NA
    A111 ** ** **
    A112 ** *** **
    A113 ** ** *
    A114 ** *** *
    A115 *** *** *
    A116 ** ** **
    A117 ** ** *
    A118 * * **
    A119 * * **
    A120 *** * **
    A121 ** ** **
    A122 ** *** **
    A123 * *** **
    A124 * *** *
    A125 * *** *
    A126 *** *** ***
    A127 ** ** **
    A128 NA NA NA
    A129 NA NA NA
    A130 *** ** ***
    A131 *** *** ***
    A132 *** *** ***
    A133 * * *
    A134 *** ** ***
    A135 ** ** *
    A136 ** ** **
    A137 ** *** ***
    A138 NA NA NA
    A139 NA NA NA
    A140 * * *
    A141 * *** *
    A142 * ** *
    A143 *** *** ***
    A144 ** ** *
    A145 * * *
    A146 *** ** *
    A147 * ** *
    A148 *** * ***
    A149 *** *** *
    A150 NA NA NA
    A151 NA NA NA
    A152 NA NA NA
    A153 ** *** **
    A154 NA NA NA
    A155 * ** **
    A156 ** ** *
    A157 NA NA NA
    A158 NA NA NA
    A159 NA NA NA
    A160 NA NA NA
    A161 *** *** NA
    A162 * ** *
    A163 NA NA NA
    A164 *** *** ***
    A165 NA NA NA
    A166 NA NA NA
    A167 NA NA NA
    A168 *** *** **
    A169 *** *** ***
    A170 *** *** ***
    A171 *** *** **
    A172 *** *** ***
    A173 *** *** NA
    A174 ** *** NA
    A175 NA NA NA
    A176 NA NA NA
    A177 NA NA NA
    A178 NA NA NA
    A179 NA NA NA
    A180 NA NA NA
    A181 NA NA NA
    A182 NA NA NA
    A183 NA NA NA
    A184 *** *** ***
    A185 ** *** **
    A186 NA NA NA
    A187 NA NA NA
    A188 NA NA NA
    A189 *** *** NA
    A190 * *** *
    A191 NA NA NA
    A192 ** ** *
    A193 * * **
    A194 *** *** ***
    A195 *** *** **
    A196 *** ** ***
    A197 *** *** ***
    A198 ** *** **
    A199 * *** **
    A200 *** *** **
    A201 *** *** ***
    A202 * *** *
    A203 NA NA NA
    A204 *** *** ***
    A205 NA NA NA
    A206 *** *** ***
    A207 *** *** ***
    A208 *** *** ***
    A209 NA NA NA
    A210 NA NA NA
    A211 NA NA NA
    1For MLM and HLM: *** represent Cl < 12 μL/min/mg protein, ** represents Cl in the range of 12-48 μL/min/mg protein, * represents Cl in the range of ≥48 μL/min/mg protein.
    2For MH: *** represent Cl <4 μL/min/million cells, ** represents Cl in the range of 4-18 μL/min/million cells, * represents Cl in the range of ≥18 μL/min/million cells.
  • TABLE 2B
    Human Hepatocyce Stability Results of Representative
    Compounds from Tables A and B
    Number of Compounds
    Cl < 4 μL/minimillion cells 14 compounds
    Cl in the range of 4-18 μL/minimillion 31 compounds
    cells
    Cl in the range of ≥18 μL/minimillion 36 compounds
    cells
  • TABLE 2C
    Liver Microsome Half-Life
    Liver microsome T½ (min)
    Compound ID Human Mouse Rat Dog Monkey
    A13 >120 >120 64 >120 >120
    A28 >120  74 23 >120 73
    A38 >120 >120 12 >120 >120
    A74 >120 >120 >120 >120 >120
    A93 >120 >120 83 >120 101
    A109 >120 >120 >120 >120 >120
    X >120 NA NA NA NA
    Enzalutamide >120 >120 >120 >120 >120
  • TABLE 2D
    Hepatocytes Half-Life
    Hepatocytes T½ (min)
    Compound ID Human Mouse Rat
    A13 290 203 75
    A28 >360 86 31
    A38 >360 >360 56
    A74 >360 NA >360
    A93 >360 >360 94
    A109 >360 >360 >360
    X >360 33 NA
    Enzalutamide >360 NA NA
  • Example 37: In Vivo Pharmacokinetic Properties
  • The purpose of this study is to determine the pharmacokinetics of the tested compounds in plasma, following oral gavage administration (PO) to male CD-1 mice.
  • Animal Husbandry: Animals were group housed during acclimation and the study. The animal room environment were controlled (target conditions: temperature 18 to 26° C., relative humidity 30 to 70%, 12 hours artificial light and 12 hours dark). Temperature and relative humidity were monitored daily. Animals were fasted at least 12 hours prior to the administration. All animals had access to Certified Rodent Diet ad libitum 4 hours post dosing.
  • Dose Formulation: prepared 85-100 uL of a stock solution of representative compounds of the disclosure in DMSO at 50 mM; 1.5% v/v of Tween 80 were added and mixed gently; 95.5% v/v saline was added gently to the organic phase. The solution was mixed slowly by reverse pipetting to get a clear solution.
  • Dose Administration: the dose formulations were administered via oral gavage per facility SOPs. The dose volume were determined by the animals' body weight collected on the morning of dosing day
  • Sample Collection: blood collection (about 0.05 mL per time point) was performed from saphenous vein of each animal into polypropylene tubes at each timepoints (0, 0.25, 0.5, 1, 2, 4, 8, 12, and 24 hr). All blood samples were transferred into pre-chilled tubes containing 2 μL of K2-EDTA (0.5M) and placed on wet ice until centrifugation. Each collected blood sample was in the wet-ice before centrifugation. Each collected blood was under centrifugation for 15 minutes at 4° C. and 3000 g for plasma collection. Plasma samples were stored in polypropylene tubes, quickly frozen over dry ice and kept at −70±10° C. until LC/MS/MS analysis.
  • Bioanalytical analysis: A LC-MS/MS method for the quantitative determination of tested compound in biological matrix were developed under non-GLP compliance. A calibration curve with at least 7 non-zero calibration standards were applied for the method including LLOQ.
  • Plasma concentration versus time data were analyzed by non-compartmental approaches using the Phoenix WinNonlin 6.3 software program. C1, Vdss, C0, Cmax, Tmax, T½, AUC(0-t), AUC(0-inf), MRT(0-t), MRT(0-inf), % F (oral availability) and graphs of plasma concentration versus time profile were reported. FIG. 3 shows the time-concentration curve of representative compounds in mice as measured in plasma.
  • Tables 3A shows PK parameters determined from single PO dose of representative compounds in mice (5 mg/kg).
  • TABLE 3A
    Summary of PO PK Parameters After Single
    Dose (5 mg/kg) in Male CD-1 Mice
    AUC/D
    Compound tmax Cmax AUClast (hr*kg*ng/ F
    ID (hr) (hr) (ng/mL) (hr*ng/mL) mL/mg) (%)
    A13 8.3 1.0 1143.3 8309.3 1661.9 85.8
    A28 2.8 0.5 2013.3 8425.4 1685.1 50.3
    A29 2.5 0.5 2010.0 5186.6 1037.3 60.6
    A35 4.0 0.5 818.7 2415.1 483.0 49.6
    A38 4.9 3.3 1270.0 12805.1 2561.0 86.4
    A66 12.0 0.8 1613.3 11927.2 2385.4 71.4
    A74 12.5 2.7 6306.7 72711.4 14542.3 71.9
    A93 17.1 0.8 2973.3 32878.6 6575.7 62.8
    A109 8.1 2.2 2673.3 30714.8 6143.0 33.6
    A122 1.1 0.5 4080.0 6800.0 1360.0 92.4
    A126 4.7 1.3 3113.0 24547.0 4909.4 86.0
    A131 5.0 1.3 7053.0 76047.0 15209.4 81.0
    A136 10.2 1.5 3427.0 42038.0 8407.6 14.0
    A170 3.0 3.0 8503.0 80038.0 16007.6 150.0
  • Example 38: In Vivo Activity of Representative Compounds in LNCaP Xenografts Model
  • Tumor growth was measured in male NCG mice bearing LNCaP tumors. Castration was performed when tumors reached ˜100 mm3 and dosing (60 mg/kg PO qd) started 1 week after castration. See FIG. 4. Body weight of the mice were captured biweekly in the animals which showed no drug related toxicity. See FIG. 5. Individual tumor volume change from baseline measured at the end of the experiment. See FIG. 6. Data demonstrated in FIGS. 4-6 shows that the representative compounds showed activity and induced partial regressions of tumor growth. The Cmin at 5 mg/kg PO and extrapolated Cmin in efficacy of the representative compounds are shown in Table 4.
  • TABLE 4
    Cmin at 5 mg/kg PO and Cmin at 5 mg/kg PO
    Compound Cmin at 5 mg/kg PO Cmin at 5 mg/kg PO
    ID (μM) (μM)
    A13 0.23 2.78
    A74 2.42 29.1
    A109 0.68 8.18
    Enzalutamide 3.80 22.8
  • Numbered Embodiments
  • In some embodiments, the present disclosure relates to the following embodiments.
  • Embodiment 1
  • A compound having a structure of formula (I):
  • Figure US20200123117A1-20200423-C00362
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-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—, —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, —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)-CON14R15, —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, 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 R8b 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 or 2;
      • 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.
    Embodiment 2
  • The compound of Embodiment 1, wherein A and B are each independently 5- or 6-membered aryl or heteroaryl.
  • Embodiment 3
  • The compound of Embodiment 1 or 2, wherein A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene.
  • Embodiment 4
  • The compound of any one of Embodiments 1-3, wherein A has a meta or para connectivity with X and Y.
  • Embodiment 5
  • The compound of any one of Embodiments 1-4, wherein B has a meta or para connectivity with X and Z.
  • Embodiment 6
  • The compound of any one of Embodiments 1-5, wherein C is 5- to 10-membered heteroaryl or aryl.
  • Embodiment 7
  • The compound of any one of Embodiments 1-6, wherein C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 8
  • The compound of any one of Embodiments 1-7, wherein C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, or pyrimidyl.
  • Embodiment 9
  • The compound of any one of Embodiments 1-7, wherein C, which is optionally substituted with R3, is selected from
  • Figure US20200123117A1-20200423-C00363
    Figure US20200123117A1-20200423-C00364
    Figure US20200123117A1-20200423-C00365
  • wherein R3a is C1-C3 alkyl.
  • Embodiment 10
  • The compound of any one of Embodiments 1-6, wherein C is phenyl or naphthyl.
  • Embodiment 11
  • The compound of any one of Embodiments 1-5, wherein C is 5- to 10-membered heterocyclyl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 12
  • The compound of Embodiment 11, wherein
      • C is
  • Figure US20200123117A1-20200423-C00366
      • D is —O—, —NH— or —NR3—; and
      • U is each independently O, S, or NR16.
    Embodiment 13
  • The compound of Embodiment 11, wherein C is imidazolidine, imidazolidine-dione, or dihydrooxazole.
  • Embodiment 14
  • The compound of any one of Embodiments 1-13, wherein Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)-.
  • Embodiment 15
  • The compound of any one of Embodiments 1-14, wherein W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—, wherein R7 is H or C1-C3 alkyl.
  • Embodiment 16
  • The compound of any one of Embodiments 1-15, wherein:
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—; and
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—.
    Embodiment 17
  • The compound of any one of Embodiments 1-16, wherein:
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is —CH2—, —CH2CH2—, —CH2CH2CH2—; and
      • L is halogen, —NH2, or —CF3.
    Embodiment 18
  • The compound of any one of Embodiments 1-17, wherein X is a bond or —(CR5R6)t—.
  • Embodiment 19
  • The compound of any one of Embodiments 1-18, wherein X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—.
  • Embodiment 20
  • The compound of any one of Embodiments 1-19, wherein R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, C1-C3 alkyl, C1-C3 alkoxy, —(C1-C3 alkyl)-(C1-C3 alkoxy), —(C1-C3 alkyl)-OH, —NR13R14, —(C1-C3 alkyl)-NR3R4, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2R16, —(C1-C3 alkyl)-SO2R16, 3- to 7-membered carbocyclyl, 3- to 7-membered heterocyclyl, phenyl, or 5- to 6-membered heteroaryl.
  • Embodiment 21
  • The compound of any one of Embodiments 1-20, wherein R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, C1-C3 alkyl, C1-C3 alkoxy, —(C1-C3 alkyl)-(C1-C3 alkoxy), —(C1-C3 alkyl)-OH, —NR3R4, —(C1-C3 alkyl)-NR3R4, —NR14SO2R16, —(C1-C3 alkyl)NR14SO2R16, —NR14COR16, —(C1-C3 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2R16, or —(C1-C3 alkyl)-SO2R16.
  • Embodiment 22
  • The compound of any one of Embodiments 1-21, wherein R1 and R2 are each independently halogen, —CN, —CF3, —OH, methyl, methoxy, or —CONH2.
  • Embodiment 23
  • The compound of any one of Embodiments 1-22, wherein R1 and R2 are each independently C1, —CN, —CF3, —OH, methyl, methoxy, or —CONH2.
  • Embodiment 24
  • The compound of any one of Embodiments 1-23, wherein n1 and n2 are each independently 0, 1, or 2.
  • Embodiment 25
  • The compound of any one of Embodiments 1-24, wherein the sum of n1 and n2 is 0, 1, 2, 3, or 4.
  • Embodiment 26
  • The compound of any one of Embodiments 1-25, wherein R3 is selected from hydrogen, halogen, oxo, ═S, or ═NR16, —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)-NR3R4, —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).
  • Embodiment 27
  • The compound of any one of Embodiments 1-26, wherein R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl).
  • Embodiment 28
  • The compound of any one of Embodiments 1-27, wherein R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, methyl, —SCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3.
  • Embodiment 29
  • The compound of any one of Embodiments 1-28, wherein at least one of R3 is —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3.
  • Embodiment 30
  • The compound of any one of Embodiments 1-29, wherein at least one of R3 is oxo, ═S, ═NR16.
  • Embodiment 31
  • The compound of any one of Embodiments 1-30, wherein 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.
  • Embodiment 32
  • The compound of any one of Embodiments 1-30, wherein R8a and R9a are not —OH.
  • Embodiment 33
  • The compound of Embodiment 1, wherein the compound has the structure of formula (II):
  • Figure US20200123117A1-20200423-C00367
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —NHCO—, —N(C1-C3 alkyl)CO—, or —CONH—, or —CON(C1-C3 alkyl)-;
      • V is a bond, —(CR8aR9a)m—, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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;
      • 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 R8b taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R11, R12, 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 a 3- to 6-membered heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or 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.
    Embodiment 34
  • The compound of Embodiment 1, wherein the compound has the structure of formula (III):
  • Figure US20200123117A1-20200423-C00368
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —(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—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 R8b 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or 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.
    Embodiment 35
  • The compound of Embodiment 33 or 34, wherein 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.
  • Embodiment 36
  • The compound of Embodiment 33 or 34, wherein R8a and R9a are not —OH.
  • Embodiment 37
  • The compound of Embodiments 1, wherein the compound has the structure of formula (IV):
  • Figure US20200123117A1-20200423-C00369
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y and Z are each independently a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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.
    Embodiment 38
  • The compound of any one of Embodiments 33-37, wherein C is 5- to 10-membered heteroaryl or aryl.
  • Embodiment 39
  • The compound of any one of Embodiments 33-38, wherein C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 40
  • The compound of any one of Embodiments 33-39, wherein C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, or pyrimidyl.
  • Embodiment 41
  • The compound of any one of Embodiments 33-39, wherein C, which is optionally substituted with R3, is selected from
  • Figure US20200123117A1-20200423-C00370
    Figure US20200123117A1-20200423-C00371
  • wherein R3a is C1-C3 alkyl.
  • Embodiment 42
  • The compound of any one of Embodiments 33-38, wherein C is phenyl or naphthyl.
  • Embodiment 43
  • The compound of any one of Embodiments 33-37, wherein C is 5- to 10-membered heterocyclyl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 44
  • The compound of Embodiment 43, wherein
      • C is
  • Figure US20200123117A1-20200423-C00372
      • D is —O—, —NH— or —NR3—; and
      • U is each independently O, S, or NR16.
    Embodiment 45
  • The compound of Embodiment 43, wherein C is imidazolidine, imidazolidine-dione, or dihydrooxazole.
  • Embodiment 46
  • The compound of any one of Embodiments 33-45, wherein A has a meta or para connectivity with X and Y.
  • Embodiment 47
  • The compound of any one of Embodiments 33-46, wherein B has a meta or para connectivity with X and Z.
  • Embodiment 48
  • The compound of any one of Embodiments 33-47, wherein n1 and n2 are each independently 0, 1, or 2.
  • Embodiment 49
  • The compound of Embodiments 1, wherein the compound has the structure of formula (V):
  • Figure US20200123117A1-20200423-C00373
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 5- to 10-membered heteroaryl or aryl;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, halogen, oxo, ═S, —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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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.
    Embodiment 50
  • The compound of Embodiment 49, wherein C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 51
  • The compound of Embodiment 49 or 50, wherein C is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, or pyrimidyl.
  • Embodiment 52
  • The compound of Embodiment 49, wherein C, which is optionally substituted with R3, is selected from
  • Figure US20200123117A1-20200423-C00374
    Figure US20200123117A1-20200423-C00375
  • wherein R3a is C1-C3 alkyl.
  • Embodiment 53
  • The compound of any one of Embodiments 49-52, wherein A has a meta or para connectivity with X and Y.
  • Embodiment 54
  • The compound of any one of Embodiments 49-53, wherein B has a meta or para connectivity with X and Z.
  • Embodiment 55
  • The compound of any one of Embodiments 49-54, wherein A and B are each phenyl.
  • Embodiment 56
  • The compound of any one of Embodiments 49-55, wherein —Z—V-L is —Z—CH2CH2C1, —Z—CH2CH2CH2C1, —Z—CH2CH2NH2, or —Z—CH2CH2CH2NH2, wherein Z is a bond, —O—, —NH—, or —N(COCH3)-.
  • Embodiment 57
  • The compound of any one of Embodiments 49-56, wherein —Y—W— is a bond, —OCH2—, —OCH2CH2—, —OCH(CH3)—, —NH—, —NHCH2—, —NHC(═O)—, or —C(═O)NH—.
  • Embodiment 58
  • The compound of any one of Embodiments 49-57, wherein X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—.
  • Embodiment 59
  • The compound of Embodiment 1, wherein the compound has the structure of formula (VI):
  • Figure US20200123117A1-20200423-C00376
  • wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 5- to 10-membered heterocyclyl;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl; and
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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.
    Embodiment 60
  • The compound of Embodiment 57, wherein C is 5- to 7-membered saturated or partially saturated heterocycle comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 61
  • The compound of Embodiment 59 or 60, wherein C is imidazolidine, imidazolidine-dione, or dihydrooxazole.
  • Embodiment 62
  • The compound of any one of Embodiments 59-61, wherein C, which is optionally substituted with R3, is selected from
  • Figure US20200123117A1-20200423-C00377
  • Embodiment 63
  • The compound of any one of Embodiments 59-62, wherein A has a meta or para connectivity with X and Y.
  • Embodiment 64
  • The compound of any one of Embodiments 59-63, wherein B has a meta or para connectivity with X and Z.
  • Embodiment 65
  • The compound of any one of Embodiments 59-64, wherein A and B are each phenyl.
  • Embodiment 66
  • The compound of any one of Embodiments 59-65, wherein -V-L is —CH2CH2C1, —CH2CH2CH2C1, —CH2CH2NH2, or —CH2CH2CH2NH2, and Z is a bond, —O—, —NH—, or —N(COCH3)—.
  • Embodiment 67
  • The compound of any one of Embodiments 59-66, wherein —Y—W— is a bond, —OCH2—, —OCH2CH2—, —OCH(CH3)—, —NH—, —NHCH2—, —NHC(═O)—, or —C(═O)NH—.
  • Embodiment 68
  • The compound of any one of Embodiments 59-67, wherein X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—.
  • Embodiment 69
  • The compound of any one of Embodiments 59-68, R3 is oxo, ═S, ═NR16, C1-C3 alkyl, —SO2(C1-C3 alkyl), or —NHSO2(C1-C3 alkyl).
  • Embodiment 70
  • The compound of Embodiment 1, wherein the compound has the structure of formula (A):
  • Figure US20200123117A1-20200423-C00378
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a phenyl or a 5- to 7-membered monocyclic heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • 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.
    Embodiment 71
  • The compound of Embodiment 70, wherein C is 5- to 7-membered monocyclic heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 72
  • The compound of Embodiment 1, wherein the compound has the structure of formula (B)
  • Figure US20200123117A1-20200423-C00379
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a 5- to 7-membered saturated or partially saturated monocyclic heterocycle comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, ═S, ═NR16, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • R16 is hydrogen or C1-C3 alkyl;
      • 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.
    Embodiment 73
  • The compound of any one of Embodiments 70-72, wherein -V-L is —CH2CH2C1, —CH2CH2CH2C1, —CH2CH2NH2, or —CH2CH2CH2NH2.
  • Embodiment 74
  • The compound of any one of Embodiments 70-73, wherein —Y—W— is a bond, —OCH2—, —OCH2CH2—, —OCH(CH3)—, —NH—, —NHCH2—, —NHC(═O)—, or —C(═O)NH—.
  • Embodiment 75
  • The compound of any one of Embodiments 70-74, wherein X is a bond, —CH2—, —C(CH3)H—, —C(CH3)2—, or —CH2CH2—.
  • Embodiment 76
  • The compound of Embodiment 1, wherein the compound has the structure of formula (C):
  • Figure US20200123117A1-20200423-C00380
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen, —NH2, or —CF3;
      • D is —NH or —NR3;
      • U is each independently O, S, or NR16;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, halogen, —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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy; or R5 and R6 taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R7 is H or C1-C6 alkyl;
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl; or R14 and R15 taken together form a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, 2, or 3; and
      • t is 0, 1 or 2.
    Embodiment 77
  • The compound of Embodiment 1, wherein the compound has the structure of formula (D):
  • Figure US20200123117A1-20200423-C00381
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a 5- or 6-membered heteroaryl comprising 1 or 2 heteroatoms selected from O, S, or N as a ring member;
      • X is —(CR5R6)t— or —NR7—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —NH2, or —CF3;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • 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.
    Embodiment 78
  • The compound of Embodiments 1, wherein the compound has the structure of formula (E)
  • Figure US20200123117A1-20200423-C00382
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00383
      • X is —(CR5R6)t— or —NR7—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is hydrogen or halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NHSO2CH3, —N(CH3)SO2CH3, —CH2NHSO2CH3, —CH2N(CH3)SO2CH3, —SO2NH2, —CONH2, —CON(C1-C3 alkyl)2, —CONH(C1-C3 alkyl), —NHCO(C1-C3 alkyl), or —N(CH3)CO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, or 2; and
      • t is 1 or 2.
    Embodiment 79
  • The compound of Embodiment 1, wherein the compound has the structure of formula (F):
  • Figure US20200123117A1-20200423-C00384
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00385
      • X is —(CR5R6)t— or —NR7—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is hydrogen or halogen;
      • R1 and R2 are each independently halogen or —CN;
      • R3 is selected from —NHSO2CH3, —N(CH3)SO2CH3, or —SO2CH3;
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, or 2; and
      • t is 1 or 2.
    Embodiment 80
  • The compound of Embodiment 1, wherein the compound has the structure of formula (G):
  • Figure US20200123117A1-20200423-C00386
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00387
      • X is —(CR5R6)t— or —NR7—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2— and L is hydrogen;
      • or alternatively, V is —CH2CH2— or —CH2CH2CH2—, and L is halogen
      • R1 and R2 are each independently Cl or —CN;
      • R3 is selected from —NHSO2CH3, —N(CH3)SO2CH3, or —SO2CH3;
      • R5 and R6 are each independently hydrogen or methyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1, or 2; and
      • t is 1.
    Embodiment 81
  • The compound of Embodiment 1, wherein the compound has the structure of formula (H)
  • Figure US20200123117A1-20200423-C00388
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00389
      • X is —(CR5R6)t— or —NR7—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2—, —CH2CH2— or —CH2CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • R5 and R6 are each independently hydrogen or methyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • t is 1.
    Embodiment 82
  • The compound of any one of Embodiments 1-69, wherein A and B are each monocyclic ring.
  • Embodiment 83
  • The compound of any one of Embodiments 1-69, wherein B is phenyl, pyridyl, or pyrimidyl.
  • Embodiment 84
  • The compound of any one of Embodiments 1-36, wherein Z and V are not both a bond.
  • Embodiment 85
  • The compound of any one of Embodiments 1-78, wherein Y and W are not both a bond.
  • Embodiment 86
  • The compound of any one of Embodiments 1-48, wherein C is a 4- to 10-membered ring.
  • Embodiment 87
  • A compound selected from Compounds A1-A97 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • Embodiment 88
  • A compound selected from Compounds A98-A186 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • Embodiment 89
  • A compound selected from Compounds A187-A211 or a pharmaceutically acceptable salt thereof.
  • Embodiment 90
  • A compound selected from Compounds B1-B11 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • Embodiment 91
  • A compound having a structure of formula (IA):
  • Figure US20200123117A1-20200423-C00390
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-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—, —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, —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, 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, —NR14COOR16, —NR14COR16, —NR14CONR14R15, 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;
      • 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 R8b 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, C1-C6 haloalkyl, C2-C6 haloalkenyl, C2-C6 haloalkynyl, optionally substituted carbocyclyl, optionally substituted —CO(C1-C6 alkyl), —CO(optionally substituted heterocyclyl), 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 —COO(C1-C6 alkyl), 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 or 2;
      • 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.
    Embodiment 92
  • The compound of Embodiment 91, wherein the compound has the structure of formula (VA):
  • Figure US20200123117A1-20200423-C00391
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 5- to 10-membered heteroaryl or aryl;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —OH, —NH2, or —CF3;
      • 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, halogen, oxo, ═S, —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, —NR14CONR14R15, —(C1-C6 alkyl)-NR14COR16, —CONR14R15, —(C1-C3 alkyl)-CONR14R15, —SO2NR14R15, —(C1-C3 alkyl)-SO2NR14R15, —SO2(C1-C3 alkyl), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, —NH2, 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 H, C1-C6 alkyl, —CO(C1-C6 alkyl);
      • R13, R14 and R15 are each independently hydrogen, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, or —COO(C1-C6 alkyl); or R14 and R15 taken together form a 3- to 6-membered heterocyclyl;
      • R16 is hydrogen, C1-C3 alkyl, C1-C3 haloalkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
      • 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.
    Embodiment 93
  • The compound of any one of Embodiments 1-7, 33-39, and 49, wherein C, which is optionally substituted with R3, is selected from
  • Figure US20200123117A1-20200423-C00392
    Figure US20200123117A1-20200423-C00393
  • wherein R3a is C1-C3 alkyl.
  • Embodiment 94
  • The compound of Embodiment 1, wherein the compound has the structure of formula (E-I):
  • Figure US20200123117A1-20200423-C00394
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00395
      • X is —(CR5R6)t— or —NRT—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, —CH2CH2CH2—, or —CH2CHClCH2—;
      • L is hydrogen, —OH, or halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 0, 1, or 2; and
      • t is 1 or 2.
    Embodiment 95
  • The compound of Embodiment 94, wherein R3 is selected from hydrogen, 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).
  • Embodiment 96
  • The compound of Embodiment 1, wherein the compound has the structure of formula (G-I):
  • Figure US20200123117A1-20200423-C00396
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
  • C is
  • Figure US20200123117A1-20200423-C00397
  • X is —(CR5R6)t—;
  • Y is —O—;
  • Z is —O—;
  • W is —CH2— or —C(CH3)H—;
  • V is —CH2CH2— or —CH2CH2CH2—;
  • L is halogen
  • R1 and R2 are each independently Cl or —CN;
  • R3 is selected from —NHSO2CH3, —N(CH3)SO2CH3, or —SO2CH3;
  • R5 and R6 are each independently hydrogen or methyl;
  • n1 and n2 are each independently 0, 1, or 2;
  • n3 is 1 or 2; and
  • t is 1.
  • Embodiment 97
  • The compound of Embodiment 1, wherein the compound has the structure of formula (H-I):
  • Figure US20200123117A1-20200423-C00398
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C—I is
  • Figure US20200123117A1-20200423-C00399
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2—, —CH2CH2— or —CH2CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • R5 and R6 are each independently hydrogen or methyl;
      • R7 is H or C1-C6 alkyl;
      • n1 and n2 are each independently 0, 1, or 2; and
      • t is 1.
    Embodiment 98
  • The compound of Embodiment 1, wherein the compound has the structure of formula (E-II)
  • Figure US20200123117A1-20200423-C00400
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00401
      • X is —(CR5R6)—;
      • Y is a bond, —CH2—, —O—, or —NH—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is —CH2—, —CH2CH2—, or —CH2CH2CH2—;
      • L is halogen;
      • R1, R2A and R2B are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from hydrogen, F, Cl, Br, I, oxo, —CN, —CF3, —OH, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, C1-C3 alkoxy, —S(C1-C3 alkyl), —SO2(C1-C3 alkyl), —NH2, —NHSO2CH3, —NHSO2CF3, —N(CH3)SO2CH3, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or C1-C3 alkyl;
      • n1 is 0, 1, or 2; and
      • n3 is 1 or 2;
  • wherein when C—R3 is
  • Figure US20200123117A1-20200423-C00402
  • R2A and R2B are not both Cl.
  • Embodiment 99
  • A compound having the structure of formula (IB):
  • Figure US20200123117A1-20200423-C00403
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, —NR7—, —N(R7)CO—, —CON(R7)—, or —NSO2R7—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —(CR8aR9a)m—, —N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CN, —OR10; —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, deuterium, 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, —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, 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, —NR14COOR16, —NR14COR16, —NR14CONR14R15, 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;
      • 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, 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 R8b 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 C1-C6 alkyl-NH2, C1-C6 haloalkyl, C2-C6 haloalkenyl, C2-C6 haloalkynyl, optionally substituted carbocyclyl, optionally substituted —CO(C1-C6 alkyl), —CO(optionally substituted heterocyclyl), 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 —COO(C1-C6 alkyl), optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or (R11 and R12) or (R14 and R15) or (R14 and R16) taken together form an optionally substituted heterocyclyl;
      • each m is independently 0, 1 or 2;
      • 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.
    Embodiment 100
  • A compound having the structure of formula (IIA):
  • Figure US20200123117A1-20200423-C00404
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —CH2—, —C(CH3)H—, —NHCO—, —N(C1-C3 alkyl)CO—, or —CONH—, or —CON(C1-C3 alkyl)-;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —CF2R1, —CF3, —CN, —OR10; —NR11R12, or —CONR11R12;
      • R1 and R2 are each independently hydrogen, deuterium, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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;
      • R8 and R9 are each independently hydrogen, halogen, or C1-C3 alkyl;
      • R11, R12, 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 a 3- to 6-membered heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl or C1-C6 alkyl-NH2; or R14 and R16 taken together form a 3- to 6-membered heterocyclyl;
      • 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.
    Embodiment 101
  • A compound having the structure of formula (IIB):
  • Figure US20200123117A1-20200423-C00405
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, or —NR7—;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • V is a bond, —(CR8aR9a)m—, —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, deuterium, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from hydrogen, 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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;
      • 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 R8b taken together form an optionally substituted 3- to 6-membered carbocyclyl or heterocyclyl;
      • R11, R12, 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 a 3- to 6-membered heterocyclyl;
      • R7, R10 and R16 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl or C1-C6 alkyl-NH2; or R14 and R16 taken together form a 3- to 6-membered heterocyclyl;
      • 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.
    Embodiment 102
  • A compound having the structure of formula (IC):
  • Figure US20200123117A1-20200423-C00406
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently aryl or heteroaryl;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, —O—, —C(═O)—, —S—, —S(═O)—, —SO2—, —NR7—, —N(R7)CO—, —CON(R7)—, or —NSO2R7—;
      • Y is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • W is a bond, —(CR8aR9a)m—, —C(═O)—, N(R7)CO—, —CONR7—, or —NSO2R7—;
      • Z is a bond, —(CR8R9)m—, —O—, —S—, —S(═O)—, —SO2—, or —NR7—;
      • V is —CH2—, —CH2CH2—, —CH(CH3)CH2—, —CH2CH(CH3)—, or —CH2CH2CH2—;
      • L is hydrogen, halogen, —CF2R10, —CF3, —CCl2R10, —CCl3, —CN, —OR10; —NR11R12 or —CONR11R12;
      • R1 and R2 are each independently hydrogen, deuterium, 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, —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, 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, —NR14COOR16, —NR14COR16, —NR14CONR14R15, 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;
      • 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, —CONR13R14, —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, —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 R8b 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 C1-C6 alkyl-NH2, C1-C6 haloalkyl, C2-C6 haloalkenyl, C2-C6 haloalkynyl, optionally substituted carbocyclyl, optionally substituted —CO(C1-C6 alkyl), —CO(optionally substituted heterocyclyl), 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 —COO(C1-C6 alkyl), optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or (R11 and R12) or (R14 and R15) or (R14 and R16) taken together form an optionally substituted heterocyclyl;
      • each m is independently 0, 1 or 2;
      • 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.
    Embodiment 103
  • A compound having a structure of formula (IIIA):
  • Figure US20200123117A1-20200423-C00407
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, 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)—, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 R8b 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 a 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.
    Embodiment 104
  • A compound having a structure of formula (G-II)
  • Figure US20200123117A1-20200423-C00408
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00409
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or methyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1 or 2; and
      • t is 1.
    Embodiment 105
  • The compound of any one of Embodiments 1-104, wherein at least one of R3 is —CN, —OCH3, —SO2CH3, —NHSO2CH3, —CH2NHSO2CH3, —SO2NH2, —CONH2, or —NHCOCH3.
  • Embodiment 106
  • A compound selected from Compounds A212-A234 or a pharmaceutically acceptable salt thereof.
  • Embodiment 107
  • A pharmaceutical composition comprising a compound of any one of Embodiments 1-106 and a pharmaceutically acceptable carrier.
  • Embodiment 108
  • The pharmaceutical composition of Embodiment 107, further comprising one or more additional therapeutic agents.
  • Embodiment 109
  • The pharmaceutical composition of Embodiment 108, wherein the one or more additional therapeutic agents is for treating 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, or age-related macular degeneration.
  • Embodiment 110
  • The pharmaceutical composition of Embodiment 108, wherein 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, SKF105,111; 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; or a mammalian target of rapamycin (mTOR) inhibitor including but not limited to everolimus, temsirolimus.
  • Embodiment 111
  • A method for modulating androgen receptor activity, comprising administering a compound of any one of Embodiments 1-106, to a subject in need thereof.
  • Embodiment 112
  • The method of any one of Embodiment 111, wherein the modulating androgen receptor activity is for treating a condition or disease selected from 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, or age-related macular degeneration.
  • Embodiment 113
  • A method for treating cancer, comprising administering a compound of any one of Embodiments 1-106, to a subject in need thereof.
  • Embodiment 114
  • The method of Embodiment 113, wherein the cancer is selected from prostate cancer, breast cancer, ovarian cancer, bladder cancer, pancreatic cancer, hepatocellular cancer, endometrial cancer, or salivary gland carcinoma.
  • Embodiment 115
  • The method of Embodiment 113, wherein the cancer is prostate cancer.
  • Embodiment 116
  • The method of Embodiment 115, wherein the prostate cancer is primary or localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, advanced prostate cancer, metastatic prostate cancer, metastatic castration-resistant prostate cancer, and hormone-sensitive prostate cancer. The method of Embodiment 115, wherein the prostate cancer is primary or localized prostate cancer, locally advanced prostate cancer, recurrent prostate cancer, advanced prostate cancer, metastatic prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-resistant prostate cancer, and hormone-sensitive prostate cancer.
  • Embodiment 117
  • The method of Embodiment 115, wherein the prostate cancer is metastatic castration-resistant prostate cancer.
  • Embodiment 118
  • The method of Embodiment 115, wherein the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.
  • Embodiment 119
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 1000 nM.
  • Embodiment 120
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 900 nM.
  • Embodiment 121
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 800 nM.
  • Embodiment 122
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 700 nM.
  • Embodiment 123
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 600 nM.
  • Embodiment 124
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 500 nM.
  • Embodiment 125
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 400 nM.
  • Embodiment 126
  • The compound of any one of Embodiments 1-106, wherein the compound has an in vitro IC50 in a PSA-luciferase assay of less than about 300 nM.
  • Embodiment 127
  • The compound of any one of Embodiments 1-106 or 119-126, wherein the compound has an in vitro metabolic clearance of less than 12 μL/min/mg protein in mammalian microsomes.
  • Embodiment 128
  • The compound of Embodiment 126, wherein the mammalian microsomes are human microsomes.
  • Embodiment 129
  • The compound of any one of Embodiments 1-106 or 119-128, wherein the compound has an in vitro metabolic clearance in mammalian hepatocytes of less than 4 μL/min/million cells.
  • Embodiment 130
  • The compound of Embodiment 128, wherein the mammalian microsomes are human microsomes.
  • Embodiment 131
  • The compound of any one of Embodiment 1-106, wherein the compound has:
  • (a) an in vitro IC50 in a PSA-luciferase assay of less than about 1000 nM;
  • (b) an in vitro metabolic clearance of less than 12 μL/min/mg protein in mammalian microsomes; and
  • (c) in vitro metabolic clearance in mammalian hepatocytes of less than 4 μL/min/million cells.
  • Embodiment 132
  • The compound of any one of Embodiments 1-106, wherein the compound has:
  • (a) an in vitro IC50 in a PSA-luciferase assay of less than about 1000 nM;
  • (b) an in vitro metabolic clearance of less than 12 μL/min/mg protein in human microsomes; and
  • (c) in vitro metabolic clearance in human hepatocytes of less than 4 μL/min/million cells.
  • Embodiment 133
  • The compounds of any one of Embodiments 119-132, wherein the PSA-luciferase assay is performed as described in Example 35.
  • Embodiment 134
  • The compounds of any one of Embodiments 119-132, wherein the metabolic clearance is measured according to Example 36.
  • Embodiment 135
  • A compound having the structure of formula (IIIA):
  • Figure US20200123117A1-20200423-C00410
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
      • C is a 3- to 10-membered ring;
      • X is a bond, —(CR5R6)t—, 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)—, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
      • 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 R8b 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 a 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.
    Embodiment 136
  • The compound of embodiment 135, wherein the compound has the structure of formula (IVA):
  • Figure US20200123117A1-20200423-C00411
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
  • A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
  • C is a 3- to 10-membered ring;
  • X is a bond, —(CR5R6)t—, or —NR7;
  • Y and Z are each independently a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
  • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
  • V is —CH2— and L is halogen, —NH2, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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, optionally substituted —(C1-C6 alkyl)-SO2R16;
  • R3 is selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
  • 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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
  • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
  • 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.
  • Embodiment 137
  • The compound of embodiment 135 or 136, wherein C is 5- to 10-membered heteroaryl or aryl
  • Embodiment 138
  • The compound of embodiment 137, wherein C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
  • Embodiment 139
  • The compound of embodiment 138, wherein C, which is substituted with (R3)n3, is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl.
  • Embodiment 140
  • The compound of any one of embodiments 135-139, wherein C, which is substituted with (R3)n3, is selected from
  • Figure US20200123117A1-20200423-C00412
    Figure US20200123117A1-20200423-C00413
  • wherein R3a is C1-C3 alkyl.
  • Embodiment 141
  • The compound of any one of Embodiments 135-140, wherein R1 and R2 are each independently C1, —CN, —CF3, —OH, methyl, methoxy, or —CONH2.
  • Embodiment 142
  • The compound of any one of Embodiments 135-141, wherein:
      • A and B are phenyl;
      • X is —(CR5R6)t—;
      • Y and Z are each —O—,
      • V is —CH2— or —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, or optionally substituted C1-C6 alkyl;
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl; and
      • R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.
    Embodiment 143
  • The compound of any one of Embodiments 135-142, wherein:
      • R5 and R6 are each independently hydrogen, or C1-C3 alkyl;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2CH2—; and
      • R1 and R2 are each independently hydrogen, halogen, or —CN.
    Embodiment 144
  • The compound of any one of Embodiments 135-143, wherein the compound has the structure of formula (A-I):
  • Figure US20200123117A1-20200423-C00414
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is a 5- to 7-membered monocyclic heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
      • X is a bond, —(CR5R6)t—, or —NR7;
      • Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
      • Z is a bond, —CH2—, —O—, or —NH—;
      • W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
      • V is —CH2— and L is halogen, —NH2, or —CF3; or
      • V is —CH2CH2- and L is halogen or —NH2;
      • R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
      • R3 is selected from —CN, C1-C3 alkoxy, CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
      • R7 is H or C1-C6 alkyl;
      • 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.
    Embodiment 145
  • The compound of any one of Embodiments 135-144, wherein: at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl).
  • Embodiment 146
  • The compound of any one of Embodiments 135-145, wherein:
      • X is a bond or —(CR5R6)t;
      • W is a bond, —CH2—, or —C(CH3)H—;
      • Y is —O—;
      • Z is —O—;
      • V is —CH2— or —CH2CH2—; and
      • L is halogen.
    Embodiment 147
  • The compound of any one of Embodiments 135-146, wherein the compound has the structure of formula (G-II):
  • Figure US20200123117A1-20200423-C00415
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
      • C is
  • Figure US20200123117A1-20200423-C00416
      • X is —(CR5R6)t—;
      • Y is —O—;
      • Z is —O—;
      • W is —CH2— or —C(CH3)H—;
      • V is —CH2CH2—;
      • L is halogen;
      • R1 and R2 are each independently Cl or —CN;
      • at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl);
      • R5 and R6 are each independently hydrogen or methyl;
      • n1 and n2 are each independently 0, 1, or 2;
      • n3 is 1 or 2; and
      • t is 1.
    Embodiment 148
  • The compound of any one of Embodiments 135-147, wherein: at least one R3 is selected from —NHSO2CH3, —NHSO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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).
  • Embodiment 149
  • A compound selected from Compounds A1-A96, A98-A116, A118-A159, A161-A175, or A177-A234 or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • Embodiment 150
  • The compound of Embodiment 149, wherein the compound is selected from
  • Figure US20200123117A1-20200423-C00417
    Figure US20200123117A1-20200423-C00418
    Figure US20200123117A1-20200423-C00419
    Figure US20200123117A1-20200423-C00420
  • or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
  • Embodiment 151
  • A pharmaceutical composition comprising a compound of any one of Embodiments 135-150 and a pharmaceutically acceptable carrier
  • Embodiment 152
  • A method for treating cancer, comprising administering the compound, pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug of any one of the compound of Embodiments 135-150 or any pharmaceutical composition of Embodiment 151, to a subject in need thereof.
  • Embodiment 153
  • The method of Embodiment 152, wherein the cancer is prostate cancer.
  • Embodiment 154
  • The method of Embodiment 153, wherein the prostate cancer is metastatic castration-resistant prostate cancer.
  • Embodiment 155
  • The method of Embodiment 154, wherein the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.
  • 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 (27)

What is claimed is:
1. A compound having the structure of formula (IIIA):
Figure US20200123117A1-20200423-C00421
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
C is a 3- to 10-membered ring;
X is a bond, —(CR5R6)t—, 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)—, —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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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 selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
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 R8b 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 a 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.
2. The compound of claim 1, wherein the compound has the structure of formula (IVA):
Figure US20200123117A1-20200423-C00422
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
A and B are each independently selected from phenyl, pyridyl, pyrimidyl, or thiophene;
C is a 3- to 10-membered ring;
X is a bond, —(CR5R6)t—, or —NR7;
Y and Z are each independently a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
V is —CH2— and L is halogen, —NH2, 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, —NR13R14, optionally substituted —(C1-C6 alkyl)-NR13R14, —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, optionally substituted —(C1-C6 alkyl)-SO2R16;
R3 is selected from 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), or —(C1-C6 alkyl)-SO2(C1-C3 alkyl);
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 H or C1-C6 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 a 3- to 6-membered heterocyclyl;
R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl;
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.
3. The compound of claim 2, wherein C is 5- to 10-membered heteroaryl or aryl.
4. The compound of claim 3, wherein C is 5- to 7-membered heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member.
5. The compound of claim 4, wherein C, which is substituted with (R3)n3, is pyrazole, imidazole, oxazole, oxadiazole, oxazolone, isoxazole, thiazole, pyridyl, pyrazine, furan or pyrimidyl.
6. The compound of claim 5, wherein C, which is substituted with (R3)n3, is selected from
Figure US20200123117A1-20200423-C00423
Figure US20200123117A1-20200423-C00424
Figure US20200123117A1-20200423-C00425
wherein R3a is C1-C3 alkyl.
7. The compound of claim 6, wherein R1 and R2 are each independently C1, —CN, —CF3, —OH, methyl, methoxy, or —CONH2.
8. The compound of claim 2, wherein:
A and B are phenyl;
X is —(CR5R6)t—;
Y and Z are each —O—;
V is —CH2— or —CH2CH2—;
L is halogen;
R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, —OH, or optionally substituted C1-C6 alkyl;
R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl; and
R16 is hydrogen, C1-C3 alkyl, C2-C3 alkenyl, or C2-C3 alkynyl.
9. The compound of claim 8, wherein:
R5 and R6 are each independently hydrogen, or C1-C3 alkyl;
W is —CH2— or —C(CH3)H—;
V is —CH2CH2—; and
R1 and R2 are each independently hydrogen, halogen, or —CN.
10. The compound of claim 1, wherein the compound has the structure of formula (A-I)
Figure US20200123117A1-20200423-C00426
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
C is a 5- to 7-membered monocyclic heteroaryl comprising 1, 2, or 3 heteroatoms selected from O, S, or N as a ring member;
X is a bond, —(CR5R6)t—, or —NR7;
Y is a bond, —CH2—, —C(CH3)H—, —O—, —S—, —NH—, —NCH3—, or —N(COCH3)—;
Z is a bond, —CH2—, —O—, or —NH—;
W is a bond, —CH2—, —C(CH3)H—, —C(═O)—, —N(R7)CO—, or —CONR7—;
V is —CH2— and L is halogen, —NH2, or —CF3; or
V is —CH2CH2- and L is halogen or —NH2;
R1 and R2 are each independently hydrogen, halogen, —CN, —CF3, methyl, or —CONH2;
R3 is selected from —CN, C1-C3 alkoxy, —CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, —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), or —N(CH3)COO(C1-C3 alkyl);
R5 and R6 are each independently hydrogen, halogen, —OH, or C1-C3 alkyl;
R7 is H or C1-C6 alkyl;
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.
11. The compound of claim 10, wherein:
at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl).
12. The compound of claim 10, wherein:
X is a bond or —(CR5R6)t;
W is a bond, —CH2—, or —C(CH3)H—;
Y is —O—;
Z is —O—;
V is —CH2— or —CH2CH2—; and
L is halogen.
13. The compound of claim 1, wherein the compound has the structure of formula (G-II)
Figure US20200123117A1-20200423-C00427
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof, wherein:
C is
Figure US20200123117A1-20200423-C00428
X is —(CR5R6)t—;
Y is —O—;
Z is —O—;
W is —CH2— or —C(CH3)H—;
V is —CH2CH2—;
L is halogen;
R1 and R2 are each independently Cl or —CN;
at least one R3 is selected from —CN, C1-C3 alkoxy, —CONH2, —NHSO2CH3, —N(CH3)SO2CH3, —NHSO2CH2CH3, —N(CH3)SO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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), or —N(CH3)COO(C1-C3 alkyl);
R5 and R6 are each independently hydrogen or methyl;
n1 and n2 are each independently 0, 1, or 2;
n3 is 1 or 2; and
t is 1.
14. The compound of claim 13, wherein:
at least one R3 is selected from —NHSO2CH3, —NHSO2CH2CH3, or —SO2CH3 and the other R3, if present, is selected from —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).
15. A compound selected from the group consisting of:
Figure US20200123117A1-20200423-C00429
Figure US20200123117A1-20200423-C00430
Figure US20200123117A1-20200423-C00431
Figure US20200123117A1-20200423-C00432
Figure US20200123117A1-20200423-C00433
Figure US20200123117A1-20200423-C00434
Figure US20200123117A1-20200423-C00435
Figure US20200123117A1-20200423-C00436
Figure US20200123117A1-20200423-C00437
Figure US20200123117A1-20200423-C00438
Figure US20200123117A1-20200423-C00439
Figure US20200123117A1-20200423-C00440
Figure US20200123117A1-20200423-C00441
Figure US20200123117A1-20200423-C00442
Figure US20200123117A1-20200423-C00443
Figure US20200123117A1-20200423-C00444
Figure US20200123117A1-20200423-C00445
Figure US20200123117A1-20200423-C00446
Figure US20200123117A1-20200423-C00447
Figure US20200123117A1-20200423-C00448
Figure US20200123117A1-20200423-C00449
Figure US20200123117A1-20200423-C00450
Figure US20200123117A1-20200423-C00451
Figure US20200123117A1-20200423-C00452
Figure US20200123117A1-20200423-C00453
Figure US20200123117A1-20200423-C00454
Figure US20200123117A1-20200423-C00455
Figure US20200123117A1-20200423-C00456
Figure US20200123117A1-20200423-C00457
Figure US20200123117A1-20200423-C00458
Figure US20200123117A1-20200423-C00459
Figure US20200123117A1-20200423-C00460
Figure US20200123117A1-20200423-C00461
Figure US20200123117A1-20200423-C00462
Figure US20200123117A1-20200423-C00463
Figure US20200123117A1-20200423-C00464
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
16. The compound of claim 15, wherein the compound is selected from
Figure US20200123117A1-20200423-C00465
Figure US20200123117A1-20200423-C00466
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
17. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier.
18. A pharmaceutical composition comprising a compound of claim 4 and a pharmaceutically acceptable carrier.
19. A pharmaceutical composition comprising a compound of claim 10 and a pharmaceutically acceptable carrier.
20. A pharmaceutical composition comprising a compound of claim 13 and a pharmaceutically acceptable carrier.
21. A pharmaceutical composition comprising a compound of claim 15 and a pharmaceutically acceptable carrier.
22. A pharmaceutical composition comprising a compound of claim 16 and a pharmaceutically acceptable carrier.
23. 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.
24. The method of claim 23, wherein the cancer is prostate cancer.
25. The method of claim 24, wherein the prostate cancer is metastatic castration-resistant prostate cancer.
26. The method of claim 25, wherein the prostate cancer expresses full-length androgen receptor or truncated androgen receptor splice variant.
27. The method of claim 24, wherein the compound is selected from
Figure US20200123117A1-20200423-C00467
Figure US20200123117A1-20200423-C00468
or a pharmaceutically acceptable salt, tautomer, stereoisomer or prodrug thereof.
US16/657,625 2018-10-18 2019-10-18 Androgen receptor modulators and methods for their use Abandoned US20200123117A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/657,625 US20200123117A1 (en) 2018-10-18 2019-10-18 Androgen receptor modulators and methods for their use
US16/852,810 US11059795B2 (en) 2018-10-18 2020-04-20 Androgen receptor modulators and methods for their use
US17/363,534 US20220380325A1 (en) 2018-10-18 2021-06-30 Androgen receptor modulators and methods for their use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862747209P 2018-10-18 2018-10-18
US201962803516P 2019-02-10 2019-02-10
US201962857516P 2019-06-05 2019-06-05
US16/657,625 US20200123117A1 (en) 2018-10-18 2019-10-18 Androgen receptor modulators and methods for their use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/852,810 Continuation US11059795B2 (en) 2018-10-18 2020-04-20 Androgen receptor modulators and methods for their use

Publications (1)

Publication Number Publication Date
US20200123117A1 true US20200123117A1 (en) 2020-04-23

Family

ID=70280422

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/657,625 Abandoned US20200123117A1 (en) 2018-10-18 2019-10-18 Androgen receptor modulators and methods for their use
US16/852,810 Active US11059795B2 (en) 2018-10-18 2020-04-20 Androgen receptor modulators and methods for their use
US17/363,534 Pending US20220380325A1 (en) 2018-10-18 2021-06-30 Androgen receptor modulators and methods for their use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/852,810 Active US11059795B2 (en) 2018-10-18 2020-04-20 Androgen receptor modulators and methods for their use
US17/363,534 Pending US20220380325A1 (en) 2018-10-18 2021-06-30 Androgen receptor modulators and methods for their use

Country Status (16)

Country Link
US (3) US20200123117A1 (en)
EP (1) EP3867216A4 (en)
JP (1) JP2022504949A (en)
KR (1) KR20210093886A (en)
CN (1) CN113195441A (en)
AU (1) AU2019362061A1 (en)
BR (1) BR112021007222A8 (en)
CA (1) CA3115101A1 (en)
CL (1) CL2021000977A1 (en)
CO (1) CO2021006354A2 (en)
IL (1) IL282257A (en)
MX (2) MX2021004427A (en)
PE (1) PE20211543A1 (en)
PH (1) PH12021550800A1 (en)
SG (1) SG11202103325WA (en)
WO (1) WO2020081999A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11059795B2 (en) 2018-10-18 2021-07-13 Essa Pharma, Inc. Androgen receptor modulators and methods for their use
US11142508B2 (en) 2016-04-15 2021-10-12 The University Of British Columbia Bisphenol derivatives and their use as androgen receptor activity modulators
US11242324B2 (en) 2020-04-17 2022-02-08 Essa Pharma, Inc. Solid forms of an n-terminal domain androgen receptor inhibitor and uses thereof
WO2022051330A1 (en) 2020-09-02 2022-03-10 Propella Therapeutics, Inc. Abiraterone prodrugs
US11345670B2 (en) 2015-01-13 2022-05-31 The University Of British Columbia Heterocyclic compounds for cancer imaging and treatment and methods for their use
WO2022174134A1 (en) 2021-02-15 2022-08-18 Propella Therapeutics, Inc. Abiraterone prodrugs
US11485713B2 (en) 2018-05-25 2022-11-01 Essa Pharma, Inc. Androgen receptor modulators and methods for their use
WO2023034916A1 (en) * 2021-09-01 2023-03-09 Essa Pharma, Inc. Pharmaceutical compositions and combinations comprising inhibitors of the androgen receptor and uses thereof
WO2023038933A1 (en) 2021-09-08 2023-03-16 Propella Therapeutics, Inc. Oral abiraterone formulations
WO2023056423A1 (en) * 2021-09-30 2023-04-06 Essa Pharma, Inc. Androgen receptor modulators and methods for use as bifunctional degraders
WO2023167783A1 (en) 2022-03-01 2023-09-07 Propella Therapeutics, Inc. Abiraterone decanoate prodrugs and use in therapy
US11779550B2 (en) 2015-03-12 2023-10-10 The University Of British Columbia Bisphenol ether derivatives and methods for using the same
WO2024107928A1 (en) 2022-11-16 2024-05-23 Propella Therapeutics, Inc. Abiraterone decanoate composition and use in therapy

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3947353A4 (en) * 2019-03-28 2023-07-05 Essa Pharma, Inc. Pharmaceutical compositions and combinations comprising inhibitors of the androgen receptor and uses thereof
JP2021050161A (en) 2019-09-25 2021-04-01 武田薬品工業株式会社 Heterocyclic compound and use thereof
JP2021080177A (en) 2019-11-14 2021-05-27 武田薬品工業株式会社 Heterocyclic compounds and uses thereof
US20220105093A1 (en) * 2020-09-16 2022-04-07 Essa Pharma, Inc. Pharmaceutical compositions and combinations comprising inhibitors of the androgen receptor and uses thereof
CN113603695B (en) * 2021-08-05 2022-06-14 青岛恒宁生物科技有限公司 Substituted pyrimidinamine compound or salt thereof acceptable as pesticide, composition and application thereof
WO2023031371A1 (en) 2021-09-01 2023-03-09 Janssen Pharmaceutica Nv Combination therapies for metastatic castration-resistant prostate cancer
WO2023110473A1 (en) 2021-12-14 2023-06-22 Basf Se Heterocyclic compounds for the control of invertebrate pests
EP4198033A1 (en) 2021-12-14 2023-06-21 Basf Se Heterocyclic compounds for the control of invertebrate pests
WO2024006207A1 (en) * 2022-06-26 2024-01-04 Essa Pharma Inc. An n-terminal domain androgen receptor inhibitor and uses thereof

Family Cites Families (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2571217A (en) 1951-10-16 Horace s
US2890189A (en) 1954-06-14 1959-06-09 Johnson & Son Inc S C Alkali soluble resins and compositions containing the same
US3074974A (en) 1957-12-06 1963-01-22 Monsanto Chemicals Method for the preparation of diglycidyl ether of tetrachlorobisphenol-a
US3162615A (en) 1961-01-03 1964-12-22 Dow Chemical Co Polyesters from cyclic polyhaloalkane polyols and unsaturated dicarboxylic acids
FR1389005A (en) 1963-01-09 1965-02-12 Bayer Ag Advanced process for curing polyepoxides
SU638596A1 (en) 1975-09-01 1978-12-25 Государственный Научно-Исследовательский И Проектный Институт Полимерных Клеев Им. Э.Л.Тер-Газаряна Diallylisocyanuric acid chlorohydrin derivative as plasticizer of polyvinyl acetate emulsion
US4284574A (en) 1979-06-15 1981-08-18 Ciba-Geigy Corporation Diglycidyl ethers of di-secondary alcohols, their preparation, and curable compositions containing them
SU929630A1 (en) 1980-02-06 1982-05-23 Научно-Исследовательский Институт Биологии При Иркутском Государственном Университете Им.А.А.Жданова Process for producing di- or trioxydiphenyl sulfones
US4369298A (en) 1980-05-27 1983-01-18 Tokuyama Soda Kabushiki Kaisha Novel cured resin, process for production thereof, and lens composed of said resin from bis(alkyleneoxyphenyl)-diacrylate, bis(alkyleneoxyphenyl)diallyl ether, bis(alkyleneoxyphenyl)diallyl carbonate monomers
DE3170223D1 (en) 1981-01-13 1985-06-05 Teijin Ltd Ion-permeable composite membrane and its use in selective separation
IN169231B (en) 1984-03-15 1991-09-14 Ciba Geigy Ag
PL141793B1 (en) 1984-10-08 1987-08-31 Politechnika Warszawska Method of obtaining bisphenolic resins
US5753730A (en) 1986-12-15 1998-05-19 Mitsui Toatsu Chemicals, Inc. Plastic lenses having a high-refractive index, process for the preparation thereof and casting polymerization process for preparing sulfur-containing urethane resin lens and lens prepared thereby
JPH0832844B2 (en) 1987-02-09 1996-03-29 パイロツトインキ株式会社 Opaque ink for writing board
US4904760A (en) 1987-04-27 1990-02-27 Mitsubishi Gas Chemical Co., Inc. Thermosetting resin composition from cyanate ester and non-branched aromatic compound
JPH01503541A (en) 1987-06-01 1989-11-30 ザ ダウ ケミカル カンパニー Method for producing propargyl ether of hydroxy aromatic compound
US5155196A (en) 1987-06-01 1992-10-13 The Dow Chemical Company Polymer resulting from the cure of a preformed chromene-containing mixture
JP2572020B2 (en) 1987-06-19 1997-01-16 竹本油脂株式会社 Flame retardant for thermosetting resin
DE3821585A1 (en) 1987-09-13 1989-03-23 Hoechst Ag POSITIVELY WORKING RADIATION-SENSITIVE MIXTURE AND PRODUCTION OF RADIATION-SENSITIVE RECORDING MATERIAL FOR HIGH-ENERGY RADIATION
US4855184A (en) 1988-02-02 1989-08-08 Minnesota Mining And Manufacturing Company Radiation-curable protective coating composition
DE3939760A1 (en) 1989-12-01 1991-06-06 Bayer Ag METHOD FOR LACQUERING PLASTICS, LACQUERED PLASTICS AND THE USE OF ADHESIVES SUITABLE FOR THIS
EP0515128A1 (en) 1991-05-23 1992-11-25 Konica Corporation Silver halide color photographic light-sensitive material
JPH0649473A (en) 1992-08-04 1994-02-22 Asahi Chem Ind Co Ltd Refrigerant composition
DE4323512A1 (en) 1992-09-01 1994-04-28 Agfa Gevaert Ag Photographic recording material
JPH07117349A (en) 1993-10-27 1995-05-09 Asahi Denka Kogyo Kk Thermal recording material
JPH10509962A (en) 1994-11-29 1998-09-29 ヘキスト・マリオン・ルセル・インコーポレイテツド Use of triaryl-ethylene derivatives in the treatment and prevention of osteoporosis
DE19526146A1 (en) 1995-07-07 1997-01-09 Schering Ag Triphenylethylenes, processes for their preparation, pharmaceutical preparations containing these triphenylethylenes and their use for the preparation of medicaments
JPH09176240A (en) 1995-12-27 1997-07-08 Mitsubishi Chem Corp Photopolymerizable composition
JPH10133427A (en) 1996-11-05 1998-05-22 Fuji Xerox Co Ltd Carrier for developing electrostatic latent image, electrostatic latent image developer, and image forming method
AR011626A1 (en) 1997-02-07 2000-08-30 Shell Int Research A PROCEDURE FOR THE ELABORATION OF EPOXID COMPOUNDS, EPOXID RESINS OBTAINED THROUGH SUCH A PROCEDURE AND A PROCEDURE FOR THE ELABORATION OF INTERMEDIATE COMPOUNDS FOR THE ELABORATION OF EPOXID COMPOUNDS
JPH10316803A (en) 1997-05-16 1998-12-02 Teijin Chem Ltd Flame-retardant resin composition
IL125840A (en) 1997-08-22 2002-12-01 Teijin Chemicals Ltd Bromine compound production method
JPH11166087A (en) 1997-12-04 1999-06-22 Teijin Chem Ltd Flame-retardant resin composition
WO2000001813A2 (en) 1998-06-30 2000-01-13 The University Of British Columbia Peptide inhibitors of androgen-independent activation of androgen receptor
US6245117B1 (en) 1998-08-07 2001-06-12 Ipposha Oil Industries Co., Ltd. Modifier of cellulose fibers and modification method of cellulose fibers
US6218430B1 (en) 1998-08-24 2001-04-17 Ligand Pharmaceuticals Incorporated Vitamin D3 mimics
JP3795679B2 (en) 1998-09-01 2006-07-12 帝人化成株式会社 Method for producing bromine compound
AU775928B2 (en) 1999-10-14 2004-08-19 Bristol-Myers Squibb Company Crystallographic structure of the androgen receptor ligand binding domain
US6534621B2 (en) 2000-05-18 2003-03-18 Dow Global Technologies Inc. Process for manufacturing a hydroxyester derivative intermediate and epoxy resins prepared therefrom
US6472436B1 (en) 2000-07-17 2002-10-29 The Salk Institute For Biological Studies Methods for protecting cells from amyloid toxicity and for inhibiting amyloid protein production
WO2002018334A2 (en) 2000-08-31 2002-03-07 Theravance, Inc. Sodium channel modulators
DE10102322A1 (en) * 2001-01-19 2002-07-25 Merck Patent Gmbh New disubstituted and trisubstituted benzene derivatives useful as coagulation inhibitors for the treatment of thromboembolic diseases
US6646102B2 (en) 2001-07-05 2003-11-11 Dow Global Technologies Inc. Process for manufacturing an alpha-dihydroxy derivative and epoxy resins prepared therefrom
US20030092724A1 (en) 2001-09-18 2003-05-15 Huaihung Kao Combination sustained release-immediate release oral dosage forms with an opioid analgesic and a non-opioid analgesic
US7344700B2 (en) 2002-02-28 2008-03-18 University Of Tennessee Research Corporation Radiolabeled selective androgen receptor modulators and their use in prostate cancer imaging and therapy
GB0324551D0 (en) 2003-10-21 2003-11-26 Karobio Ab Novel compounds
CA2544501A1 (en) 2003-11-20 2005-06-09 Eli Lilly And Company Vitamin d receptor modulators
US8114381B2 (en) 2004-02-13 2012-02-14 The University Of British Columbia Radiolabeled compounds and compositions, their precursors and methods for their production
JP2005325301A (en) 2004-05-17 2005-11-24 Fuji Photo Film Co Ltd Cellulose acylate dope composition and cellulose acylate film
EP1781280A2 (en) 2004-08-18 2007-05-09 Warner-Lambert Company LLC Androgen modulators
JP2006208607A (en) 2005-01-26 2006-08-10 Fuji Photo Film Co Ltd Pattern forming material and device, and permanent pattern forming method
JP4753601B2 (en) 2005-03-23 2011-08-24 旭化成イーマテリアルズ株式会社 Photosensitive composition
EP1717235A3 (en) 2005-04-29 2007-02-28 Bioprojet Phenoxypropylpiperidines and -pyrrolidines and their use as histamine H3-receptor ligands
FR2885904B1 (en) 2005-05-19 2007-07-06 Aventis Pharma Sa NOVEL FLUORENE DERIVATIVES, COMPOSITIONS CONTAINING SAME AND USE THEREOF
EP1792622A1 (en) 2005-11-11 2007-06-06 GPC Biotech AG Anti-proliferative combination therapy comprising a platinum-based chemotherapeutic agent and EGFR inhibitors or pyrimidine analogues
HUE031972T2 (en) * 2005-12-29 2017-08-28 Celtaxsys Inc Diamine derivatives as inhibitors of leukotriene a4 hydrolase
JP2007290980A (en) 2006-04-21 2007-11-08 Shin Etsu Chem Co Ltd Fluorine-containing (meth)acrylic acid ester
DE102006019044A1 (en) 2006-04-25 2007-10-31 Merck Patent Gmbh antioxidants
TW200819421A (en) 2006-10-31 2008-05-01 Univ Nat Chunghsing The method of synthesizing biphenol A, BPA having di-alkoxyl group by using polycarbonate or its waste
JP2010508358A (en) * 2006-11-01 2010-03-18 ブリストル−マイヤーズ スクイブ カンパニー Glucocorticoid receptor, AP-1 and / or modulator of NF-κB activity, and use thereof
KR20090115797A (en) 2007-02-20 2009-11-06 바스프 에스이 High refractive index monomers, compositions and uses thereof
RU2015107733A (en) 2008-07-02 2015-06-27 Бритиш Коламбиа Кэнсер Эйдженси Бранч THERAPEUTIC BASES ON THE BASIS OF DERIVATIVES OF DIGLYCYLIC ETHERS AND WAYS OF THEIR APPLICATION
AU2009279936A1 (en) 2008-08-05 2010-02-11 Banyu Pharmaceutical Co., Ltd. Therapeutic compounds
US20130109758A1 (en) 2010-01-06 2013-05-02 The University Of British Columbia Bisphenol derivative therapeutics and methods for their use
CA2786319C (en) 2010-01-06 2019-03-12 British Columbia Cancer Agency Branch Bisphenol derivatives and their use as androgen receptor activity modulators
US20140248263A1 (en) 2011-04-08 2014-09-04 The University Of British Columbia Bisphenol compounds and methods for their use
WO2012145330A1 (en) 2011-04-18 2012-10-26 The University Of British Columbia Fluorene-9-bisphenol compounds and methods for their use
WO2012145328A1 (en) 2011-04-18 2012-10-26 The University Of British Columbia Dibenzylphenyl compounds and methods for their use
WO2013028572A1 (en) 2011-08-19 2013-02-28 British Columbia Cancer Agency Branch Fluorinated bisphenol ether compounds and methods for their use
WO2013028791A1 (en) 2011-08-22 2013-02-28 British Columbia Cancer Agency Branch 18f compounds for cancer imaging and methods for their use
US9365510B2 (en) 2012-04-16 2016-06-14 British Columbia Cancer Agency Branch Aziridine bisphenol ethers and related compounds and methods for their use
CN105358522A (en) * 2013-05-10 2016-02-24 不列颠哥伦比亚癌症局分支机构 Ester derivatives of androgen receptor modulators and methods for their use
CN103342892A (en) 2013-06-06 2013-10-09 西安交通大学 Bimaleimide resin toughening modifiers and preparation method thereof
US20150010469A1 (en) 2013-07-03 2015-01-08 British Columbia Cancer Agency Branch Bisphenol ether compounds with novel bridging groups and methods for their use
EP3044197B1 (en) 2013-09-09 2018-11-07 British Columbia Cancer Agency Branch Halogenated compounds for cancer imaging and treatment and methods for their use
WO2016058080A1 (en) 2014-10-14 2016-04-21 British Columbia Cancer Agency Branch Fluoro-chloro bisphenol ether compounds and methods for their use
WO2016058082A1 (en) 2014-10-14 2016-04-21 British Columbia Cancer Agency Branch 18f compounds for cancer imaging and methods for their use
AU2016206412B2 (en) 2015-01-13 2020-10-08 British Columbia Cancer Agency Branch Heterocyclic compounds for cancer imaging and treatment and methods for their use
US10471023B2 (en) 2015-03-12 2019-11-12 British Columbia Cancer Agency Branch Bisphenol ether derivatives and methods for using the same
US20170056336A1 (en) 2015-09-02 2017-03-02 British Columbia Cancer Agency Branch Co-targeting androgen receptor splice variants and mtor signaling pathway for the treatment of castration-resistant prostate cancer
US20170298033A1 (en) * 2016-04-15 2017-10-19 The University Of British Columbia Bisphenol derivatives and their use as androgen receptor activity modulators
WO2017210771A1 (en) 2016-06-06 2017-12-14 British Columbia Cancer Agency Branch Compounds and compositions for radiation therapy and methods of using the same
WO2018045450A1 (en) * 2016-09-09 2018-03-15 British Columbia Cancer Agency Branch Bisphenol a compounds and methods for treating drug-resistant androgen receptor mediated cancers
BR112021007222A8 (en) * 2018-10-18 2022-11-22 Provincial Health Services Authority ANDROGEN RECEPTOR MODULATORS AND METHODS FOR THEIR USE

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11345670B2 (en) 2015-01-13 2022-05-31 The University Of British Columbia Heterocyclic compounds for cancer imaging and treatment and methods for their use
US11779550B2 (en) 2015-03-12 2023-10-10 The University Of British Columbia Bisphenol ether derivatives and methods for using the same
US11142508B2 (en) 2016-04-15 2021-10-12 The University Of British Columbia Bisphenol derivatives and their use as androgen receptor activity modulators
US11919874B2 (en) 2016-04-15 2024-03-05 The University Of British Columbia Bisphenol derivatives and their use as androgen receptor activity modulators
US11485713B2 (en) 2018-05-25 2022-11-01 Essa Pharma, Inc. Androgen receptor modulators and methods for their use
US11059795B2 (en) 2018-10-18 2021-07-13 Essa Pharma, Inc. Androgen receptor modulators and methods for their use
US11518747B2 (en) 2020-04-17 2022-12-06 Essa Pharma, Inc. Solid forms of an N-terminal domain androgen receptor inhibitor and uses thereof
US11358938B2 (en) 2020-04-17 2022-06-14 Essa Pharma, Inc. Solid forms of an N-terminal domain androgen receptor inhibitor and uses thereof
US11814357B2 (en) 2020-04-17 2023-11-14 Essa Pharma Inc. Solid forms of an N-terminal domain androgen receptor inhibitor and uses thereof
US11242324B2 (en) 2020-04-17 2022-02-08 Essa Pharma, Inc. Solid forms of an n-terminal domain androgen receptor inhibitor and uses thereof
WO2022051330A1 (en) 2020-09-02 2022-03-10 Propella Therapeutics, Inc. Abiraterone prodrugs
WO2022174134A1 (en) 2021-02-15 2022-08-18 Propella Therapeutics, Inc. Abiraterone prodrugs
WO2023034916A1 (en) * 2021-09-01 2023-03-09 Essa Pharma, Inc. Pharmaceutical compositions and combinations comprising inhibitors of the androgen receptor and uses thereof
WO2023038933A1 (en) 2021-09-08 2023-03-16 Propella Therapeutics, Inc. Oral abiraterone formulations
WO2023056423A1 (en) * 2021-09-30 2023-04-06 Essa Pharma, Inc. Androgen receptor modulators and methods for use as bifunctional degraders
WO2023167783A1 (en) 2022-03-01 2023-09-07 Propella Therapeutics, Inc. Abiraterone decanoate prodrugs and use in therapy
WO2024107928A1 (en) 2022-11-16 2024-05-23 Propella Therapeutics, Inc. Abiraterone decanoate composition and use in therapy

Also Published As

Publication number Publication date
BR112021007222A8 (en) 2022-11-22
BR112021007222A2 (en) 2021-08-10
PH12021550800A1 (en) 2021-10-04
CA3115101A1 (en) 2020-04-23
CL2021000977A1 (en) 2021-10-22
AU2019362061A8 (en) 2021-06-17
MX2023010043A (en) 2023-09-05
JP2022504949A (en) 2022-01-13
CN113195441A (en) 2021-07-30
KR20210093886A (en) 2021-07-28
PE20211543A1 (en) 2021-08-16
AU2019362061A1 (en) 2021-05-06
WO2020081999A1 (en) 2020-04-23
EP3867216A1 (en) 2021-08-25
MX2021004427A (en) 2021-11-12
US20200247763A1 (en) 2020-08-06
IL282257A (en) 2021-05-31
EP3867216A4 (en) 2022-07-13
CO2021006354A2 (en) 2021-05-31
US11059795B2 (en) 2021-07-13
SG11202103325WA (en) 2021-05-28
US20220380325A1 (en) 2022-12-01

Similar Documents

Publication Publication Date Title
US11059795B2 (en) Androgen receptor modulators and methods for their use
US20220202780A1 (en) Pharmaceutical compositions and combinations comprising inhibitors of the androgen receptor and uses thereof
US11485713B2 (en) Androgen receptor modulators and methods for their use
US20220218632A1 (en) Pharmaceutical compositions and combinations comprising inhibitors of the androgen receptor and uses thereof
US20230078913A1 (en) Androgen receptor modulators and methods for use as proteolysis targeting chimera ligands
US8846704B2 (en) Thiopyrimidine-based compounds and uses thereof
JP6033835B2 (en) Benzo [b] [1,4] oxazine derivatives as calcium-sensing receptor modulators
US20050165074A1 (en) Amide derivatives as C-KIT modulators
US10682358B2 (en) Substituted 2, 3-dihydro-1H-inden-1-one retinoic acid-related orphan nuclear receptor antagonists for treating multiple sclerosis
US8569345B2 (en) Compounds and compositions as LXR modulators
US20150376174A1 (en) Azole benzene derivative
US20170137415A1 (en) Sulfonamide compounds as voltage gated sodium channel modulators
US20220177459A1 (en) Aromatic amine compound and use thereof in preparation of ar and brd4 dual inhibitors and regulators
RU2797622C2 (en) Androgen receptor modulators and methods for their use
US20230373934A1 (en) Androgen receptor modulators and methods for their use
JP2009507815A (en) Thiazole compounds and their use as PGD2 antagonists
US20220380378A1 (en) Androgen receptor modulators and methods for their use
CZ2022277A3 (en) Triterpenes with inhibitory activity on the Hedgehog signalling pathway for the use in the treatment of cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: ESSA PHARMA, INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZHOU, HAN-JIE;VIRSIK, PETER;REEL/FRAME:050890/0146

Effective date: 20191022

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP., ISSUE FEE NOT PAID

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE